(Methods in Molecular Biology) - Artificial Riboswitches - Methods and Protocols (2014, Humana Press)
(Methods in Molecular Biology) - Artificial Riboswitches - Methods and Protocols (2014, Humana Press)
(Methods in Molecular Biology) - Artificial Riboswitches - Methods and Protocols (2014, Humana Press)
Artificial
Riboswitches
Methods and Protocols
METHODS IN M O L E C U L A R B I O LO G Y
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Atsushi Ogawa
Ehime University, Matsuyama, Ehime, Japan
Editor
Atsushi Ogawa
Ehime University
Matsuyama
Ehime, Japan
Ribonucleic acids or RNAs are versatile. mRNAs serve as mediators of hereditary informa-
tion from DNAs to proteins, tRNAs carry amino acids into the ribosome on mRNA, and
rRNAs in the ribosome promote peptide bond formation between the amino acids. In
addition, many diverse noncoding RNAs directly regulate gene expression in cis and/or
trans. There are also many other types of RNAs known in nature, including RNA enzymes
(ribozymes). It is considered that in an ancient RNA world (where proteins and DNAs did
not still exist), a greater variety of functional RNAs would have flourished, though almost
all of them are now defunct.
RNA functions are dependent on their sequences, which are composed of four kinds of
nucleotides: A, G, U, and C. The sequences determine the higher order structures, which
in turn make possible their unique functions. In other words, we could create new func-
tional RNAs by appropriately lining up the nucleotides. Although RNAs are not as diverse
as proteins, which have 20 amino acids as building block, RNAs have a large advantage in
that they are easy to be amplified with the following three steps: reverse transcription, poly-
merase chain reaction (PCR), and transcription. We can therefore obtain functional RNAs
from a randomized RNA pool by repeating selection–amplification cycles (in vitro selec-
tion). In fact, many types of functional RNAs have been selected via in vitro selection. The
representative examples are aptamers that tightly bind to their specific ligand molecules and
ribozymes that catalyze specific biochemical reactions. In vitro selection also enables us to
obtain a ligand-responsive ribozyme called aptazyme, which is generally a conjugate
between two foregoing functional RNAs, an aptamer and a ribozyme.
A riboswitch is also a functional RNA: a ligand-dependent and cis-acting gene regulator
in mRNA. This regulatory RNA is composed of an aptamer domain, which binds to the
specific ligand as described above, and an expression platform, which regulates transcription
termination, translation initiation, self-cleavage, or splicing of its own mRNA. The interac-
tion between the aptamer domain and the ligand induces conformational changes of the
expression platform to up- or down-regulate gene expression. Natural riboswitches acting
in response to endogenous metabolites have been identified mainly in bacteria and rarely in
eukaryotes over the last decade. In parallel with these discoveries, a variety of methods have
also been reported for artificially constructing arbitrary molecule-dependent riboswitches
with the corresponding in vitro-selected aptamers or self-cleaving aptazymes.
This volume is mainly focused on the state-of-the-art methods developed in recent
years for creating artificial riboswitches. Approximately half of the total chapters are devoted
to screening or rational design methods for obtaining artificial riboswitches that function in
either bacterial or eukaryotic translation systems. In these methods, an aptamer or an apta-
zyme is generally required for a gene regulator to acquire ligand responsiveness. Although
an already identified aptamer or aptazyme is available, several chapters cover in vitro
selection methods for obtaining a new aptamer or aptazyme for a ligand molecule that the
reader might be interested in (a small molecule, protein, or photo-responsive molecule).
In this context, one chapter is devoted to a computational method for designing a starting
library of RNA sequences for in vitro selection. Protocols for evaluating the activities of the
v
vi Preface
resultant riboswitches are also presented. Some other chapters include protocols for con-
struction of ligand-dependent, trans-acting gene regulators.
Artificial riboswitches and other ligand-responsive gene regulators that are obtainable
through the cutting-edge methods described here make it possible to switch protein syn-
thesis ON or OFF with arbitrary ligand molecules, which can be freely chosen when select-
ing the corresponding aptamers to be implemented in the gene regulators. Therefore, this
book can be regarded as a collection of recipes for the gene circuit elements in synthetic
biology and metabolic engineering. However, I would recommend this cookbook not only
to bioengineers who aim to reprogram cell behaviors and molecular biologists who leverage
these regulators for genetic studies but also to all researchers who just want to regulate the
expression of a specific gene by an arbitrary molecule in various organisms, to detect a spe-
cific molecule with reporter protein expression in vitro or in vivo, or to design ligand-
dependent RNA switches by using aptamers or aptazymes.
All chapters are written by experts from all around the world who are active in the front
lines of the relevant research areas. I would like to express my gratitude to the contributors,
all of whom were willing to write their lab protocols in an easily comprehensible manner,
despite their busy research lives. I believe that the readers will be able to easily understand
and follow the experimental procedures, thanks to the intelligible explanations and notes,
and to construct their own artificial riboswitches. Last but not least, I am also grateful to
Prof. John Walker for giving me the opportunity to edit this book.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
Contributors
ix
x Contributors
Abstract
Selection of aptamers that bind a specific ligand usually begins with a random library of RNA sequences,
and many aptamers selected from such random pools have a simple stem–loop structure. We present here
a computational approach for designing a starting library of RNA sequences with increased formation of
complex structural motifs and enhanced affinity to a desired target molecule. Our approach consists of two
steps: (1) generation of RNA sequences based on customized patterning of nucleotides with increased
probability of forming a base pair and (2) a high-throughput virtual screening of the generated library to
select aptamers with binding affinity to a small-molecule target. We developed a set of criteria that allows
one to select a sequence with potential binding affinity from a pool of random sequences and designed a
protocol for RNA 3D structure prediction. The proposed approach significantly reduces the RNA sequence
search space, thus accelerating the experimental screening and selection of high-affinity aptamers.
Key words Screening library design, Patterned sequences, RNA structure prediction, Virtual screening,
RNA–small molecule docking
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_1, © Springer Science+Business Media New York 2014
1
2 Yaroslav G. Chushak et al.
Fig. 1 Generation of initial RNA sequence pool. Step 1: Generate large sequence pool of sequences using
patterned libraries with the alternation of purines (R = A, G), pyrimidines (Y = U, C), and random bases (N = A, C,
G, U). At this step generated sequences are not target specific and can be used for SELEX-like selection of
binding aptamers or can be used in step 2. At step 2 sequences are filtered based on their secondary structure,
their 3D structure is generated, and virtual screening is used to identify RNA molecules that bind to a target
molecule. Selected RNA molecules that are now specific to the desired target are placed into a pool of
sequences for experimental screening and selection of high-affinity aptamers
2 Materials
3 Methods
3.1 RNA Secondary Analysis of experimentally selected aptamers shows a significant cor-
Structure Analysis relation between the dissociation constant and the free energy of
secondary structure formation. Based on the literature, free energy
of aptamers is significantly lower than the median same-length ran-
dom sequence value [18]. Figure 2 shows the distribution of folding
energies for 10,000 randomly chosen members of patterned libraries
in comparison with the random N40 library. The mean folding energy
for random library ΔGmean = −6.3 kcal/mol, and the standard devia-
tion SD = 3.2 kcal/mol. The mean energy for pattern PT1 is
−8.98 kcal/mol with standard deviation of 3.55 kcal/mol, for pat-
tern PT2 −7.85 kcal/mol and 3.8 kcal/mol, and for pattern PT3
−8.03 kcal/mol and 3.8 kcal/mol, respectively. The cutoff energy
for sequence selection was chosen as ΔGmean − SD = −9.5 kcal/mol
based on values for the random sequences which is higher than the
mean energy value for the patterned sequences.
Next, we analyzed the interaction of nucleic acids with ligands
for several aptamer–ligand complexes based on their experimental
three-dimensional structures [19]. It was found that RNA bases
involved in molecular recognition do not form Watson–Crick pairs
with other bases. There are two possible reasons for this: first,
6 Yaroslav G. Chushak et al.
unpaired RNA bases are more flexible, so they can easily change
their conformation to form a binding pocket and accommodate a
ligand; secondly, unpaired bases have available donor/acceptor
atoms for potential formation of hydrogen bonds with the ligand.
Therefore, we set a constraint that the secondary structure of our
sequences with 40 bases should have at least 11 unpaired bases.
This number seemed optimal, since a higher number of unpaired
bases will significantly reduce the presence of sequences with high
secondary structure free energy while a lower number will reduce
the potential sites for ligand binding.
Based on these findings, we developed a set of criteria that
limited the presence of sequences with abundant simple structural
motifs and maximized the presence of stable low-energy structures.
For the pool of 40-mer sequences these criteria are as follows [4]:
(1) in the lowest energy conformation bases 1 and 2 form pairs
with bases 40 and 39; (2) the free energy of secondary structure
formation is less than −9.5 kcal/mol; (3) there are at least 11 bases
that do NOT form Watson–Crick pairs. Only the sequences that
passed the selection criteria were forwarded to the next step in the
selection process.
The typical steps for the secondary structure generation and
analysis are outlined below.
Computational Design 7
3.1.2 Calculation Next step is to calculate the secondary structure and folding energy
of Folding Energy of the of the generated sequence. We perform that using RNA::fold
Generated Sequences function from the ViennaRNA package:
use RNA;
($s_struct, $f_energy) = RNA::fold($sequence);
The secondary structure of the generated sequence is presented
in bracket-dot notations. In these notations unpaired bases are
presented by dots and base pair between bases i and j is presented
by “(” at position i and “)” at position j, e.g., “(((…((…))..)))”.
3.1.3 Analysis of the Now, perform analysis of the secondary structure. If it satisfies our
Secondary Structure criteria, we save the sequence and its secondary structure into the
of Generated Sequences output files:
open(fasta_FILE, ">Seq1.fasta") || die("Could not
open file:fasta_file !");
8 Yaroslav G. Chushak et al.
3.1.4 Generation of Files Saved sequences and their secondary structure are used to generate
for 3D Modeling three-dimensional structures by Rosetta package. RNA 3D struc-
ture modeling in Rosetta requires as input sequence in FASTA for-
mat (saved in step 3.1.3). As optional file it uses “.param” file that
specifies Watson–Crick base pairs, contained in $s_struct.
Parameter file looks like the following:
STEM PAIR 1 40 W W A
STEM PAIR 2 39 W W A
STEM PAIR 3 38 W W A
STEM PAIR 8 29 W W A
STEM PAIR 9 28 W W A
Our experience shows that predicted structures are more accurate
with the use of parameter file. There is no easy way to create param-
eter file from the $s_structure string. One possible solution is
to use b2c.c function that is distributed with the ViennaRNA pack-
age and is located in folder “Utilities.” This function converts
bracket-dot notations into mfold *.ct file. We modified write_
ct_file subroutine of b2c.c function to produce output file in
Rosetta parameter format. That modified function is named
b2param.c. As input it uses temporary file created in step 3.1.3,
and it is used inside the Perl script:
system (“b2param < Seq1.temp > Seq1.param”;
Computational Design 9
3.2 Generation During the last few years, several groups have proposed different
of RNA Three- approaches for de novo prediction of RNA tertiary structure (see
Dimensional ref. 20 for recent review). We used the Rosetta package [14, 15]
Structures developed by the Baker group at the University of Washington to
predict three-dimensional structures of selected sequences. In
Rosetta modeling algorithm, the RNA sequence is divided into
short fragments containing three nucleotides. The assembly of
fragment from a library of experimental RNA crystal structures is
based on Monte Carlo process with a simplified low-resolution
energy function. The generated models are then refined with the
all-atom potential to provide high-resolution atomistic structures.
This two-step protocol has been named Fragment Assembly of
RNA with Full Atom Refinement (FARFAR) [15]. In the previous
section we created two files for each of the sequences in the pool:
a sequence file in FASTA format (Seq1.fasta) and a parameter
file (Seq1.param) that contains information about the Watson–
Crick base pairing. These two files are used as input files for the 3D
structure prediction (see Note 1).
3.2.2 Sorting Structures The generated structures in output file Seq1.out are in order
Based on Their Score they were generated and not in order of their score. Therefore,
next step is to sort structures based on their score. The first line in
the Seq1.out is a sequence, e.g.:
SEQUENCE: ggcgucacaccuucgggugaagucgcccaccuucgg
10 Yaroslav G. Chushak et al.
3.2.3 Extracting Best Next, we need to extract structures with the lowest score (score is
Structures negative; therefore, best score is the lowest score) from the Seq1.
out file into the *.pdb files. It is widely accepted that ligand binding
can drastically alter the receptor’s conformation [21]. To account
for such conformational flexibility, five to ten of the lowest score
structures for each sequence can be selected and placed into a
library of RNA molecules (see Note 3). This library will be used for
virtual high-throughput screening to select aptamers with binding
affinity to a specified small molecule.
To extract structures we use Rosetta extract_pdb command:
$ROSETTA_HOME/rosetta_source/bin/extract_
pdbs.<exe> @flags_extract -database $ROSETTA_
HOME/rosetta_database
where file flags_extract looks like the following:
-in:file:silent Seq1.out
-in:file:fullatom
Computational Design 11
-out:prefix Seq1
-in:file:tags S_000469 S_000021 S_000004 S_000145 S_000358
The last line specifies a list of structures with the lowest scores
that need to be extracted. By including five 3D structures we
perform ensemble docking and thus account for conformational
flexibility of the RNA molecules.
3.3 Virtual Screening Computational docking is a common tool used to identify small-
of the RNA Molecule molecule ligands that bind to proteins [22]. While most docking
Library methods have been developed mostly for proteins, recent evalua-
tion of AutoDock and DOCK programs has demonstrated their
ability to dock compounds to RNA molecules [16]. We used the
AutoDock Vina and ADT to prepare RNA molecules and small-
molecule ligand for docking and to perform docking.
In the previous section we generated PDB files with 3D coor-
dinates of RNA molecules. Now, we need to create a PDB file with
the coordinates of a small-molecule ligand. One way of doing that
is to use SMILES representation of the molecule. Usually, SMILES
representation of molecule can be found on Wikipedia on the right
side of the screen. Under the “Chemical data” section there is a tag
SMILES. Online SMILES translator at http://cactus.nci.nih.gov/
translate/ can be used to generate 3D coordinates from SMILES
string and to save them into PDB file. Another way to generate
coordinates is to use a commercial package to build a molecule and
to create a PDB file with the coordinates.
Next step is to prepare RNA and ligand molecules for docking
using AutoDock Vina. But, before we proceed to that step, we need
to center the RNA molecules around (0,0,0) coordinate. This can
be done using Open Babel package. The command is as follows:
babel -ipdb Seq1.pdb -opdb Seq1_Ct.pdb -C
where the output file Seq1_Ct.pdb contains the centered
coordinates of the molecule.
To prepare molecules for docking we can use GUI version of
ADT or scripts provided by ADT for batch mode. To use ADT,
please follow the tutorial at http://autodock.scripps.edu/faqs-
help/tutorial/using-autodock-4-with-autodocktools. To prepare
files in batch mode we used two commands provided by ADT. To
prepare ligand molecule:
prepare_ligand4.py -l ligand.pdb
The input file for this command is a PDB file with 3D of ligand
created above. As output it produces ligand.pdbqt file that
includes Gasteiger charges, AutoDock atom types, and special key-
words that establish the torsional flexibility of ligand molecule
(see Note 4).
The similar command is used to prepare receptor (in our case
RNA) molecules:
prepare_receptor4.py -r Seq1_Ct.pdb
12 Yaroslav G. Chushak et al.
4 Notes
Disclaimer
The opinions and assertions contained herein are the private views
of the authors and are not to be construed as official or as reflecting
the views of the US Air Force or the US Department of Defense.
This chapter has been approved for public release with unlimited
distribution.
References
1. Ellington AD, Szostak JW (1990) In vitro pools for in vitro selection. Bioinformatics
selection of RNA molecules that bind specific 23:2959–2960
ligands. Nature 346:818–822 10. Luo X, McKeague M, Pitre S et al (2010)
2. Tuerk C, Gold L (1990) Systematic evolution Computational approaches toward the design
of ligands by exponential enrichment: RNA of pools for the in vitro selection of complex
ligands to bacteriophage T4 DNA polymerase. aptamers. RNA 16:2252–2262
Science 249:505–510 11. Lorenz R, Bernhart SH, Hoener Zu
3. Osborne SE, Ellington AD (1997) Nucleic Siederdissen C et al (2011) ViennaRNA pack-
acid selection and the challenge of combinato- age 2.0. Algorithms Mol Biol 6:26
rial chemistry. Chem Rev 97:349–370 12. Mathews DH, Sabina J, Zuker M, Turner DH
4. Chushak Y, Stone MO (2009) In silico selec- (1999) Expanded sequence dependence of
tion of RNA aptamers. Nucleic Acids Res thermodynamic parameters improves predic-
37:e87 tion of RNA secondary structure. J Mol Biol
5. Davis JH, Szostak JW (2002) Isolation of 288:911–940
high-affinity GTP aptamers from partially 13. Mathews DH, Turner DH (2009) NNDB: the
structured RNA libraries. Proc Natl Acad Sci nearest neighbor parameter database for pre-
99:11616–11621 dicting stability of nucleic acid secondary
6. Carothers JM, Oestreich SC, Davis JH, structure. Nucleic Acids Res 38:280–282
Szostak JW (2004) Informational complexity 14. Leaver-Fay A, Tyka M, Lewis SM et al (2011)
and functional activity of RNA structures. J ROSETTA3: an object-oriented software suite
Am Chem Soc 126:5130–5135 for the simulation and design of macromole-
7. Ruff KM, Snyder TM, Liu DR (2010) cules. Methods Enzymol 487:545–574
Enhanced functional potential of nucleic acid 15. Das R, Karanicolas J, Baker D (2010) Atomic
aptamer libraries patterned to increase sec- accuracy in predicting and designing nonca-
ondary structure. J Am Chem Soc 132: nonical RNA structure. Nat Methods
9453–9464 7:291–294
8. Kim N, Gan HH, Schlick T (2007) A compu- 16. Detering C, Varani G (2004) Validation of
tational proposal for designing structured automated docking programs for docking and
RNA pools for in vitro selection of RNAs. database screening against RNA drug targets.
RNA 13:478–492 J Med Chem 47:4188–4201
9. Kim N, Shin JS, Elmetwaly S et al (2007) 17. Trott O, Olson AJ (2010) AutoDock Vina:
RagPools: RNA-as-graph-pools—a web server improving the speed and accuracy of dock-
for assisting the design of structured RNA ing with a new scoring function, efficient
Computational Design 15
optimization and multithreading. J Comput 22. Taylor RD, Jewsbury PJ, Essex JW (2002) A
Chem 31:455–461 review of protein-small molecule docking
18. Carothers JM, Oestreich SC, Szostak JW methods. J Comput Aided Mol Des 16:
(2006) Aptamers selected for higher-affinity 151–166
binding are not more specific for the target 23. Cruz JA, Blanchet MF, Boniecki M et al
ligand. J Am Chem Soc 128:7929–7937 (2012) RNA-Puzzles: a CASP-like evaluation
19. Patel DJ, Suri AK (2000) Structure, recogni- of RNA three-dimensional structure predic-
tion and discrimination in RNA aptamer com- tion. RNA 18:610–625
plexes with cofactors, amino acids, drugs and 24. Fulle S, Gohlke H (2010) Molecular
aminoglycoside antibiotics. J Biotechnol recognition of RNA: challenges for modelling
74:39–60 interactions and plasticity. J Mol Recognit
20. Laing C, Schlich T (2011) Computational 23:220–231
approaches to RNA structure prediction, anal- 25. Brooks BR, Brooks CL III, Mackerell AD Jr
ysis, and design. Curr Opin Struct Biol et al (2009) CHARMM: the biomolecular
21:306–318 simulation program. J Comput Chem
21. May A, Sieker F, Zacharias M (2008) How to 30:1545–1614
efficiently include receptor flexibility during 26. Case DA, Cheatham TE, Darden T et al
computational docking. Curr Comput Aided (2005) The amber biomolecular simulation
Drug Des 4:143–153 programs. J Comput Chem 26:1668–1688
Chapter 2
Abstract
Artificial riboswitches can be generated by functional coupling of an RNA aptamer for synthetic small
molecule to an expression platform. RNA aptamers that can bind strongly and selectively to their target are
feasible to be used for obtaining more potent artificial riboswitches. In this chapter, we describe tips and
notes for in vitro selection of RNA aptamers targeting synthetic small molecules.
Key words RNA aptamer, In vitro selection, Small molecule, Riboswitch, Chemical biology
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_2, © Springer Science+Business Media New York 2014
17
18 Asako Murata and Shin-ichi Sato
Randomized
T7 promoter
sequence (N60)
DNA pool
Transcription by T7
RNA Polymerase
Regeneration by
transcription
Binding to compound-
immobilized resin
RNA pool
DNA pool
Amplification by
PCR
cDNA
Washing
Reverse
transcription
Unbound RNA
Elution
Fluorescent !
Quencher dye Detection of
(BHQ-1) target RNA
Fig. 2 Application of in vitro-selected aptamers against a small-molecule dye for RNA tagging
2 Materials
3 Methods
Table 1
Reaction mixture for PCR amplification of DNA pools
3.2 In Vitro Selection In vitro selection was performed to isolate RNA aptamers for
Procedures BHQ-1 according to the standard procedure (Fig. 1).
3.2.1 Preparation 1. Assemble the reaction mix in PCR tubes as described in Table 1
of DNA Pools (see Note 2). *1Template DNA: 5′-GAA TTC CGC GTG TGC
ACA CC-N60–GTC CGT TGG GAT CCT CAT GG-3′ (for
preparation of first DNA pool a reverse-transcribed product
was used for the other selection round). *2Forward primer:
5′-GCT AAT ACG ACT CAC TAT AGG GAA TTC CGC
GTG TGC ACA CC-3′ (T7 promoter sequence is underlined).
*3
Reverse primer: 5′-CCA TGA GGA TCC GAA CGG AG-3′.
2. Place the tubes into a thermal cycler and perform the PCR
amplification using the following program: 2-min initial
denaturation at 94 °C, followed by sequential cycles of dena-
turation at 94 °C for 30 s, annealing at 55 °C for 30 s, exten-
sion at 68 °C for 1 min, and final extension at 68 °C for 7 min
(see Note 3).
3. Precipitate the DNA by adding a half volume of 7.5 M ammo-
nium acetate solution and 1.5 volume of isopropanol. Centrifuge
the sample at 10,000 × g for 30 min to pellet the DNA.
4. Remove the supernatant carefully, and rinse the pellet with
70 % ethanol.
5. Air-dry the pellet for 5 min, and dissolve it in 50 μL of RNase-
free water.
Table 2
Reaction mixture for in vitro transcription of reaction
3.2.3 Binding of RNA 1. Place 50 μL of the BHQ-1-immobilized resin (25 % slurry)
Pool to BHQ-Immobilized into an empty column (Micro Bio-Spin Chromatography
Resin Columns) (see Note 7), and wash the resin three times with
150 μL of the binding buffer.
2. Apply the RNA pool solution to the resin and incubate for
30 min on ice. Vortex the sample briefly every 10 min.
3. Drain and wash the resin to remove the unbound RNA species
(see Note 6).
4. Elute bound RNA species three times with 130 μL of the binding
buffer saturated with free BHQ-1 amine (see Note 7).
24 Asako Murata and Shin-ichi Sato
Table 3
Reverse transcription reaction
a
Template RNA 8 μL
10 mM dNTP mix 1 μL
10 μM reverse primer* 1 μL
Total 10 μL
[*Reverse primer: 5′-CCATGAGGATCCGAACGG AC-3′]
↓ 65 °C, 5 min
b
Template RNA/primer mixture 10 μL
5× PrimeScript buffer 4 μL
RNase inhibitor 0.5 μL
PrimeScript reverse transcriptase 1 μL
RNase-free water 4.5 μL
Total 20 μL
↓ 42 °C, 60 min, then 70 °C, 15 min
3.2.4 Reverse 1. Prepare the following mixture (Table 3a) in a PCR tube. Heat
Transcription the sample for 5 min at 65 °C and then chill immediately on
ice. Assemble the reaction mix as shown in Table 3b, and per-
form cDNA synthesis reaction for 60 min at 42 °C, followed
by heat-inactivation of RTase at 70 °C for 15 min.
2. Use 10 μL of the reverse-transcribed product as a template for
the PCR amplification to generate the DNA pool for the next
round of selection (see Note 8).
3. Repeat the selection steps for several rounds to enrich the RNA
species with high affinity to BHQ-1.
Aptamers for Small-Molecules 25
3.3 Cloning and 1. After the final round of selection, digest the resulting DNA
Sequencing of Each pool with EcoRI and BamHI as well as pUC19.
RNA Species in the 2. Ligate the digested insert and vector using DNA Ligation Kit.
Enriched Pool
3. Transform DH5α with the ligation product, and heat shock
and spread the cells on LB plate containing 100 μg/mL of
ampicillin. Incubate the plate at 37 °C overnight.
4. Culture from a single colony, and isolate the plasmid using
standard plasmid isolation procedures.
5. Sequence the insert using primers in the vector backbone.
3.4 Fluorescence To determine if the selected aptamers have the ability to restore the
Measurements fluorescence of the fluorophore–BHQ-1 conjugates, fluorescence
titration of the fluorophore–BHQ-1 conjugates with the aptamers
can be performed. The synthesis of the fluorophore–BHQ-1 con-
jugate was described in our previous report [14].
1. Prepare the dsDNA template containing T7 promoter and each
BHQ-1 aptamer by PCR amplification using the individual clone
as a template. Primers used in the reaction are as follows: the for-
ward primer [5′-GCT AAT ACG ACT CAC TAT AGG GAA
TTC CGC GTG TGC ACA CC-3′ (T7 promoter sequence is
underlined)] and the reverse primer [5′-CCA TGA GGA TCC
GAA CGG AC-3′].
2. Perform a transcription reaction, and purify RNA as described
above in Subheading 3.2.2.
3. Prepare a solution of conjugate AL at the concentration of
2 μM in the binding buffer.
4. Prepare RNA solution at the concentrations of 0, 0.6, 2, 6, 20,
60, and 200 μM in the binding buffer.
5. Mix an equal volume of the RNA solution with the conjugate
solution, and measure the fluorescence of each sample using either
a plate reader or a fluorophotometer. The excitation maximum of
each conjugate and the emission maximum in the RNA-bound
state are as follows: conjugate AL (Ex/Em = 543 nm/610 nm),
conjugate Cy (Ex/Em = 519 nm/562 nm), and conjugate FL
(Ex/Em = 497 nm/515 nm).
The Kd values of each aptamer can be obtained by fitting fluo-
rescence titration data using the following equation:
æ ([conjugate ] + [aptamer ] + K d ) - ö
ç T T
÷
( )
1/ 2
ç ([conjugate ] + [aptamer ] + K )2 - 4 [conjugate ] [aptamer ] ÷
ç T T d T T ÷
Fobs = Aç ÷
è 2 [conjugate ]T ø
26 Asako Murata and Shin-ichi Sato
Table 4
In transcription/translation reaction mixture
Final conc.
Template DNA X μL 0.5 μg
E. coli lysate 4.8 μL
Reaction mix 4 μL
Amino acids 4.8 μL
Methionine 0.4 μL
Reconstitution buffer 2 μL
100 μM fluorophore–BHQ-1 conjugate 0.5 μL 2.5 μM
RNase-free water 3.5 − X μL
Total 20 μL
4 Notes
are removed from the reaction after 4, 8, 12, 16, 20, 24, 28,
and 32 cycles and separated on an 8 % native polyacrylamide
gel. As the population of binding species becomes enriched, a
band of the desired product will appear at an earlier PCR cycle
than the previous selection round.
4. We use NAP-5 column to remove unincorporated NTPs, but
any other gel filtration system can be used.
5. Do not heat the RNA solution in the presence of Mg2+. This is
important to avoid possible chemical degradation of RNA with
divalent cations.
6. We usually measure the UV absorbance of the RNA pool
solution before and after binding to the resin to determine the
percentage of bound RNA in each cycle. The percentage of
RNA bound to the resin is calculated as 100(AbsT − AbsF)/
AbsT, where AbsT is UV absorbance of the RNA solution
before binding to the resin, and AbsF is UV absorbance of the
flow-through fraction after binding to the resin.
7. Do not use a spin column with a microporous membrane such
as PVDF, because BHQ-1 amine may adsorb to the membrane,
probably due to the hydrophobicity of BHQ-1.
8. We usually save a half of the reverse-transcribed product for
archival purposes. If subsequent amplification or selection
reactions fail for any reason, the procedure can be started from
the archival pool.
Acknowledgment
References
1. Roth A, Breaker RR (2009) The structural and 6. Wieland M, Benz A, Klauser B, Hartig JS
functional diversity of metabolite-binding (2009) Artificial ribozyme switches containing
riboswitches. Annu Rev Biochem 78:305–334 natural riboswitch aptamer domains. Angew
2. Breaker RR (2011) Prospects for riboswitch Chem Int Ed 48:2715–2718
discovery and analysis. Mol Cell 16(43): 7. Dixon N, Duncan JN, Geerlings T, Dunstan
867–879 MS, McCarthy JE, Leys D, Micklefield J
3. Serganov A, Nudler E (2013) A decade of (2010) Reengineering orthogonally selective
riboswitches. Cell 152:17–24 riboswitches. Proc Natl Acad Sci U S A 107:
4. Winkler W, Nahvi A, Breaker RR (2002) 2830–2835
Thiamine derivatives bind messenger RNAs 8. Werstuck G, Green MR (1998) Controlling
directly to regulate bacterial gene expression. gene expression in living cells through small mol-
Nature 419:952–956 ecule-RNA interactions. Science 282:296–298
5. Nomura Y, Yokobayashi Y (2007) Reengineering 9. Suess B, Hanson S, Berens C, Fink B,
a natural riboswitch by dual genetic selection. Schroeder R, Hillen W (2003) Conditional
J Am Chem Soc 129:13814–13815 gene expression by controlling translation with
28 Asako Murata and Shin-ichi Sato
tetracycline-
binding aptamers. Nucleic Acids 12. Weigand JE, Sanchez M, Gunnesch EB, Zeiher
Res 31:1853–1858 S, Schroeder R, Suess B (2008) Screening for
10. Suess B, Fink B, Berens C, Stentz R, Hillen W engineered neomycin riboswitches that con-
(2004) A theophylline responsive riboswitch trol translation initiation. RNA 14:89–97
based on helix slipping controls gene expres- 13. Ellington AD, Szostak JW (1990) In vitro
sion in vivo. Nucleic Acids Res 32:1610–1614 selection of RNA molecules that bind specific
11. Lynch SA, Desai SK, Sajja HK, Gallivan JP ligands. Nature 346:818–822
(2007) A high-throughput screen for synthetic 14. Murata A, Sato S, Kawazoe Y, Uesugi M (2011)
riboswitches reveals mechanistic insights into Small-molecule fluorescent probes for specific
their function. Chem Biol 14:173–184 RNA targets. Chem Commun 47:4712–4714
Chapter 3
Abstract
Photoresponsive artificial riboswitch has the potential to offer a de novo method for spatiotemporal control
of gene expression in living cells. Because, even today, it is difficult to design a small molecule binding to
a specific RNA sequence, generating an artificial riboswitch that possesses highly specific affinity to a ligand
of interest basically depends on in vitro selection procedure where a variety of RNA–ligand complexes can
be obtained in established methods. Here, we describe the protocol for in vitro aptamer selection against
a photoresponsive peptide ligand containing azobenzene moiety that undergoes photoisomerization
through light irradiation. Furthermore, we explain a procedure for surface plasmon resonance assay to
detect photoswitchable association and dissociation of RNA–ligand complex on gold surface.
Key words In vitro selection, RNA aptamer, Azobenzene, Photoresponsive association and dissocia-
tion, Gene regulation
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_3, © Springer Science+Business Media New York 2014
29
30 Gosuke Hayashi and Kazuhiko Nakatani
2 Materials
3 Methods
3.1 In Vitro Selection 1. Shake the bottle of Affi-Gel 10® Gel to uniformly suspend gel.
Against KRAzR Peptide 2. Take 1 mL of the agarose gel (about 2 mL of the slurry) into
3.1.1 Preparation Econo column (see Note 2).
of KRAzR- and Glycine- 3. Discard the preloaded solution from the bottom of the
Agarose Columns column.
4. Add 3-column volume of gel washing solution (3 mL) to com-
pletely remove the preloaded solution.
5. Equilibrate the gel by adding 2-column volume of coupling
buffer (2 mL).
6. Cap the bottom of the column and add 2 mL of 5 mM KRAzR
dissolved in the coupling buffer.
7. Leave the column on a rotating shaker for 1 h (see Note 3).
8. Drain the reaction solution, and then wash the column with
5-column volume of binding buffer (5 mL).
9. To make agarose gel for negative selection (see Note 4), use
0.5 mL of gel slurry and 1 mL of 10 mM glycine dissolved in
coupling buffer instead of 1 mL gel and 5 mM KRAzR solu-
tion, respectively. The other procedures are same as that
described above.
Fig. 1 Apparatus used in our in vitro selection against KRAzR peptide. Glycine-
agarose packed column and KRAzR-agarose packed column were tandemly
placed to efficiently transfer flow-through of glycine column onto KRAzR column
34 Gosuke Hayashi and Kazuhiko Nakatani
3.1.5 Reverse 1. Incubate the recovered RNA solution at 80 °C for 3 min, and
Transcription (RT)-PCR then place it on ice (see Note 11).
2. Add appropriate amount of dNTPs, reverse transcription buf-
fer, MgCl2, RNase inhibitor, primer, and AMV reverse tran-
scriptase according to the manufacturer’s protocol.
3. Incubate the total volume of 50 μL reaction mixture at 42 °C
for 30 min.
4. Using all of RT reaction mixture, prepare 500 μL of PCR reac-
tion with ExTaq polymerase, according to the procedure as
described above. Dispense the reaction mixture into ten tubes
(50 μL each).
5. To determine the optimum PCR cycles, begin with the 50 μL
volume and take 5 μL in each four cycles. PCR amplification
should be done in the condition of 98 °C, 10 s; 55 °C, 30 s;
and 72 °C, 30 s.
6. From PAGE analysis, determine the appropriate PCR cycle
number (Fig. 2a) (see Note 12). In our case, we chose 11 cycles.
7. Run PCR reaction with the remaining 450 μL of the reaction
mixture, and check the PCR product by PAGE (Fig. 2b).
Photoswitchable Ligand-Aptamer Complexes 35
Fig. 2 Native PAGE analysis of RT-PCR products obtained at first round of selection. (a) Optimization of PCR
cycles using 50 μL of PCR mixture. In each four cycles, 5 μL of the reaction mixture was taken for PAGE analysis.
After electrophoresis, the gel was stained with SYBR Green. (b) Confirmation of migration of RT-PCR products
(11 cycles) obtained in large-scale reaction. After electrophoresis, the gel was stained with SYBR Green
3.1.6 Next Rounds 1. To proceed to the second round of the selection, prepare
of Selection 200 μL of transcription mixture and react with the same
condition described above using half of the transcribed RNA
for loading to selection column. The other half should be
stored in −20 °C refrigerator. To cycle selection procedures
including negative and positive selection, RT-PCR, and
transcription, basically follow the protocol described above
(see Notes 13 and 14).
2. When you confirm the enrichment of RNA bound to the
KRAzR-gel after several rounds of selection (see Note 15),
stop the selection cycle after RT-PCR.
3.1.7 Isolation and 1. Add BamHI and EcoRI to the obtained PCR product (DNA
Identification of Aptamer library), which is dissolved in appropriate buffer according to
Sequences the manufacturer’s protocol and incubate at 37 °C for 3 h.
2. Treat pUC18 in the same condition in a 200 μL tube.
3. After 37 °C incubation, place both the DNA library and
pUC18 at 70 °C for 10 min for deactivation of restriction
enzymes.
4. Prepare 10 μL of ligation solution including 5 μL of Mighty
Mix, about 25 fmol of cleaved pUC18, and about 25–250 fmol
of cleaved DNA library.
36 Gosuke Hayashi and Kazuhiko Nakatani
3.2 SPR Analysis Basically, follow the instruction manual of Biacore 2000. Use CM5
of Photoresponsive sensor chip throughout all procedures. Immobilization of KRAzR
Interaction Between onto the gold surface was done through amide bond formation.
RNA Aptamers and
KRAzR Peptide
3.2.2 Sample 1. To prepare RNA aptamer samples for Biacore analysis, PCR-
Preparation amplify template DNAs from plasmid DNAs.
2. Transcribe RNA aptamers, and purify them by 8 % denaturing
PAGE and 2-propanol precipitation.
3. Dissolve the obtained pellet with binding buffer used in the
selection procedure, and prepare 1.0 μM of each sample
(see Note 16).
3.2.3 SPR Analyses 1. Set running buffer (binding buffer) and RNA samples in the
Biacore 2000.
2. Flow running buffer for 90 s, and then inject aptamer sample
for 120 s for binding to the KRAzR surface (association).
3. Flow the running buffer again for 120 s for dissociation.
Photoswitchable Ligand-Aptamer Complexes 37
360 nm 430 nm
a b
5 min. 5 min.
Response Unit
Response Unit
Response Unit
buffer gold surface 300 300 300
carrying Az-peptide
glass 0 0 0
0 100 200 300 400 0 100 200 300 400 0 100 200 300 400
Time (sec.) Time (sec.) Time (sec.)
Fig. 3 Photoresponsive interaction between RNA aptamers and KRAzR peptide. (a) Illustration of light irradia-
tion to the SPR sensor chip covered with droplet of running buffer. (b) SPR sensorgrams before and after light
irradiation. Two different RNA aptamers (red and blue line) were analyzed. As negative control, running buffer
was injected instead of RNA aptamers (green line)
4 Notes
Acknowledgements
References
1. Mandal M, Breaker RR (2004) Gene regula- underlying riboswitch function. Annu Rev
tion by riboswitches. Nat Rev Mol Cell Biol Biophys 41:343–370
5:451–463 5. Soukup GA, Breaker RR (1999) Engineering
2. Serganov A, Patel DJ (2007) Ribozymes, ribo- precision RNA molecular switches. Proc Natl
switches and beyond: regulation of gene Acad Sci U S A 96:3584–3589
expression without proteins. Nat Rev Genet 6. Werstuck G, Green MR (1998) Controlling
8:776–790 gene expression in living cells through small
3. Winkler WC, Breaker RR (2005) Regulation molecule-RNA interactions. Science
of bacterial gene expression by riboswitches. 282:296–298
Annu Rev Microbiol 59:487–517 7. Bauer G, Suess B (2006) Engineered ribo-
4. Serganov A, Patel DJ (2012) Metabolite rec- switches as novel tools in molecular biology.
ognition principles and molecular mechanisms J Biotechnol 124:4–11
40 Gosuke Hayashi and Kazuhiko Nakatani
8. Sinha J, Reyes SJ, Gallivan JP (2010) 11. Wilson DS, Szostak JW (1999) In vitro selec-
Reprogramming bacteria to seek and destroy tion of functional nucleic acids. Annu Rev
an herbicide. Nat Chem Biol 6:464–470 Biochem 68:611–647
9. Suess B, Fink B, Berens C, Stentz R, Hillen W 12. Hayashi G, Hagihara M, Dohno C, Nakatani
(2004) A theophylline responsive riboswitch K (2007) Photoregulation of a peptide-RNA
based on helix slipping controls gene expres- interaction on a gold surface. J Am Chem Soc
sion in vivo. Nucleic Acids Res 32:1610–1614 129:8678–8679
10. Suess B, Hanson S, Berens C, Fink B, Schroeder R, 13. Hayashi G, Hagihara M, Nakatani K (2009)
Hillen W (2003) Conditional gene expression by RNA aptamers that reversibly bind photores-
controlling translation with tetracycline-binding ponsive azobenzene-containing peptides.
aptamers. Nucleic Acids Res 31:1853–1858 Chemistry 15:424–432
Chapter 4
Abstract
Artificial riboswitches that respond to the concentrations of intracellular proteins are a promising tool with
a variety of applications. They can be designed and engineered using existing RNA aptamers that target
proteins. Aptamers are generated via an iterative selection–amplification process, known as systematic
evolution of ligands by exponential enrichment (SELEX). This chapter describes a SELEX procedure for
the identification of RNA aptamers against hexa-histidine-tagged proteins. For the efficient enrichment of
higher affinity aptamers, the selection stringency should be gradually increased. Undesired RNA species
that bind to affinity resins can be eliminated from the pool by using a negative selection step and alternat-
ing different types of resins.
Key words Aptamer, SELEX, His-tag, In vitro selection, RNA, Artificial riboswitch, Synthetic gene
circuit
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_4, © Springer Science+Business Media New York 2014
41
42 Shoji Ohuchi
Fig. 1 Schematic diagram of SELEX for the identification of RNA aptamers against a hexa-histidine-tagged
protein. Through negative selection, undesired RNA species that bind to the resin (indicated by dark gray) are
eliminated. The unbound RNA is recovered and used for positive selection. After extensive washing, RNA spe-
cies that bind tightly to the target (indicated by black) are isolated and amplified by RT-PCR. Following in vitro
transcription regenerates an enriched RNA pool for the next round of SELEX. These processes are repeated
until the desired aptamers are enriched in the pool
2 Materials
3 Methods
3.1 Random DNA Two alternative methods are described blow (Fig. 2). The first
Pool Construction method (Subheading 3.1.1) is more common. The second method
(Subheading 3.1.2) requires longer sequences of synthetic DNA
but no enzymatic reaction.
3.1.1 DNA Pool 1. Extension reaction (Fig. 2a): The total reaction volume is
Preparation by Extension 500 μL: 1× PCR buffer (supplemented), 0.2 mM of each
Reaction dNTP (supplemented), 1 μM R3-N40 (template DNA), 5 μM
Fig. 2 Procedures for the preparation of the random DNA pool: (a) Extension reaction-based procedure.
(b) Annealing-based procedure. Boxes and “N” represent the primer regions and the random sequence,
respectively. Gray arrows indicate the orientation of the extension reaction
SELEX Against His-Tagged Proteins 45
3.1.2 DNA Pool By using single-stranded DNA template that contains a full pro-
Preparation by Primer moter region, the DNA pool can be prepared without enzymatic
Annealing (Alternative reaction. The annealing of a forward primer to the template makes
Method) a transcriptionally competent, double-stranded promoter region
(Fig. 2b) [8].
The total volume is 40 μL: 0.1× PCR buffer, 10 μM
R3-N30+T7p, and 10 μM R3f. After denaturing for 1 min at
96 °C, anneal the DNA under the following conditions using a
thermocycler: 70 °C for 1 min; 60 °C for 1 min; 50 °C for 1 min;
and 40 °C for 5 min. All of the annealed DNA will be used for in
vitro transcription.
3.3.3 Positive Selection 1. Spin down the target-immobilized resin (Subheading 3.3.2),
and remove the supernatant. Add the RNA solution from the
negative selection (Subheading 3.3.1) to the resin. Incubate
the tube at room temperature with gentle rotation for 30 min
using a rotator (see Note 10).
2. Spin down the resin, and remove the supernatant containing
unbound RNA. To remove weakly bound RNA, wash the resin
with 100 μL of buffer A four times.
3. Spin down the resin, and remove the supernatant. To release
the target protein from the resin, resuspend the resin in 100 μL
of buffer C. Incubate the tube at room temperature with gen-
tle rotation for 10 min using a rotator (see Notes 10 and 12).
4. Collect the supernatant containing the released protein
and RNA bound to the protein. Recover the bound RNA
by PCI extraction followed by ethanol precipitation with a
co-precipitant (see Note 13).
Fig. 3 Desired and undesired reactions during PCR. (a) Desired reactions during the extension step.
(b) Undesired side reactions. Because of the random sequence in the DNA pool, unintended mis-annealing may
occur. These side reactions generate longer and shorter by-products. Boxes and “N” represent the primer
regions and the random sequence, respectively. Gray arrows indicate the orientation of the extension reaction
3.4 Analysis of The number of SELEX rounds necessary depends on the properties
SELEX Progress of the target protein and the selection conditions employed. Thus,
it is impossible to know how many rounds to perform a priori, and
the progress of SELEX should be experimentally confirmed by
analyzing the affinity enrichment of the RNA pool (see Note 20).
48 Shoji Ohuchi
Fig. 4 Optimization of PCR cycle number. Too many PCR cycles generate unde-
sired longer and shorter by-products, which can be recognized by a smear band
pattern in PAGE analysis. Therefore, the number of cycles that is necessary and
sufficient should be determined. In the case shown, 16–18 cycles are optimal
Table 1
Conditions for positive selection against a His-tagged target protein
3.4.1 Pull-Down Assay 1. Prepare 6 μL of the resin with 200 pmol of the target protein
immobilized as described above (Subheading 3.3.2). Divide it
equally into two 0.2-mL PCR tubes. As a resin control, wash
6 μL of empty resin and divide it equally into two 0.2-mL PCR
tubes (see Notes 21 and 22).
2. Dilute 200 pmol of the RNA pool to be tested with water to
96 μL, fold the RNA, and add tRNA and BSA as described
above (Subheading 3.3.1). The total volume of the RNA sam-
ple is 200 μL. As an RNA control, use 200 pmol of RNA from
the initial random pool.
3. Remove the supernatant from the resin tubes, and add 90 μL
of each RNA sample onto each resin.
4. Incubate the resin tubes at room temperature with gentle rota-
tion for 30 min using a rotator (see Note 10).
5. Spin down the resin and wash with 100 μL of buffer A twice.
After removing the supernatant, resuspend the resin in 10 μL
of the denaturing solution. As an input sample control, transfer
9 μL of the remaining folded RNA to a 0.2-mL PCR tube and
add 9 μL of the denaturing solution.
6. Heat the samples at 90 °C for 3 min and chill quickly to 4 °C
using a thermocycler. Separate the samples on 12 % denaturing
PAGE containing 7 M urea. Soak the gel with SYBR Green II
to stain the RNA bands, and analyze the band image using a
phosphorimager. If SELEX works successfully, and the desired
aptamers are enriched in the pool, the RNA pool binds to the
protein-immobilized resin but not to the empty resin (Fig. 5).
50 Shoji Ohuchi
Fig. 6 Example of EMSA results. When SELEX progresses successfully, the RNA
pool (in this case, the Round 10 pool, designated “R10”) forms complexes with
the target in a protein concentration-dependent manner. Not all RNA molecules
form complexes even when the protein concentration is high due to mis-folding
of the aptamers and the presence of inactive RNA species in the pool. “40N”
indicates the control RNA
3.4.2 Electrophoretic 1. Dilute 5 pmol of the RNA pool to be tested with water to
Mobility Shift Assay 12 μL, fold the RNA, and add tRNA and BSA as described
(Alternative Method) above (Subheading 3.3.1). The total volume of the RNA sam-
ple is 25 μL. As an RNA control, use 5 pmol of RNA from the
initial random pool (see Note 21).
2. Dilute 2.5, 10, or 40 pmol of the target protein with buffer A
to 12.5 μL.
3. Mix 5 μL of each folded RNA and 5 μL of the diluted protein.
As a no-protein control, mix 5 μL of each folded RNA and
5 μL of buffer A. The final concentrations of RNA and protein
are 100 nM and 0 nM, 100 nM, 400 nM, or 1,600 nM,
respectively.
4. Incubate at 37 °C for 30 min.
5. Chill on ice, and add 2 μL of the 6× loading solution.
6. Run the TBM gel at 75 V for 2 h in a cold room (see Note 23).
7. Soak the gel with SYBR Green II to stain the RNA bands, and
analyze the band image using a phosphorimager. If SELEX
works successfully, and the desired aptamers are enriched in the
pool, the RNA pool forms a complex with the protein, and
mobility-shifted bands can be detected (Fig. 6).
3.4.3 Filter-Binding 1. Dilute 0.1 pmol of the radioisotope-labeled RNA pool with
Assay (Alternative Method) water to 120 μL, fold the RNA, and add tRNA and BSA as
described above (Subheading 3.3.1). The total volume of the
SELEX Against His-Tagged Proteins 51
3.5 Cloning Once affinity enrichment of the RNA pool is confirmed, clone the
and Evaluation variants by TA cloning. Several TA cloning vectors are commercially
of Aptamers available [i.e., pGEM-T Easy Vector System (Promega); TA Cloning
Kit with pCR (Invitrogen)]. Sequencing more than 30 clones is
3.5.1 Cloning
recommended. Typically, similar or even identical sequences appear
and Sequencing
multiple times. This sequence convergence indicates successful
enrichment of certain clones via SELEX (see Note 28).
3.5.2 Affinity Estimation To evaluate the affinity of each clone, perform a binding assay as
of Each Clone described for the analysis of SELEX progress (Subheading 3.4). At
this stage, determining the precise value of the affinity constant is
desirable. Thus, an EMSA (Subheading 3.4.2) or a filter-binding
assay (Subheading 3.4.3) with radioisotope-labeled RNA is recom-
mended. Alternatively, other interaction analysis techniques, like
surface plasmon resonance analysis or fluorescence correlation
spectroscopy, are useful [9–12].
3.5.3 Evaluation of Typically, the full-length sequence of the original isolate is not
Essential Elements required for the binding activity. In addition, a shorter sequence
for Binding Activity and simpler structure are beneficial for the design and engineer-
ing of artificial riboswitches employing the obtained aptamers.
52 Shoji Ohuchi
Fig. 7 Generation of a shortened aptamer. Initially, truncations from the 5′- and 3′-ends of the original isolate,
an aptamer against T7 RNA polymerase (T06), were examined. A shortened derivative (T06-38) was generated
without reducing the affinity. Based on the shortened derivative, secondary SELEX was performed, and further
truncation was examined. The resulting 29-nt aptamer (T230-29) is 42 nt shorter than the original aptamer;
its affinity for the target protein is higher. In addition, the secondary SELEX revealed the sequence and struc-
tural features essential for activity (for details, see ref. 13)
4 Notes
References
Abstract
Riboswitches have a number of characteristics that make them ideal regulatory elements for a wide range of
synthetic biology applications. To maximize their utility, methods are required to create custom riboswitches
de novo or to modify existing riboswitches to suit specific experimental needs. This chapter describes such a
method, which exploits fluorescence-activated cell sorting (FACS) to quickly and efficiently sort through
large libraries of riboswitch-like sequences to identify those with the desired activity. Suggestions for the
experimental setup are provided, along with detailed protocols for testing and optimizing FACS conditions
FACS selection steps, and follow-up assays to identify and characterize individual riboswitches.
Key words Artificial riboswitch, Aptamer, Biosensor, Fluorescence-activated cell sorting (FACS),
High-throughput, Enrichment
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_5, © Springer Science+Business Media New York 2014
57
58 Zohaib Ghazi et al.
Fig. 1 Isolation of functional riboswitches in E. coli using FACS. Following the transformation of the plasmid
library (small circle), which contains riboswitch candidates placed within the 5′ UTR of an FP gene. E. coli cells
are sorted based on fluorescence intensity to isolate cells that are highly fluorescent in the presence of the
target molecule that regulates the riboswitch, but less so in its absence
2 Materials
2.3 96-Well Plate 1. 96-well, flat bottom polystyrene non-treated sterile assay plates
Fluorescence Assay (Corning).
2. 96-well, half-area flat bottom polystyrene NBS plates
(Corning).
3. ABgene 96-well, square-well storage plates (Thermo Fisher
Scientific).
3 Methods
3.1.2 Library Design Once an aptamer has been chosen, the next step before beginning
the riboswitch selection is to design a plasmid-borne library of can-
didate riboswitches. The only region of the plasmid that will vary
between individual members of the library is the “riboswitch
region,” which resides in the 5′ untranslated region (UTR) of a
fluorescent protein. To drive the expression of the transcript that
contains the riboswitch region and the fluorescent protein, it is
advisable to use a well-characterized “constitutive” promoter,
which is as stable as possible under the selection conditions that
will be used. Two important considerations for the library plasmid
are its origin of replication (copy number) and the strength of the
promoter. For each of these two factors, the required signal
strength for the FACS steps must be balanced with the potential
complications and intrusiveness associated with protein overex-
pression. In addition, for riboswitches with high-affinity aptamers
that detect low levels of the target molecule, it is possible that,
within the aptamer’s dynamic range, the target molecule may not
be in excess over the transcript when using high copy plasmids and
strong promoters.
The methodology described in this chapter is intended to be
generally applicable for a range of riboswitch designs and mecha-
nisms. The focus is therefore on setting up and performing the
selection that allows for the identification of active riboswitches
from a starting library and on follow-up experiments to identify
and characterize individual sequences. There are limitless options
for the design of the starting library, which could target ribo-
switches that act at the level of transcription [19, 20], translation
[10–13, 21], or RNA processing [22–24]. Specific library designs
are not discussed here. Generally, the library will consist of a
combination of invariant and random residues in the 5′ UTR
shortly upstream of a fluorescent protein (FP). The aptamer domain,
which is typically held largely constant to maintain the binding
62 Zohaib Ghazi et al.
3.1.3 The Fluorescent Decades of intense research has yielded an abundance of FPs with
Protein diverse properties. Different FPs can vary greatly in their excitation/
emission spectra, extinction coefficient and quantum yield, matu-
ration speed and efficiency, turnover rate, environmental sensitivity,
photostability, oligomerization state, etc. A number of excellent
Selecting Artificial Riboswitches by FACS 63
3.1.4 Instrument High-speed cell sorters are available from several manufacturers,
Considerations such as the FACSAria™ (BD Biosciences), EPICS® ALTRA™
(Beckman Coulter), or the MoFlo® (Dako) cell sorters. Flow
cytometers, particularly those with sorting capabilities, are tremen-
dously valuable for a wide range of applications and are very com-
plex and expensive instruments. Accordingly, many research
institutes have a limited number of flow cytometers that are shared
in common facilities, often operated exclusively by specially trained
personnel. For those that do not have ready access to a capable
instrument that allows the sorting of bacteria, many centralized
facilities permit access to users from other institutes at hourly rates.
While most modern instruments are compatible with the experi-
ments described here, there are several issues to consider before
64 Zohaib Ghazi et al.
3.2 Establishing Prior to beginning selection steps, it is important to test the feasibility
Controls and FACS of your particular project, to experience running your samples on the
Parameters flow cytometer that will be employed, to gain insight into the proper-
ties of your samples, and to determine the gating and other settings
that will best suit your selection. This section presents a set of experi-
ments that will help accomplish these goals. Depending upon the
success of these experiments, additional tests along these same lines
may be required in order to prepare for the selection.
At a minimum, these tests require two plasmids to be stably
introduced in the strain that will be used for the selection:
1. The library plasmid containing the FP you have chosen but
lacking any potential regulatory elements in its 5′ UTR
(FP-positive control plasmid).
2. A similar plasmid that contains a different antibiotic resistance
cassette and that lacks a fluorescent protein (FP-negative control
plasmid).
Selecting Artificial Riboswitches by FACS 65
Fig. 3 Schematic illustrations of the expected fluorescence histograms for (a) cells containing the FP-positive
control plasmid, (b) cells containing the FP-negative control plasmid, (c) a 50:50 mixture of the FP-positive and
FP-negative cells, (d) a 95:5 mixture of the FP-positive and FP-negative cells, and (e) a 95:5 mixture of the
FP-negative and FP-positive cells. Each dash line with an arrow indicates an example of the gating threshold
settings that can be used for sorting, with the arrow indicating the cells to be kept from the sort
3.4 Identifying The two-step FACS selection described in Subheading 3.3 can be
and Characterizing repeated any number of times prior to checking the progress of the
Individual selection and seeking individual riboswitch candidates. For most
Riboswitches projects, two to three rounds are likely to be sufficient; however,
this will depend on factors such as the frequency of desirable
sequences in the library, the size of the library, and the extent of
purification that is achieved at each FACS step. Importantly, the
pool of clones that remains after each FACS step must be saved
(as described in the previous section), and so it is always possible to
return to any stage in the selection process. One addition that can
help improve the final outcome of your project is to identify
the most active riboswitch sequences from a given selection
(populations 2A, 2B, 3A, #B, etc) and then to build a second
library based on these sequences with random residues introduced
at key locations [9]. The process of identifying and improving
upon promising sequences can allow you to more shrewdly survey
the almost limitless sequence space that exists in a riboswitch-sized
oligonucleotide library.
Even after your library has been significantly enriched, it will
still contain a large number of unique sequences with varying levels
of activity. The procedure described below allows for the identifica-
tion of the most active individual sequences from your final, most
enriched population.
1. Thaw an aliquot of the population of cells that you wish to
survey on ice for 10–15 min.
2. Create several serial tenfold dilutions of the thawed cells in LB,
plate onto LB agar plates containing the appropriate antibiotic,
and incubate overnight at 37 °C. The goal of this step is to
generate plates containing large numbers of well-separated,
single colonies.
3. Fill a 96-well plate with 200 μL of LB plus the appropriate
antibiotic in each well. Inoculate each well with a single colony
from step 2. Set aside a few wells and inoculate them with cells
carrying the FP-positive and the FP-negative control vectors to
serve as controls for subsequent fluorescence assays. Grow
overnight (~16 h) with shaking at 37 °C.
4. The following day, fill a 96-well storage plate (ABgene) with
100 μL per well of LB + 30 % glycerol. Using a multichannel
pipette, add 100 μL per well of the plate from step 3 to the
Selecting Artificial Riboswitches by FACS 71
4 Notes
Acknowledgments
References
1. Roth A, Breaker RR (2009) The structural and metabolite binding kinetics operate an FMN
functional diversity of metabolite-binding ribo- riboswitch. Mol Cell 18:49–60
switches. Annu Rev Biochem 78:305–334 17. Wickiser JK, Cheah MT, Breaker RR et al
2. Fowler CC, Brown ED, Li Y (2010) Using a (2005) The kinetics of ligand binding by an
riboswitch sensor to examine coenzyme B(12) adenine-sensing riboswitch. Biochemistry 44:
metabolism and transport in E. coli. Chem Biol 13404–13414
17:756–765 18. Gilbert SD, Stoddard CD, Wise SJ et al
3. Topp S, Gallivan JP (2007) Guiding bacteria (2006) Thermodynamic and kinetic charac-
with small molecules and RNA. J Am Chem terization of ligand binding to the purine
Soc 129:6807–6811 riboswitch aptamer domain. J Mol Biol 359:
4. Blount KF, Breaker RR (2006) Riboswitches as 754–768
antibacterial drug targets. Nat Biotechnol 24: 19. Trausch JJ, Ceres P, Reyes FE, Batey RT
1558–1564 (2011) The structure of a tetrahydrofolate-
5. Sudarsan N, Hammond MC, Block KF et al sensing riboswitch reveals two ligand binding
(2006) Tandem riboswitch architectures sites in a single aptamer. Structure 19:
exhibit complex gene control functions. 1413–1423
Science 314:300–304 20. Wachsmuth M, Findeiß S, Weissheimer N et al
6. Jin Y, Watt RM, Danchin A, Huang J (2009) (2013) De novo design of a synthetic ribo-
Use of a riboswitch-controlled conditional switch that regulates transcription termination.
hypomorphic mutation to uncover a role for Nucleic Acids Res 41:2541–2551
the essential csrA gene in bacterial autoaggre- 21. Suess B, Fink B, Berens C et al (2004) A the-
gation. J Biol Chem 284:28738–28745 ophylline responsive riboswitch based on helix
7. Djordjevic M (2007) SELEX experiments: new slipping controls gene expression in vivo.
prospects, applications and data analysis in Nucleic Acids Res 32:1610–1614
inferring regulatory pathways. Biomol Eng 24: 22. Win MN, Smolke CD (2007) A modular and
179–189 extensible RNA-based gene-regulatory platform
8. Stoltenburg R, Reinemann C, Strehlitz B for engineering cellular function. Proc Natl
(2007) SELEX—a (r)evolutionary method to Acad Sci U S A 104:14283–14288
generate high-affinity nucleic acid ligands. 23. Wieland M, Hartig JS (2008) Improved apta-
Biomol Eng 24:381–403 zyme design and in vivo screening enable
9. Fowler CC, Brown ED, Li Y (2008) A FACS- riboswitching in bacteria. Angew Chem Int Ed
based approach to engineering artificial ribo- 47:2604–2607
switches. ChemBioChem 9:1906–1911 24. Ogawa A, Maeda M (2007) Aptazyme-based
10. Lynch SA, Gallivan JP (2009) A flow cytometry- riboswitches as label-free and detector-free
based screen for synthetic riboswitches. Nucleic sensors for cofactors. Bioorg Med Chem Lett
Acids Res 37:184–192 17:3156–3160
11. Lynch SA, Desai SK, Sajja HK et al (2007) 25. Beisel CL, Smolke CD (2009) Design princi-
A high-throughput screen for synthetic ribo- ples for riboswitch function. PLoS Comput Biol
switches reveals mechanistic insights into their 5:e1000363
function. Chem Biol 14:173–184 26. Regulski EE, Breaker RR (2008) In-line prob-
12. Topp S, Gallivan JP (2008) Random walks to ing analysis of riboswitches. Methods Mol Biol
synthetic riboswitches—a high-throughput 419:53–67
selection based on cell motility. ChemBioChem 27. Tsien RY (1998) The green fluorescent protein.
9:210–213 Annu Rev Biochem 67:509–544
13. Muranaka N, Abe K, Yokobayashi Y (2009) 28. Shaner NC, Steinbach PA, Tsien RY (2005)
Mechanism-guided library design and dual A guide to choosing fluorescent proteins. Nat
genetic selection of synthetic OFF riboswitches. Methods 2:905–909
ChemBioChem 10:2375–2381 29. Telford WG, Hawley T, Subach F et al (2012)
14. Shapiro HM (2003) Practical flow cytometry, Flow cytometry of fluorescent proteins.
4th edition. Wiley-Liss, New York, NY Methods 57:318–330
15. Macey MG (2007) Flow cytometry: principles 30. Chudakov D, Matz M, Lukyanov S et al (2010)
and applications. Humana Press, Totowa, NJ Fluorescent proteins and their applications in
16. Wickiser JK, Winkler WC, Breaker RR et al imaging living cells and tissues. Physiol Rev
(2005) The speed of RNA transcription and 90:1103–1163
Chapter 6
Abstract
Artificial riboswitches are engineered to regulate gene expression in response to a variety of non-endogenous
small molecules and, therefore, can be useful tools to reprogram cellular behavior for different applica-
tions. A new synthetic riboswitch can be created by linking an in vitro-selected aptamer with a randomized
expression platform followed by in vivo selection and screening. Here, we describe an in vivo selection and
screening technique to discover artificial riboswitches in E. coli cells that is based on TEV protease–FRET
substrate reporter system.
Key words Aptamer, Riboswitch, In vivo selection, TEV protease, FRET-based fusion protein
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_6, © Springer Science+Business Media New York 2014
77
78 Svetlana V. Harbaugh et al.
Fig. 1 TEV protease–FRET substrate reporter system for monitoring riboswitch performance in E. coli cells.
In the absence of ligand, extensive pairing of riboswitch with ribosome-binding site (RBS) prevents translation
of TEV protease. Ligand binding induces conformational change of the riboswitch, unpairing RBS and initiating
translation of the TEV protease mRNA. The produced protease cleaves the FRET-based protein substrate, yield-
ing an increase in fluorescent emission at 510 nm upon excitation at 457 nm
2 Materials
3 Methods
(continued)
Water To 10 μL final
Temperature 25 ºC
Time 3h
Fig. 5 In vivo screening for functional riboswitches. (a) Obtained after transformation E. coli colonies are picked
and streaked on three LB-agar plates: without inducer and ligand; with inducer (expression of the fusion
protein-encoding gene) and without ligand (no riboswitch activation); with inducer (expression of the
fusion protein-encoding gene) and with ligand (riboswitch activation and TEV protease production). (b) Colonies
that show increased fluorescence in the presence of ligand are selected for screening in liquid medium
(an example is circled ). (c) The gene expression of the selected riboswitch clones is quantified in the presence
and absence of ligand
10. The following day, inoculate four 24-well plates for each plate
with overnight cultures with 20 μL of overnight culture. Each
well of 24-well plates should contain 1 mL of LB supplemented
with appropriate antibiotics.
11. Incubate the 24-well plates with new cultures for 2.5 h at
37 ºC with shaking (215 rpm) until appropriate absorbance
at 600 nm is reached (0.3–0.5 when corrected to 1-cm path
length cuvette). Add an appropriate amount of an inducer for
expression of eGFP-TL-REACh protein gene. (In our work,
gene encoding eGFP-TL-REACh protein was cloned in
pHWG640 plasmid vector under control of Rhamnose-
inducible promoter. For induction of the expression, we have
used 0.4 % Rhamnose.)
12. Incubate cultures for 30 min at 37 ºC with shaking (215 rpm).
Then, divide each group of four plates with the same cultures
on two sets of two. Add ligand to the first set of plates (this set
will contain cells with activated riboswitches). Add equivalent
volume of the solvent where ligand is dissolved to the second
set of plates (this set will contain cells with non-activated
riboswitch) (Fig. 5c). Measure the absorbance at 600 nm and
fluorescence at 510 nm (excitation 457 nm) of the cell cultures
in each plate.
13. Incubate the 24-well plates for 6 h at 37 ºC with shaking
(215 rpm). Measure the absorbance at 600 nm and fluores-
cence at 510 nm (excitation 457 nm) of the cell cultures in
each plate. To normalize the data, divide fluorescence intensity
by absorbance for each well.
14. Compare the ratios of the normalized data for cultures
grown in the presence of ligand to those grown in the absence
of ligand (the “activation ratio”) to identify functional
riboswitches.
to the second set of flasks (this set will contain cells with
non-activated riboswitch). Measure the absorbance at 600 nm
and fluorescence at 510 nm (excitation 457 nm) of the cell
cultures in each flask (it will be time 0 h). Record the absor-
bance at 600 nm and fluorescence at 510 nm (excitation
457 nm) for each cell culture at time 1, 2, 3, 4, 5, and 6 h after
riboswitch activation (ligand addition). At each time point,
collect 10 mL of each culture for preparation of cellular lysates.
Harvest the cells by centrifugation at 3,200 rpm for 10 min at
4 ºC. Discard the supernatant. Store cell pellets at −80 ºC until
ready for processing.
5. For preparation of cellular lysates, thaw cell pellets on ice for
approximately 15–20 min, vortex, and resuspend in 0.4 mL of
lysis buffer. Incubate cell suspensions on ice for 30 min and
mix two to three times by gently swirling. Remove cell debris
by centrifugation at 13,200 rpm for 30 min at 4 ºC. Transfer
supernatant from each sample into new tubes, and measure
fluorescence at 510 nm (excitation 457 nm). Determine pro-
tein concentrations of the clarified cellular lysates using BCA
assay; follow the protocol provided by the manufacturer. To
normalize the data, divide fluorescence intensity by total pro-
tein concentration for each sample.
6. Compare the ratios of the normalized data for lysates from
cultures grown in the presence of ligand to those grown in the
absence of ligand (the “activation ratio”) to determine the
riboswitch performance at different time points (time course).
4 Notes
Acknowledgement
References
1. Jin Y, Watt RM, Danchin A, Huang JD (2009) dual genetic selection. J Am Chem Soc 130:
Use of a riboswitch-controlled conditional 16310–16315
hypomorphic mutation to uncover a role for 7. Tuerk C, Gold L (1990) Systematic evolution
the essential csrA gene in bacterial autoaggre- of ligands by exponential enrichment: RNA
gation. J Biol Chem 284:28738–28745 ligands to bacteriophage T4 DNA polymerase.
2. Lee ER, Blount KF, Breaker RR (2009) Science 249:505–510
Roseoflavin is a natural antibacterial compound 8. Roth A, Breaker RR (2004) Selection in vitro
that binds to FMN riboswitches and regulates of allosteric ribozymes. In: Sioud M (ed)
gene expression. RNA Biol 6:187–194 Ribozymes and siRNA protocols, methods in
3. Kim JN, Blount KF, Lim J, Link KH, Breaker R molecular biology, 2nd edn. Humana Press,
(2009) Design and antimicrobial action of Totowa, NJ, pp 145–164
purine analogues that bind guanine riboswitches. 9. Harbaugh S, Kelley-Loughnane N, Davidson
ACS Chem Biol 4:915–927 M, Narayanan L, Trott S, Chushak YG, Stone
4. Jise AM, Soukup GA, Breaker RR (2001) MO (2009) FRET-based optical assay for moni-
Cooperative binding of effectors by an toring riboswitch activation. Biomacromolecules
allosteric ribozyme. Nucleic Acids Res 29: 10:1055–1060
1631–1637 10. Van der Berg S, Löfdahl P-A, Härd T, Berglund
5. Win MN, Smolke CD (2008) Higher-order H (2006) Improved solubility of TEV protease by
cellular information processing with synthetic directed evolution. J Biotechnol 121:291–298
RNA devices. Science 322:456–460 11. Ganesan S, Ameer-beg SM, Ng TT, Vojnovic
6. Sharma V, Nomura Y, Yokobayashi Y (2008) B, Wouters FS (2006) A dark yellow fluorescent
Engineering complex riboswitch regulation by protein (YFP)-based resonance energy-accepting
FRET-Based Riboswitch Selection 91
Abstract
Riboswitches are RNA-based regulatory devices that mediate ligand-dependent control of gene expression.
However, there has been limited success in rationally designing riboswitches. Moreover, most previous
riboswitches are confined to a particular gene and only perform one-way regulation. Here, we describe a
library screening strategy for efficient creation of ON riboswitches of lacI of Escherichia coli. An ON ribo-
switch of lacI is then integrated with the lac promoter, generating a hybrid device to achieve portable
sequential OFF-and-ON gene regulation.
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_7, © Springer Science+Business Media New York 2014
93
94 Ye Jin and Jian-Dong Huang
2 Materials
All media and buffers are prepared using sterilized distilled deion-
ized water (ddH2O). All containers and pipette tips are sterilized
by heating (121 °C, 15 min) and stored at room temperature.
2.1 Incubation 1. Bacterial strain: E. coli strain MG1655 (F- lambda- ilvG- rfb-
of Bacteria 50 rph-1).
2. Growth medium: Luria-Bertani (LB) medium supplemented
with 2 mM magnesium sulfate. The antibiotics 50 μg/ml
ampicillin, 50 μg/ml kanamycin, and 12.5 μg/ml chloram-
phenicol are used for selection when appropriate.
3. Glycerol.
4. Temperature-controlled microplate shaker.
5. Temperature-controlled flask shaker.
6. Sterilized 96-well microtiter plate.
7. Spectrophotometer and cuvettes.
8. Flasks (500 ml).
9. Refrigerators at 4 °C and freezers at −20 and −80 °C.
Portable Two-Way Riboswitches 95
3 Methods
3.1 Construction 1. A cat fragment is PCR amplified using pCm as a template. The
of a Randomized resulting DNA product carries the cat gene to confer chloram-
Riboswitch Cassette phenicol resistance. Upstream of cat is a constitutive promoter
(Fig. 1) to drive expression of the chloramphenicol resistance gene.
The cat gene is followed by a 3′ untranslated region, which
serves as a linker between the cat gene and the downstream
theophylline-responsive aptamer (see Note 3).
2. The theophylline-responsive aptamer is synthesized by annealing
complementary primers. Sequence of the theophylline aptamer is
GAAATACCAGCATCGTCTTGATGCCCTTGGCAGTTC
(5′–3′). Design a forward primer GAAATACCAGCATCGT
CTTGATGCCCTTGGCAGTTC (5′–3′) and a reverse primer
GAACTGCCAAGGGCATCAAGACGATGCTGGTATTTC
(5′–3′) which is the reverse complement of the forward primers.
The two primers are denatured at 94 °C for 30 s and then annealed
Portable Two-Way Riboswitches 97
Fig. 1 Construction of a theophylline-aptamer cassette. NNNNN denotes a 5-nt random sequence which is
added to the theophylline aptamer by PCR. The loxp-cat-loxp cassette is linked to the theophylline aptamer
followed by the 5-nt random sequence by fusion PCR. The resulting cassette is flanked with arms (45 nt)
homologous to the regions surrounding the ribosome-binding site of the lacI gene
Fig. 2 Creation of ON riboswitches for the lacI gene using a library screening-
based method. A random cassette mixture containing a theophylline-responsive
aptamer and five random bases is “recombineered” into the chromosome of
MG1655, generating a riboswitch library for lacI. Cells are plated on X-gal plates
without theophylline but with 2 mM caffeine. Relatively blue colonies are searched
for and transferred to X-gal plates with 2 mM theophylline. Those that turn into
white are considered to carry ON riboswitches of lacI. The blue arrow denotes
activating effects; the blunt red arrow denotes repressing effects. Reproduced
with permission from Oxford University Press [12] (color figure online)
Fig. 3 Predicted structures of an ON riboswitch of lacI. In the non-ligand-bound state, there is a 9 bp stem
structure immediately upstream of the ribosome-binding site and is likely to block the gene translation. In the
theophylline-bound state, the long stem does not form and consequently permits the gene translation. The
random sequence is shown in red; the ribosome-binding site in blue; and the start codon in green. Reproduced
with permission from Oxford University Press [12] (color figure online)
Fig. 4 Gene control mediated by the hybrid riboswitch device. (a) Constitution of the riboswitch hybrid device.
The boxed is a portable riboswitch cassette ON-lacI-LacIbs (LacI-binding sites) integrated immediately
upstream of ribosome-binding site of the rpoS gene that is a master regulator of acid resistance (AR).
(b) Tunable and two-way control of rpoS via the ON-lacI-LacIbs hybrid device, as revealed by acid survival. The
strain ON-lacI-rpoS that carries the ON-lacI-LacIbs device upstream of the ribosome-binding site becomes
increasingly sensitive to acid with theophylline (0–1,600 μM). When increasing concentrations of IPTG are added
to cells grown in media with 1,600 μM theophylline, acid survival is increased and restored to wild-type levels
in the presence of 40 μM IPTG. Error bars, SD. Reproduced with permission from Oxford University Press [12]
and inoculate the bacteria that are not treated with acid. After
overnight culture, determine colony-forming units (CFU) of
acid-treated and untreated cells. Calculate acid survival (%)
with the following formula: Acid survival (%) = 100 × (CFU of
treated/CFU of untreated).
4. We have observed that the rpoS expression is ON in the absence
of theophylline and becomes OFF in the presence of theophyl-
line and that adding IPTG restores the rpoS expression of
bacteria grown in LB plus theophylline (Fig. 4b). These confirm
the portability and two-way control features of the ON-lacI-
LacIbs device (see Note 20).
5. Unlike rpoS, expression of many genes is not linked to a
quantifiable phenotype. In those cases, expression control by
the hybrid device can be evaluated by constructing lacZ trans-
lational fusions for target genes on the chromosome and then
measuring beta-galactosidase assay.
6. To construct a chromosomal lacZ translational fusion, insert
the loxP-cat-loxP selectable cassette immediately after the stop
codon of the lacZ gene on the MG1655 chromosome using
recombineering (as described above). Next, PCR amplify the
lacZ-loxP-cat-loxP cassette, and insert it (in frame) immedi-
ately prior to the stop codon of the target gene on the chromo-
some. The inserted lacZ fragment starts from the eighth codon
of lacZ gene and is co-transcribed and translated with the fused
genes. Quantify the expression of gene-lacZ fusions using a
beta-galactosidase assay kit (see Note 21).
4 Notes
References
1. Ellington AD, Szostak JW (1990) In vitro with SPR imaging. Nucleic Acids Res 34:
selection of RNA molecules that bind specific 6416–6424
ligands. Nature 346:818–822 6. Lynch SA, Desai SK, Sajja HK et al (2007)
2. Tuerk C, Gold L (1990) Systematic evolution A high-throughput screen for synthetic ribo-
of ligands by exponential enrichment: RNA switches reveals mechanistic insights into their
ligands to bacteriophage T4 DNA polymerase. function. Chem Biol 14:173–184
Science 249:505–510 7. Lynch SA, Gallivan JP (2009) A flow cytometry-
3. Chushak Y, Stone MO (2009) In silico selec- based screen for synthetic riboswitches. Nucleic
tion of RNA aptamers. Nucleic Acids Res Acids Res 37:184–192
37:e87 8. Muranaka N, Abe K, Yokobayashi Y (2009)
4. Collett JR, Cho EJ, Lee JF et al (2005) Mechanism-guided library design and dual
Functional RNA microarrays for high- genetic selection of synthetic OFF riboswitches.
throughput screening of antiprotein aptamers. Chembiochem 10:2375–2381
Anal Biochem 338:113–123 9. Suess B, Fink B, Berens C et al (2004) A the-
5. Li Y, Lee HJ, Corn RM (2006) Fabrication and ophylline responsive riboswitch based on helix
characterization of RNA aptamer microarrays slipping controls gene expression in vivo.
for the study of protein-aptamer interactions Nucleic Acids Res 32:1610–1614
Portable Two-Way Riboswitches 105
10. Jin Y, Watt RM, Danchin A et al (2009) Use of a unit as a tool to study U6 function. RNA 4:
riboswitch-controlled conditional hypomorphic 231–238
mutation to uncover a role for the essential csrA 16. Grilly C, Stricker J, Pang WL et al (2007)
gene in bacterial autoaggregation. J Biol Chem A synthetic gene network for tuning protein
284:28738–28745 degradation in Saccharomyces cerevisiae. Mol
11. Win MN, Smolke CD (2007) A modular and Syst Biol 3:127
extensible RNA-based gene-regulatory platform 17. Cronin CA, Gluba W, Scrable H (2001) The
for engineering cellular function. Proc Natl lac operator-repressor system is functional in
Acad Sci U S A 104:14283–14288 the mouse. Genes Dev 15:1506–1517
12. Jin Y, Huang JD (2011) Engineering a porta- 18. Ellis T, Wang X, Collins JJ (2009) Diversity-
ble riboswitch-LacP hybrid device for two-way based, model-guided construction of synthetic
gene regulation. Nucleic Acids Res 39:e131 gene networks with predicted functions. Nat
13. Reuter JS, Mathews DH (2010) RNAstructure: Biotechnol 27:465–471
software for RNA secondary structure predic- 19. Small P, Blankenhorn D, Welty D et al (1994)
tion and analysis. BMC Bioinformatics 11:129 Acid and base resistance in Escherichia coli and
14. Studier FW, Moffatt BA (1986) Use of bacte- Shigella flexneri: role of rpoS and growth pH.
riophage T7 RNA polymerase to direct selective J Bacteriol 176:1729–1737
high-level expression of cloned genes. J Mol 20. Fang FC, Libby SJ, Buchmeier NA et al (1992)
Biol 189:113–130 The alternative sigma factor katF (rpoS) regu-
15. Luukkonen BG, Seraphin B (1998) Construction lates Salmonella virulence. Proc Natl Acad Sci
of an in vivo-regulated U6 snRNA transcription U S A 89:11978–11982
Chapter 8
Abstract
Riboswitches are naturally occurring RNA-based genetic switches that control gene expression in
response to the binding of small-molecule ligands, typically through modulation of transcription or
translation. Their simple mechanism of action and the expanding diversity of riboswitch classes make
them attractive targets for the development of novel gene expression tools. The essential first step in
realizing this potential is to generate artificial riboswitches that respond to nonnatural, synthetic ligands,
thereby avoiding disruption of normal cellular function. Here we describe a strategy for engineering
orthogonally selective riboswitches based on natural switches. The approach begins with saturation
mutagenesis of the ligand-binding pocket of a naturally occurring riboswitch to generate a library of
riboswitch mutants. These mutants are then screened in vivo against a synthetic compound library to
identify functional riboswitch–ligand combinations. Promising riboswitch–ligand pairs are then further
characterized both in vivo and in vitro. Using this method, a series of artificial riboswitches can be gen-
erated that are versatile synthetic biology tools for use in protein production, gene functional analysis,
metabolic engineering, and other biotechnological applications.
Key words Orthogonal riboswitches, Artificial riboswitches, Aptamer domain, Synthetic ligands,
Gene expression tools, In vivo screening, In vitro transcription, Isothermal titration calorimetry,
Synthetic biology
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_8, © Springer Science+Business Media New York 2014
107
108 Helen A. Vincent et al.
recently our laboratory has pioneered a novel strategy for the pro-
duction of artificial orthogonal riboswitches, which involves
reengineering the aptamer domain of natural riboswitches so that
they respond to synthetic ligands rather than the native metabo-
lites [10]. Applications such as the production of designer auxo-
trophs [11], bioremediation [12], development of gene expression
tools for pathogenic organisms [13, 14], and simultaneous control
of multiple genes within the same bacterial cell [15] are beginning
to be explored.
The major advantage of in vitro generation of aptamer
domains is that SELEX can, in theory, be used to produce
aptamers with high affinity and selectivity against any ligand of
choice. However, in practice, only a limited number of aptam-
ers have been applied to the development of riboswitch tech-
nologies in bacteria: the theophylline aptamer [5–9, 11, 13,
14, 16, 17] and, more recently, an atrazine aptamer [12]. This
is largely because the de novo development of aptamers in vitro
can be both costly and time consuming and, more critically,
does not guarantee in vivo functionality [18]. Functionality is
fundamentally dependent on the bioavailability of the ligand.
Even the validated synthetic ligands theophylline and atrazine
exhibit poor cell permeability, such that far higher concentra-
tions of ligand are required to elicit a response in vivo than
would be predicted from binding affinities determined in vitro
[11, 12, 18]. Also, when challenged with the intracellular
metabolome, the selectivity of the aptamer domain for the syn-
thetic ligand is often weaker than observed in vitro [19].
Finally, in vivo functionality is strongly dependent upon how
the aptamer and expression platform are linked, and this con-
nection is not easy to rationally design. Consequently, the
majority of functional in vitro-engineered artificial riboswitches
have only been isolated following further extensive in vivo
genetic selections or screens [5–9, 12].
To address the limited diversity of functional synthetic aptam-
ers, our laboratory has developed a novel strategy for reengineering
the aptamer domains of natural riboswitches to produce artificial
riboswitches that respond to diverse synthetic ligands [10]. We have
demonstrated the application of this approach with the adenine-
responsive add translational-ON switch from Vibrio vulnificus, pro-
ducing an orthogonally selective artificial riboswitch for inducible
gene expression in E. coli [10]. In this chapter, we present the pro-
tocol that we use to change the selectivity of translational ribo-
switches for use as gene expression tools in bacteria [10]. An
overview of this protocol is presented in Fig. 1. In principle, this
strategy is applicable to any riboswitch, for use in any bacterial
organism. However, for simplicity, the method presented here
focuses on E. coli, a common model organism in the laboratory.
110 Helen A. Vincent et al.
A/C/G/U A/C/G/U
R CAT
A/C/G/U A/C/G/U
2. Site-saturation mutagenesis of
ligand binding pocket to generate
library of riboswitch mutants
- Chloramphenicol + Chloramphenicol
H NH2
S N N O
N
N
HN N
H N N O2N
2
O 4. Screen riboswitch mutants against
O H O
compound library in vivo
NH2
H2N N O HO N O
N N
N N N
O N NH
1. Select natural riboswitch H 2
O O O
for reengineering
3. Build synthetic compound
library
Synthetic ligand
R eGFP
Gene Expression
5. In vivo characterization of
functional mutant riboswitch-
6. In vitro characterization of synthetic ligand pairs (dose-
functional mutant riboswitch- dependence and Natural ligand
synthetic ligand pairs orthogonality)
(binding and structure) [Ligand]
Fig. 1 Schematic overview of the strategy to engineer orthogonally selective artificial riboswitches. Individual
steps are explained in detail in the subsequent protocol
2 Materials
3 Methods
The following protocols assume that the user has a good working
knowledge of standard molecular cloning techniques. All in vitro
procedures (see Subheading 3.5 onwards) should be carried out in
an RNase-free environment (see Note 9), including the use of ster-
ile reagents and consumables. Gloves should be changed frequently
to avoid contamination from ribonucleases found on the hands
and in the environment. RNA samples should be kept on ice wher-
ever possible.
3.1 Construction of The X-ray crystal structures of aptamer domains in complex with
Mutant Riboswitch their cognate ligand have been determined for most classes of ribo-
Chloramphenicol- switch. In general, aptamer domains form tight binding pockets
Based Reporter that completely surround the ligand. With the notable exception
Plasmids of adenosylcobalamin riboswitches [20, 21], the majority of ribo-
switches appear to selectively bind the ligand through specific
hydrogen bonding interactions mediated by conserved nucleotides
within the ligand-binding pocket, with the binding of planar
ligands further stabilized through base-stacking interactions [2,
22]. When changing the specificity of natural riboswitch aptamers,
a good initial strategy is to specifically target the conserved nucleo-
tides responsible for forming the aptamer–ligand hydrogen bonds.
1. Select a suitable riboswitch for reengineering (see Note 10).
2. Identify key ligand-binding nucleotides from the X-ray crystal
structure of the aptamer–ligand complex, and select nucleo-
tides for mutagenesis (see Note 11).
3. Using established methods, construct a parental chloram-
phenicol reporter plasmid (Fig. 2a) by cloning the following
elements, in order, into a suitable high-copy-number vector
(see Note 1).
(a) The IPTG-inducible lac promoter and operator derived
from the 5′ UTR of the lacZ gene from E. coli (see Note 2).
(b) The natural riboswitch aptamer domain and expression
platform up to the native translational start site.
(c) The open reading frame of the CAT gene, which confers
chloramphenicol resistance (see Note 3).
116 Helen A. Vincent et al.
Chloramphenicol GFP
reporter plasmid reporter plasmid
Ampicillin Ampicillin
resistance gene resistance gene
Fig. 2 Schematic representation of the reporter plasmids. (a) Chloramphenicol reporter plasmid for in vivo
screening. (b) GFP reporter plasmid for quantitative in vivo characterization
3.2 Building a The choice of the screening library size and composition directly
Screening Library affects the probability of successfully identifying functional mutant
of Nonnatural riboswitch–synthetic ligand combinations. In practice, we have found
Compounds that a pool of 15 riboswitch mutants (based upon saturation muta-
genesis of two nucleotides in the ligand-binding pocket), screened
against a carefully selected library of no more than 100 compounds
(1,500 different riboswitch–ligand scenarios in total), should be suf-
ficient to find functional mutant–ligand pairs. When designing the
library it is important to consider that in order to be fit for purpose,
the compounds should have suitable physicochemical properties
(e.g., stable, at least modestly soluble in water, cell permeable) and
they should be accessible in terms of availability and cost.
1. Using the chemical structure of the natural ligand as the query,
use structural chemical research tools (e.g., CAS SciFinder)
and search commercial databases (e.g., Aurora Fine Chemicals,
TimTec, Enamine, Bionet) using chemical database software
(e.g., ChemFinder) to generate a list of analogues.
2. Filter out any compounds that are natural metabolites, since
these compounds would be expected to encounter similar
Orthogonally Selective Bacterial Riboswitches 117
1 2 3 4 1 2 3 4
- Chloramphenicol
5 6 7 8 5 6 7 8
9 10 11 12 9 10 11 12
1 2 3 4 1 2 3 4
+ Chloramphenicol
5 6 7 8 5 6 7 8
9 10 11 12 9 10 11 12
Fig. 3 Schematic representation of the expected results of chloramphenicol screening. Shaded circles indicate
bacterial growth, and white circles indicate growth inhibition. (a) “ON” riboswitches. Wells 1 and 2 represent
the no ligand and natural ligand controls and should grow only in the absence of chloramphenicol. Wells 3–N
represent different synthetic ligands. Functional riboswitch–ligand pairs are identified by growth in both the
absence and presence of chloramphenicol, e.g., wells 3 and 6. (b) “OFF” riboswitches. Wells 1 and 2 represent
the no ligand and natural ligand controls and should grow in both the absence and presence of chlorampheni-
col. Wells 3–N represent different synthetic ligands. Functional riboswitch–ligand pairs are identified by growth
only in the absence of chloramphenicol, e.g., wells 5 and 11 (color figure online)
8. To all other wells in the first screen add 180 μl of the induced
culture without chloramphenicol.
9. To all other wells in the second screen add 180 μl of the
induced culture supplemented with chloramphenicol.
10. Incubate at 37 °C for 4 h with shaking.
11. Measure the absorbance at 620 nm of each well using a plate
reader, and subtract the reading from the media-only well
from all other wells to assess the extent of bacterial growth.
No cells
Natural Uninduced
cells
Ligand
Induced
cells
No cells
Synthetic Uninduced
Ligand cells
Induced
cells
Fig. 4 Example plate layout for quantitative in vivo characterization. Testing different ligand concentrations
allows the dose-dependent response to be assessed. Comparison of the natural ligand to synthetic ligands
confirms the selectivity of the synthetic ligand
a ‘ON’ Switch
Synthetic ligand
Gene Expression
Gene Expression
Synthetic ligand
Time [Ligand]
b ‘OFF’ Switch
Natural ligand
Natural ligand
Gene Expression
Gene Expression
Synthetic ligand
Synthetic ligand
Time [Ligand]
Fig. 5 Schematic representation of expected results from quantitative in vivo characterization. Left panels
show gene expression plotted against time with different ligand concentrations plotted as individual series.
Right panels show gene expression plotted against ligand concentration for a single time point. (a) “ON” ribo-
switches. Gene expression increases with increasing time and increasing ligand concentration for the syn-
thetic ligand (dose-dependence). Gene expression is not affected by increasing time or increasing ligand
concentration for the natural ligand (orthogonality). (b) “OFF” riboswitches. Gene expression decreases with
increasing time and increasing ligand concentration (dose-dependence). Gene expression is not affected by
increasing time or increasing ligand concentration for the natural ligand (orthogonality)
122 Helen A. Vincent et al.
(f) Add 2.5 volumes (2.775 ml) of chilled 100 % ethanol and
incubate at −20 °C overnight.
(g) Pellet the DNA by centrifugation at 20,000 × g for 20 min
at 4 °C.
(h) Decant the ethanol, and wash the pellet with 2 ml chilled
70 % ethanol.
(i) Decant the ethanol, and allow the pellet to air-dry.
(j) Resuspend in 200 µl of nuclease-free H2O, and determine
the concentration by measuring the absorbance at
260 nm.
3.6 Preparation of 1. Prepare the following 5 ml scale (see Note 19) in vitro tran-
Aptamer RNA by In scription reaction:
Vitro Transcription for (a) 500 μl of 10× transcription buffer.
In Vitro Biophysical
(b) 1.2 ml NTP mix.
Characterization
(c) 50 μl of 1 M DTT.
(d) 140 μl of 1 M MgCl2.
(e) 50–100 μg/ml (final concentration) double-stranded
DNA template (see Subheading 3.5).
(f) 0.25 mg/ml (final concentration) T7 RNA polymerase.
(g) Nuclease-free H2O to 5 ml.
2. Incubate at 37 °C for 6–8 h (see Note 21).
3. Add 500 μl of 0.5 M EDTA (pH 8.0) and incubate for a fur-
ther 30 min at 37 °C.
4. Using a syringe fitted with a needle, transfer the reaction to a
Slide-A-Lyzer dialysis cassette (3,000 MWCO, 12 ml capac-
ity), and dialyze overnight at 4 °C in 2 l of freshly prepared
riboswitch buffer.
5. Using a syringe fitted with a needle, transfer the dialyzed RNA
to a Vivaspin 6 centrifugal concentrator (3,000 MWCO) and
concentrate to a final volume of ~2 ml by centrifugation at
4,000 × g at 4 °C.
6. Add 200 μl (400 units) of DNase I and incubate for 30 min at
37 °C.
7. Store at 4 °C.
3.7 Purification of We purify our RNA transcripts (from T7 RNA polymerase, DNase
Aptamer RNA for In I, and prematurely terminated transcripts) by size-exclusion chro-
Vitro Biophysical matography. This intentionally avoids denaturation or
Characterization precipitation of the RNA which could trap the RNA in inactive,
nonnative conformations [23]. The following purification proto-
col is designed to produce RNA of suitable quality and quantity
for carrying out ITC and X-ray crystallography and assumes that
124 Helen A. Vincent et al.
1. Before the first use, clean the sample cell as follows (see
Note 24):
(a) Fill the sample cell with 12 % NaOH and incubate at 65 °C
for 1 h.
(b) Wash the sample cell with DEPC-treated dH2O.
(c) Fill the sample cell with 5 % RNaseZap and incubate at
25 °C for 1 h.
(d) Wash the sample cell with DEPC-treated dH2O.
(e) Wash the sample cell thoroughly with dialysis buffer.
2. Before the first use, clean the titrant syringe and the Hamilton
syringe used to load the sample cell as follows:
(a) Fill with 5 % RNaseZap, and leave for 1 h.
(b) Wash with DEPC-treated dH2O.
(c) Wash with dialysis buffer.
3. Degas the aptamer RNA and ligand solutions using a degas-
sing unit.
4. Add 2 ml of dialysis buffer to the sample cell using a Hamilton
syringe (the blank).
5. Purge the titrant syringe twice and fill with 100 μM (see Note
25) ligand solution using a syringe fitted with tubing.
6. Dry the titrant syringe needle gently using a clean KimWipe, tak-
ing care not to touch the outlet pore at the base of the needle,
and carefully lower into the sample cell until it clicks into place.
7. Set the cell temperature to 25 °C, reference power to 5 μcal/s,
and stirrer speed to 310 rpm.
8. After a 60-s pre-titration delay, inject 2 μl of ligand over 4.8 s
followed by 24 × 12 μl injections of ligand, each over 28.8 s
with a 300-s delay between injections (ligand concentration
range of ~0.5 to ~12.5 μM when starting from a 100 μM
ligand stock solution).
9. Thoroughly wash the sample cell with dialysis buffer and then
load the sample cell with 2 ml of 10 µM (see Note 26) aptamer
RNA in dialysis buffer using a Hamilton syringe (the
experiment).
10. Carry out the same injection protocol as for ligand into buffer
(see step 8).
11. Fit the data using Origin software (see Note 27). A good data
set will have a clear sigmoidal curve (with c = 20–100) and an
appropriate stoichiometry (n = 1 for most riboswitch aptamer–
ligand interactions), and the first few injections should cover
at least 10 μcal each, with an average of 5 μcal per injection
over the course of the experiment (see Note 28).
126 Helen A. Vincent et al.
4 Notes
1. The chosen vector should not require the CAT gene for plas-
mid maintenance.
2. We use the inducible lac promoter/operator to provide tighter
gene expression control when developing novel switches, but
alternative inducible or constitutive promoters could be
substituted.
3. The CAT gene could be substituted for alternative reporter
genes, e.g., tetA (allowing for selection or screening with tet-
racycline and/or NiCl2 [5, 28–30]), lacZ (allowing enzymatic
screening for β-galactosidase activity [6, 16]), GFP (allowing
for flow cytometry-based screening for fluorescence [8, 9]), or
cheZ (allowing for screening for motility [7, 12]).
4. CAS SciFinder and ChemFinder require a site licence.
5. The F′ episome carries the lacIq gene required for inducible
expression from the lac promoter.
6. Residual DEPC can inhibit the in vitro transcription reaction.
7. We purify T7 RNA polymerase in-house from an E. coli expres-
sion strain [31], since the quantities required for the
preparative-scale (5–20 ml) in vitro transcription reactions
that we routinely carry out can become costly if commercial
sources are used.
8. In our laboratory we keep a dedicated column for RNA
purification.
9. In our laboratory we have set aside an RNA bench space,
which includes a Labcare PCR cabinet and a dedicated set of
pipettes. Apparatus is frequently treated with RNaseZap
(Invitrogen) and washed with copious amounts of DEPC-
treated dH2O.
10. Riboswitch aptamer domains that bind small, uncharged, pla-
nar ligands with four or fewer conserved ligand-interacting
nucleotides are most suitable for mutagenesis. Riboswitches
that respond to more complex coenzymes (e.g., adenosylco-
balamin and flavin mononucleotide) are not suitable as alter-
native synthetic ligands are unavailable due to synthetic
inaccessibility.
11. In the case of riboswitches for which there is no X-ray crystal
structure available, in vitro mutagenesis analysis and/or
structural probing methods (e.g., in-line probing and enzy-
matic footprinting) could provide information about ligand-
binding nucleotides.
12. This approach has been validated and is feasible for small
ligands (e.g., purines), where initially up to two ligand-binding
Orthogonally Selective Bacterial Riboswitches 127
23. ITC has the advantage over many other in vitro methods in
that the interaction is monitored directly, in solution, without
immobilization or modification of either binding partner, and
it provides a complete biophysical characterization of the
interaction including binding affinity, binding stoichiometry,
and thermodynamic parameters (enthalpy change (ΔHobs),
entropy change (ΔS), and Gibbs free energy change (ΔG)).
However, it does require significant quantities of both aptamer
RNA and ligand (10–50 nmol in the sample cell, 2.5–250 nmol
in the titrant syringe) for each experimental run. Other bio-
chemical techniques can also be used, e.g., in-line probing
[33, 34], equilibrium dialysis [35], and selective 2′-hydroxyl
acylation and primer extension (SHAPE) [34].
24. Samples can become adsorbed onto the surface of the sample
cell, interfering with heat transfer and causing erratic, noisy
baselines.
25. The concentration of the ligand in the titrant syringe should
be 10–20 times the concentration of the aptamer RNA in the
sample cell.
26. To determine the lower and upper limits for the concentration
of the aptamer in the sample cell (Mcell ), we use the equation
c = (n ´ Mcell ) / K d
where c describes the sigmoidicity of the binding curve
(should be between 20 and 100 [36]), n is the stoichiometry
of the interaction (1 for most riboswitch aptamer–ligand inter-
actions), and Kd is the expected dissociation constant. If the
expected Kd is unknown, 10 μM aptamer is a reasonable con-
centration for pilot studies.
27. The baseline and integration limits for each injection should be
checked manually before attempting to fit the data since noise
can perturb the automatic fitting performed by the software.
28. The aptamer/ligand concentrations or injection settings can
be adjusted in subsequent experiments until these criteria are
satisfied.
References
1. Breaker RR (2011) Prospects for riboswitch ecule target. Curr Protoc Nucleic Acid Chem
discovery and analysis. Mol Cell 43:867–879 Chapter 9:Unit 9.5.1–5.23
2. Serganov A, Nudler E (2012) A decade of 5. Sharma V, Nomura Y, Yokobayashi Y (2008)
riboswitches. Cell 152:17–24 Engineering complex riboswitch regulation by
3. Barrick JE, Breaker RR (2007) The distribu- dual genetic selection. J Am Chem Soc
tions, mechanisms, and structures of 130:16310–16315
metabolite-binding riboswitches. Genome Biol 6. Lynch SA, Desai SK, Sajja HK et al (2007) A
8:R239 high-throughput screen for synthetic ribo-
4. Codrea V, Hayner M, Hall B et al (2010) In switches reveals mechanistic insights into their
vitro selection of RNA aptamers to a small mol- function. Chem Biol 14:173–184
Orthogonally Selective Bacterial Riboswitches 129
7. Topp S, Gallivan JP (2008) Random walks to syn- 22. Serganov A, Patel DJ (2012) Molecular recog-
thetic riboswitches-a high-throughput selection nition and function of riboswitches. Curr Opin
based on cell motility. Chembiochem 9:210–213 Struct Biol 22:279–286
8. Fowler CC, Brown ED, Li Y (2008) A FACS- 23. Uhlenbeck OC (1995) Keeping RNA happy.
based approach to engineering artificial ribo- RNA 1:4–6
switches. Chembiochem 9:1906–1911 24. Gilbert SD, Batey RT (2009) Monitoring RNA-
9. Lynch SA, Gallivan JP (2009) A flow ligand interactions using isothermal titration
cytometry-based screen for synthetic ribo- calorimetry. Methods Mol Biol 540:97–114
switches. Nucleic Acids Res 37:184–192 25. Salim NN, Feig AL (2009) Isothermal titration
10. Dixon N, Duncan JN, Geerlings T et al (2010) calorimetry of RNA. Methods 47:198–205
Reengineering orthogonally selective riboswitches. 26. Pikovskaya O, Serganov AA, Polonskaia A et al
Proc Natl Acad Sci U S A 107:2830–2835 (2009) Preparation and crystallization of
11. Desai SK, Gallivan JP (2004) Genetic screens riboswitch-ligand complexes. Methods Mol
and selections for small molecules based on a Biol 540:115–128
synthetic riboswitch that activates protein trans- 27. Edwards AL, Garst AD, Batey RT (2009)
lation. J Am Chem Soc 126:13247–13254 Determining structures of RNA aptamers and
12. Sinha J, Reyes SJ, Gallivan JP (2010) riboswitches by X-ray crystallography. Methods
Reprogramming bacteria to seek and destroy Mol Biol 535:135–163
an herbicide. Nat Chem Biol 6:464–470 28. Nomura Y, Yokobayashi Y (2007) Reengineering
13. Topp S, Reynoso CMK, Seeliger JC et al a natural riboswitch by dual genetic selection. J
(2010) Synthetic riboswitches that induce gene Am Chem Soc 129:13814–13815
expression in diverse bacterial species. Appl 29. Muranaka N, Sharma V, Nomura Y et al (2009)
Environ Microbiol 76:7881–7884 An efficient platform for genetic selection and
14. Reynoso CMK, Miller MA, Bina JE et al screening of gene switches in Escherichia coli.
(2012) Riboswitches for intracellular study of Nucleic Acids Res 37:e39
genes involved in Francisella pathogenesis. 30. Muranaka N, Abe K, Yokobayashi Y (2009)
MBio 3:e00253–00212 Mechanism-guided library design and dual
15. Dixon N, Robinson CJ, Geerlings T et al genetic selection of synthetic OFF ribo-
(2012) Orthogonal riboswitches for tuneable switches. Chembiochem 10:2375–2381
coexpression in bacteria. Angew Chem Int Ed 31. He B, Rong M, Lyakhov D et al (1997) Rapid
51:3620–3624 mutagenesis and purification of phage RNA
16. Suess B, Fink B, Berens C et al (2004) A the- polymerases. Protein Expr Purif 9:142–151
ophylline responsive riboswitch based on helix 32. Chronopoulou EG, Labrou NE (2011) Site-
slipping controls gene expression in vivo. saturation mutagenesis: a powerful tool for
Nucleic Acids Res 32:1610–1614 structure-based design of combinatorial muta-
17. Wachsmuth M, Findeiß S, Weissheimer N et al tion libraries. Curr Protoc Protein Sci Chapter
(2013) De novo design of a synthetic ribo- 26:Unit 26.6
switch that regulates transcription termination. 33. Regulski EE, Breaker RR (2008) In-line prob-
Nucleic Acids Res 41:2541–2551 ing analysis of riboswitches. Methods Mol Biol
18. Link KH, Breaker RR (2009) Engineering 419:53–67
ligand-responsive gene-control elements: les- 34. Wakeman CA, Winkler WC (2009) Analysis of
sons learned from natural riboswitches. Gene the RNA backbone: structural analysis of ribo-
Ther 16:1189–1201 switches by in-line probing and selective
19. Carothers JM, Oestreich SC, Szostak JW 2′-hydroxyl acylation and primer extension.
(2006) Aptamers selected for higher-affinity Methods Mol Biol 540:173–191
binding are not more specific for the target 35. Mandal M, Breaker RR (2004) Adenine ribo-
ligand. J Am Chem Soc 128:7929–7937 switches and gene activation by disruption of a
20. Johnson JE Jr, Reyes FE, Polaski JT et al (2012) transcription terminator. Nat Struct Mol Biol
B12 cofactors directly stabilize an mRNA regu- 11:29–35
latory switch. Nature 492:133–137 36. Myszka DG, Abdiche YN, Arisaka F et al
21. Peselis A, Serganov A (2012) Structural (2003) The ABRF-MIRG‘02 study: assembly
insights into ligand binding and gene expres- state, thermodynamic, and kinetic analysis of
sion control by an adenosylcobalamin ribo- an enzyme/inhibitor interaction. J Biomol
switch. Nat Struct Mol Biol 19:1182–1184 Tech 14:247–269
Chapter 9
Abstract
This chapter describes a genetic selection strategy to engineer synthetic riboswitches that can chemically
regulate gene expression in Escherichia coli. Riboswitch libraries are constructed by randomizing the nucle-
otides that potentially comprise an expression platform and fused to the hybrid selection/screening marker
tetA–gfpuv. Iterative ON and OFF selections are performed under appropriate conditions that favor the
survival or the growth of the cells harboring the desired riboswitches. After the selection, rapid screening
of individual riboswitch clones is performed by measuring GFPuv fluorescence without subcloning. This
optimized dual genetic selection strategy can be used to rapidly develop synthetic riboswitches without
detailed computational design or structural knowledge.
Key words Riboswitch, Aptamer, Gene regulation, RNA engineering, Translational regulation
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_9, © Springer Science+Business Media New York 2014
131
132 Yoko Nomura and Yohei Yokobayashi
Fluorescence
Aptamer NNN…NNN SD
tetA-gfpuv
- +
Clones
Fluorescence
(Tc +ligand) (NiCl2 -ligand)
- + - +
Ligand Ligand
Fig. 1 Dual genetic selection strategy. A riboswitch library is constructed in E. coli by partially randomizing an
appropriate region within the 5′ UTR. The library is subjected to one or more cycles of ON (using Tc: tetracy-
cline) and OFF (using NiCl2) selections to enrich the functional riboswitches. The selected population of E. coli
cells can be characterized as a population or as individual clones by measuring the fluorescence reporter gene
expression
2 Materials
2.1 Cell Culture 1. LB: 1 % bacto tryptone, 0.5 % yeast extract, 1 % NaCl in diH2O
Media and Strains supplemented with an appropriate antibiotic for plasmid main-
tenance. LB agar plates are prepared with 1.5 % w/v agar.
2. M9 minimal medium: Mix separately sterilized reagents in the
following order, thoroughly mixing after adding each reagent
to prepare 1 L (see Note 1): 777 mL of diH2O, 10 mL of 40 %
v/v glycerol, 0.1 mL of 1 M CaCl2, 2 mL of 1 M MgSO4,
10 mL of 10 % w/v casamino acids, 200 mL of 5× M9 salts
(64 g/L Na2HPO4·7H2O, 15 g/L KH2PO4, 2.5 g/L NaCl,
Dual Genetic Selection of Riboswitches 133
3 Methods
3.1 Cell Culture All liquid cultures are grown in M9 minimal medium or LB
Conditions medium supplemented with an appropriate antibiotic to maintain
the plasmids and kept at 37 °C in an incubator shaker (~275 rpm).
All agar plates are incubated at 37 °C overnight after plating.
3.2 Design and 1. Successful genetic selection critically depends on the use of
Construction ON optimal selection pressures that favor the growth or the sur-
and OFF Controls vival of the desired phenotypes. The optimal selection condi-
tions depend on various parameters such as promoter strength,
plasmid copy number, and host genotype. Therefore, it is
important to carefully optimize the selection conditions prior
to the actual riboswitch selection. In order to optimize the
selection conditions, we recommend the construction of two
controls, one that represents an ON state and another that
represents an OFF state of the presumed riboswitches (Fig. 2a).
The controls should be constructed using the same plasmid
backbone and the promoter with which the researcher desires
to use the functional riboswitches. The ON control could be
any canonical 5′ UTR that allows efficient translation initiation
fused to the tetA–gfpuv marker gene (see Note 5). The OFF
control should be carefully designed so that it expresses a suf-
ficiently low level of TetA–GFPuv that is acceptable for the
desired riboswitches at their OFF state. One might consider
using a “blank” plasmid (no encoded tetA–gfpuv) as an OFF
control. In reality, however, most functional riboswitches (and
other protein-based switches) produce low levels of basal
expression even at OFF states. Using a blank plasmid as the
OFF control could result in the optimized selection condition
being too stringent such that no reasonably functional ribo-
switches would survive the selection. To obtain a “realistic”
OFF control, a stable hairpin loop may be engineered to
sequester the Shine–Dalgarno (SD) sequence to suppress
translation (Fig. 2a). Such sequestration of an SD region is
often observed in natural and engineered riboswitches.
2. After construction of the ON and OFF controls, transform
the plasmids into an appropriate selection host such as TOP10
(see Note 6).
3. Start overnight cultures of the control strains in 1.0 mL of M9
medium.
Dual Genetic Selection of Riboswitches 135
Fluorescence
ON control
5’ SD tetA-gfpuv 3’
- +
Ligand
Fluorescence
OFF control
SD
5’ tetA-gfpuv 3’
- +
b Ligand
ON
OFF
OD600
OD600
OFF ON
3.3 Optimization 1. Start overnight cultures of the ON and OFF control strains in
of the Selection 1.0 mL of M9 medium.
Conditions 2. Dilute the overnight cultures (e.g., 100-fold) into fresh 1.0 mL
of M9 medium containing varying concentrations of tetracy-
cline (e.g., 0, 10, 20, 30, 40, 50 μg/mL) or NiCl2 (0.1, 0.2,
0.3, 0.4, 0.5 mM). Depending on the type of the riboswitch
that is desired, the aptamer ligand should be added to either
tetracycline (riboswitch turns ON with the ligand) or NiCl2
(riboswitch turns OFF with the ligand) cultures. Culture the
cells for 24 h.
3. Measure OD600 of each culture using a microplate reader. The
ON and OFF controls should exhibit different sensitivity pro-
files to tetracycline and NiCl2 as illustrated in Fig. 2b. Identify
the optimal tetracycline and NiCl2 concentrations that allow
selective growth of the desired phenotype. For example, in
Fig. 2b, 30 μg/mL tetracycline and 0.3 mM NiCl2 exhibit the
best ON/OFF growth discrimination (see Note 7).
PCR/DpnI/Self-ligation
Transformation
Riboswitch library
Fig. 3 Library construction strategy. The illustration shows a strategy to randomize a short (15 base) region
between an aptamer (TPP) and a canonical SD sequence by whole plasmid PCR. The primers (blue ) are 5′
phosphorylated, and one of them contains degenerate bases (N) at its 5′ terminus. After PCR, the template
plasmid is digested by Dpn I and the PCR product is self-ligated using T4 DNA ligase. Transformation should
yield >105 colonies (color figure online)
3.5 Dual Genetic 1. Prior to initiating selection, culture the library along with the
Selection ON and OFF controls as described in Subheading 3.3 with and
without the aptamer ligand except omitting the selective reagents
(tetracycline or NiCl2). Measure the GFPuv levels of each cul-
ture as described above. The average fluorescence of the naive
library can guide the selection strategy described below. In dual
genetic selection, it is desirable to eliminate as much nonfunc-
tional mutants as possible in the first selection to avoid further
dilution of the functional clones. Therefore, if the library popu-
lation is generally OFF (low fluorescence), it is advisable to
begin with the ON selection. If the library population is gener-
ally ON, the OFF selection should be performed first.
2. Start an overnight culture of the cell population to be selected
in M9 medium.
3. Dilute the overnight culture ~100-fold into fresh M9 medium
(1.0 mL) with or without the aptamer ligand (depending on
the desired riboswitch characteristics and ON or OFF selection
to be performed) and culture for ~8 h. This nonselective
growth is necessary to enable each riboswitch mutant to adapt
to the presence or the absence of the ligand.
4. Dilute the cells ~100-fold into fresh M9 medium containing
an appropriate concentration of the selective reagent (tetracy-
cline for ON selection and NiCl2 for OFF selection) and cul-
ture for an additional 24 h.
5. Wash the cells with M9 medium and store in 20 % glycerol for
additional rounds of selection and/or characterization.
6. Complete both ON and OFF selections as described above.
Depending on the library, additional cycles of ON/OFF selec-
tions may need to be performed to enrich the functional
riboswitches.
7. After one or more rounds of ON/OFF selection, evaluate the
selected cell population’s response to the aptamer ligand in
liquid culture as described in step 1 above. If the cells respond
to the aptamer ligand as a population, it strongly suggests that
the selection successfully enriched the desired riboswitches;
therefore, one should proceed to screening of individual clones
outlined below (3.6). If no response is observed, selection
conditions and/or library design may need to be further opti-
mized or additional cycles of selection may need to be per-
formed (see Note 9).
3.6 Screening of 1. Plate the selected cells on an LB agar plate to isolate single
Individual Clones colonies.
2. Pick and culture appropriate number (>10) of colonies in M9
medium overnight.
Dual Genetic Selection of Riboswitches 139
4 Notes
Acknowledgments
References
1. Sinha J, Reyes SJ, Gallivan JP (2010) screening of gene switches in Escherichia coli.
Reprogramming bacteria to seek and destroy an Nucleic Acids Res 37:e39
herbicide. Nat Chem Biol 6:464–470 5. Nomura Y, Yokobayashi Y (2007) Dual selec-
2. Winkler WC, Breaker RR (2005) Regulation of tion of a genetic switch by a single selection
bacterial gene expression by riboswitches. Annu marker. Biosystems 90:115–120
Rev Microbiol 59:487–517 6. Podolsky T, Fong ST, Lee BT (1996) Direct
3. Wittmann A, Suess B (2012) Engineered ribo- selection of tetracycline-sensitive Escherichia coli
switches: expanding researchers’ toolbox with cells using nickel salts. Plasmid 36:112–115
synthetic RNA regulators. FEBS Lett 586: 7. Muranaka N, Abe K, Yokobayashi Y (2009)
2076–2083 Mechanism-guided library design and dual
4. Muranaka N, Sharma V, Nomura Y et al (2009) genetic selection of synthetic OFF riboswitches.
An efficient platform for genetic selection and Chembiochem 10:2375–2381
Chapter 10
Abstract
Ever-increasing repertories of RNA-based switching devices are enabling synthetic biologists to construct
compact, self-standing, and easy-to-integrate regulatory circuits. However, it is rather rare that the existing
RNA-based expression controllers happen to have the exact specification needed for particular applications
from the beginning. Evolutionary design of is powerful strategy for quickly tuning functions/specification
of genetic switches. Presented here are the steps required for rapid and efficient enrichment of genetic
switches with desired specification using recently developed nucleoside kinase-based dual selection system.
Here, the library of genetic switches, created by randomizing either the part or the entire sequence coding
switching components, is subjected to OFF (negative) selection and ON (positive) selection in various
conditions. The entire selection process is completed only by liquid handling, facilitating the parallel and
continuous operations of multiple selection projects. This automation-liable platform for genetic selection
of functional switches has potential applications for development of RNA-based biosensors, expression
controllers, and their integrated forms (genetic circuits).
Key words Thymidine kinase, Nucleoside analog, Genetic selection, Dual selection, Directed
evolution, Genetic switch
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_10, © Springer Science+Business Media New York 2014
141
142 Kohei Ike and Daisuke Umeno
Fig. 1 Dual selection of genetic switches using nucleoside kinase activity. The entire sequence or a part of the
genetic switch component is randomized either by error-prone PCR or by oligonucleotide-based randomiza-
tion. The resultant “switch library” is then placed on the condition to be at OFF state and then subjected to OFF
selection by adding nucleoside dP. The survivor pool is then placed on the condition to be ON, subjected to ON
selection by the growth in the presence of dT and 5FdU
2 Materials
2.1 Library Creation 1. Template plasmid (pTrcHis2-luxR) (see Notes 1 and 2).
(PCR Random
2. PCR primers (forward and reverse) (see Note 3):
Mutagenesis)
Primer 1: 5′-CAATCTGTGTGGGCACTCGAC-3′.
Primer 2: 5′-TACTGCCGCCAGGCAAATTC-3′.
3. Taq polymerase.
4. 10× Taq buffer.
5. 10× dNTP mixture: 2 mM each of dATP, dTTP, dCTP, dGTP.
Prepare 50 μL aliquots of this mixture (to avoid excessive
freeze/thaw cycles) and store at −20 °C.
144 Kohei Ike and Daisuke Umeno
2.2 ON/OFF Selection 1. Selector plasmid (for details, see Notes 2 and 4).
2. E. coli JW1226 (KEIO collection, tdk strain) (see Note 5, [9]).
3. 50 mg/mL Carbenicillin (1,000× stock).
4. 30 mg/mL Chloramphenicol (1,000× stock).
5. Luria-Bertani (LB): 20 g of premixed LB medium (Invitrogen)
into 1 L of deionized water. Autoclave at 121 °C for 20 min.
6. SOC media: Add 20 g of bacto-tryptone, 5 g of yeast extract,
and 0.5 g of NaCl into 950 mL of deionized water. Add 10 mL
of a 250 mM solution of KCl. Adjust the volume of the solu-
tion to 1 L with deionized water. Autoclave at 121 °C for
20 min. Just before use, add 5 mL of a sterile solution of 2 M
MgCl2 and 20 mL of a sterile 1 M solution of glucose [10].
7. dP, 6-(β-D-2-Deoxyribofuranosyl)-3,4-dihydro-8H-pyrimido
[4,5-c][1,2]oxazin-7-one) [3, 11]: Prepare 1 μM–1 mM (typi-
cally 100 nM) dP solution dissolved in DMSO as 1,000× stock.
Sample stored at 4 °C.
8. OFF selection medium: LB containing 10–1,000 nM dP [3].
9. 10 mg/mL Thymidine in deionized water (1,000× stock):
Stock stored at 4 °C.
10. 1 mg/mL Adenosine in deionized water (1,000× Stock): Stock
stored at 4 °C.
11. 20 mg/mL 5FdU in deionized water (1,000× stock): Stored
at −20 °C.
12. Tryptone broth: Add 20 g of tryptone and 5 g of NaCl into
1 L of deionized water. Autoclave at 121 °C for 20 min.
13. ON selection medium: 10 μg/mL thymidine, 1 μg/mL ade-
nosine, and 20 μg/mL 5FdU [3, 4] in tryptone broth.
14. 10 mM stock solution of 3OC6-HSL in ethyl acetate acidified
with glacial acetic acid (0.001 % (v/v)): Stored at −20 °C.
2.3 Evaluating the 1. Reporter plasmid (for details, see Note 4).
Selected Switches 2. E. coli JW1226 (see Note 5, [9]).
3. Carbenicillin stock (see Subheading 2.2, item 3).
4. Chloramphenicol (see Subheading 2.2, item 4).
5. LB-agar (see Subheading 2.2, item 5).
6. SOC media (see Subheading 2.2, item 6).
7. NaCl solution: Add 9 g of NaCl into 1 L of deionized water.
Autoclave at 121 °C for 20 min.
8. Flow cytometer: MACS Quant VYB (Miltenyi Biotec).
9. Fluorescence plate reader: Fluoroskan Ascent® (Thermo Fisher
Scientific Inc.).
10. Plate reader: SpectraMax Plus384 (molecular devices).
146 Kohei Ike and Daisuke Umeno
3 Methods
3.1 Library Creation There are a variety of established methods for the construction of
random libraries [12]. Among them, error-prone PCR is the most
popular method that could apply for a region or the entire part of
the targeted gene. The simplest is the addition of Mn2+ into the
PCR, where manganese ion lowers the fidelity of the polymerase,
inserting random base substitutions into the target sequence [13].
1. For each PCR sample, add to tube 2 fmol of template DNA
(pTrcHis2-luxR), 5 μL of 10× Taq buffer, and 5 μL of 10×
dNTP mixture. 5 μL of 5 μM each primers (to be 25 pmol in
final concentration), 1 μL of Taq polymerase (5 U), 5 μL of
10× MnCl2 solution (to be 10–100 μM in final concentration),
and nuclease-free water to a final volume of 50 μL.
2. Mix the sample by pipetting, and confirm all the solution is on
the bottom.
3. Place the tubes in a thermal cycler, and run the following PCR
program: (1) 5-min initial denature at 94 °C; (2) 30-s denature
at 94 °C, 30-s annealing at the temperature designed for prim-
ers, and 1-min extension at 72 °C; repeat for 25 cycles; and (3)
10 min at 72 °C for final extension (see Notes 6 and 7).
4. Run the product for gel electrophoresis to estimate the yield of
full-length gene (see Notes 6 and 8).
5. Clean the PCR product by using a Zymo DNA clean and con-
centrator kit. In parallel, clean up the vector in the same way.
6. Digest both PCR product and the expression vector with
appropriate enzymes (NcoI/HindIII).
7. Gel-purify the digested samples using Zymoclean™ Gel DNA
Recovery Kit (see Note 9).
8. Treat the mixture of the purified insert (100 ng) and vector
(100 ng) with ligase buffer and T4 ligase for 2 h (400 units/
reaction to total volume of 10 μL) (see Note 10).
9. Optionally, concentrate/purify the ligation product by using a
Zymo DNA clean and concentrator kit (see Note 11).
10. Electroporate the ligation mixture (1 μL) into the competent
cell (40 μL) (see Note 12).
11. Add 1 mL of SOC media immediately after the electropora-
tion, and shake it for 1 h at 37 °C. After washing the transfor-
mants with fresh LB media, resuspend the cells into 10 mL of
LB media (see Notes 13 and 14).
12. After overnight shaking at 37 °C, miniprep the portion
(1–2 mL) of the above culture to obtain plasmid library
pTrcHis2-[luxR].
Nucleotide Kinase-Based Selection System for Genetic Switches 147
3.2 Selection of the 1. Add 100 ng (1 μL) of plasmid library (pTrcHis2-[luxR]) into
LuxR Variants with 40 μL of JW1226 cell harboring pAC-Plux-hsvtk (see Note 15).
Proper Switching 2. Add 1 mL of SOC media and gently shake for 1 h at 30 °C.
3. Collect the cells by centrifugation (4,000 rcf, 3 min), resus-
pend them into the 10 mL of LB media containing chloram-
phenicol (30 μg/mL) and carbenicillin (50 μg/mL), and allow
for overnight shaking at 37 °C.
4. OFF selection: Transfer about 106 cells into the 1 mL of OFF
selection medium containing 0–1,000 nM 3OC6-HSL and
leave in the shaking incubator (37 °C) for 2 h (see Notes 16
and 17).
5. Wash the cell by centrifugation (4,000 rcf, 3 min)/resuspen-
sion to fresh LB media containing the same concentration
of 3OC6-HSL at OFF selection. Repeat this process twice
(see Note 18).
6. Shake the culture for another 6 h at 37 °C (see Notes 19 and 20).
7. ON selection: From the above culture, take 10 μL (about 106
cells) to resuspend into 1 mL of ON selection medium con-
taining desired concentration of inducer (3OC6-HSL).
8. Shake the cell culture for another 20 h at 37 °C (see Note 21).
9. Miniprep the culture to collect the plasmid containing survivor
variants.
3.3 Characterization 1. Electroporate the library plasmids before and after the selec-
of the Survivor Pools tion into JW1226 harboring pAC-Plux-gfpuv.
and Isolation of 2. Add 1 mL of SOC and incubate at 37 °C for 1 h in shaker.
Switch Variants 3. After collecting the cells by centrifugation (4,000 rcf, 3 min),
resuspend them into the 10 mL of LB media containing chlor-
amphenicol (30 μg/mL) and carbenicillin (50 μg/mL), and
allow for overnight shaking at 37 °C.
4. Take 106 cells into fresh 500 μL of LB media containing vari-
ous concentrations (0–1 μM) of 3OC6-HSL and shake them
for another 12 h. Then proceed to step 5 or 6.
5. Population analysis (Fig. 3a): Wash the cell by saline solution.
Dilute the cell by about 1,000×. The diluted cell suspension is
subjected to flow cytometry (V2 channel, laser 405 nm, filter
525/50 nm) for the analysis of the pool.
6. Isolation of the switch variants: A fraction of transformant is
plated on LB-agar plate containing chloramphenicol (30 μg/
mL) and carbenicillin (50 μg/mL) to form colonies (see Note
20). Pick several to dozens of colonies from the plates, and
inoculate them into LB (2 mL) culture containing chloram-
phenicol (30 μg/mL) and carbenicillin (50 μg/mL) in deep-
well 96 plates, allowing the overnight growth at 37 °C.
148 Kohei Ike and Daisuke Umeno
Fig. 3 (a) Flow cytometric analysis of the library of HSL-induced genetic switches before and after the OFF and
ON selection in the presence of 10 and 1,000 nM 3OC6-HSL. Original variant mixture (library) was two-peaked
in any concentration of HSL, but after single round of OFF selection in the presence of 10 nM HSL and OFF
selection in the presence of 1,000 nM HSL, the properly responding LuxR/Plux systems were enriched.
(b) [HSL]-output transfer function of representative LuxR clones isolated from the survivor pool. Filled circle
represents the switching property of wild-type LuxR, while open circle, open triangle, and open squares rep-
resent that of LuxR variants
4 Notes
References
1. Nomura Y, Yokobayashi Y (2007) Dual selec- 7. Tabor JJ, Salis HM, Simpson ZB et al (2009) A
tion of a genetic switch by a single selection synthetic genetic edge detection program. Cell
marker. Biosystems 90:115–120 137:1272–1281
2. Muranaka N, Sharma V, Nomura Y et al (2009) 8. Danino T, Mondragón-Palomino O, Tsimring
An efficient platform for genetic selection and L et al (2010) A synchronized quorum of
screening of gene switches in Escherichia coli. genetic clocks. Nature 463:326–330
Nucleic Acids Res 37:e39 9. Baba T, Ara T, Hasegawa M et al (2006)
3. Tashiro Y, Fukutomi H, Terakubo K et al Construction of Escherichia coli K-12 in-frame,
(2011) A nucleoside kinase as a dual selector single-gene knockout mutants: the Keio collec-
for genetic switches and circuits. Nucleic Acids tion. Mol Syst Biol 2:1–11
Res 39:e12 10. Sambrook J, Russell DW (2001) Molecular clon-
4. Yagila E, Rosnerb A (1971) Phosphorolysis of ing: a laboratory manual, 3rd edn. Cold Spring
5-fluoro-2′-deoxyuridine in Escherichia coli and Harbor Laboratory, Cold Spring Harbor, NY
its inhibition by nucleosides. J Bacteriol 108: 11. Negishi K, Loakes D, Schaaper RM (2002)
760–764 Saturation of DNA mismatch repair and error
5. Weiss R, Knight TF (2001) Engineered com- catastrophe by a base analogue in Escherichia
munications for microbial robotics, vol coli. Genetics 161:1363–1371
2054, Lecture Notes in Computer Science., 12. Arnold FH, Georgiou G (2003) Directed evo-
pp 1–16 lution library creation: methods and protocols.
6. Basu S, Gerchman Y, Collins CH et al (2005) Humana, Totowa, NJ
A synthetic multicellular system for pro- 13. Cirino PC, Mayer KM, Umeno D (2003)
grammed pattern formation. Nature 434: Generating mutant libraries using error-prone
1130–1134 PCR. Methods Mol Biol 231:3–9
Chapter 11
Abstract
We present a simple method to measure the real-time activity of riboswitches with purified components in
vitro and inside of artificial cells. Typically, riboswitch activity is measured in vivo by exploiting β-galactosidase
encoding constructs with a putative riboswitch sequence in the untranslated region. Additional in vitro char-
acterization often makes use of in-line probing to explore conformational changes induced by ligand binding
to the mRNA or analyses of transcript lengths in the presence and absence of ligand. However, riboswitches
ultimately control protein levels and often times require accessory factors. Therefore, an in vitro system
capable of monitoring protein production with fully defined components that can be supplemented with
accessory factors would greatly aid riboswitch studies. Herein we present a system that is amenable to such
analyses. Further, since the described system can be easily reconstituted within compartments to build artifi-
cial, cellular mimics with sensing capability, protocols are provided for building sense-response systems within
water-in-oil emulsion compartments and lipid vesicles. Only standard laboratory equipment and commer-
cially available material are exploited for the described assays, including DNA, purified transcription–transla-
tion machinery, i.e., the PURE system, and a spectrofluorometer.
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_11, © Springer Science+Business Media New York 2014
153
154 Laura Martini and Sheref S. Mansy
fluorescence
n time
atio
te riz
ac
ar
ch
ce
llu
lar
mi
mi
cs
Fig. 1 Cell-free systems for in vitro riboswitch characterization and the construction of artificial, cellular mim-
ics. The PURE system is used to characterize in real-time riboswitch activity through the expression of a
reporter protein either in vitro or inside of a compartment with dimensions similar to living cells
2 Materials
3 Methods
3.1 DNA Template The DNA template can be either a circular plasmid or a linear PCR
Preparation product that contains a series of modular elements to allow for ribo-
switch controlled protein synthesis (Fig. 2). The sequence should
contain a transcriptional promoter, a sequence encoding the ribo-
switch that contains a ribosome binding site (preferably the natural
ribosome binding site sequence, if possible), a gene coding for a fluo-
rescent protein to act as a reporter, and a transcriptional terminator.
The transcriptional promoters for T7 and E. coli RNA polymerases are
typically used. However, since the activity of a riboswitch can depend
Cell-free Riboswitch Activity 157
Fig. 2 The composition of a theophylline riboswitch for the real-time observation of riboswitch activity. The
construct shown here contains a standard T7 transcriptional promoter, a theophylline riboswitch that contains
a ribosome binding site, a gene that codes for A206K YPet (mYPet), and a T7 transcriptional terminator. Only
the start and stop codons of mYPet are shown. The riboswitch sequence is shown in bold
3.2 In Vitro The major advantage of working in vitro, compared with in vivo, is
Transcription and that the system operates only with what is provided. In other
Translation Reaction words, in vitro activity cannot depend on unidentified cellular
components, because they are not present. To date, only E. coli
[24] and Thermus thermophilus [25] translation machinery have
been reconstituted in vitro from purified components. Of these
two, only the E. coli system, i.e., the PURE system, is commercially
available. It should be noted that in contrast to in vivo or cell-
extract conditions, reactions with purified transcription–translation
machinery do not contain nucleases. The lack of nucleases decreases
the amount of DNA template needed. Whether protein produc-
tion is more or less efficient with the PURE system in comparison
with cell-extract based systems depends on the specific folding
properties of the expressed protein.
1. The PURE system components should be aliquoted on ice and
stored in 0.2 mL microcentrifuge tubes. Convenient volumes
are 10 µL aliquots of solution A and 7.5 µL aliquots of solu-
tion B. Aliquots are stored at −80 °C.
2. Assemble the reaction components, except for the DNA tem-
plate, on ice following the manufacturer’s instructions.
Supplementary reagents can also be added, such as RNase
inhibitor (e.g., 20 U RiboLock RNase Inhibitor) or the ribo-
switch ligand (e.g., 0.5 mM theophylline).
3. The assembled reaction is transferred to a quartz cuvette and
incubated at 37 °C.
4. The reaction is initiated by the addition of the DNA template
and monitored by fluorescence spectroscopy for 6 h (Fig. 3)
(see Note 2). The DNA template concentration should be
screened. We used 250 ng of plasmid template in 25.5 µL
total reaction volume. If YPet is used as the reporter protein,
the excitation and emission wavelengths are 517 nm and
530 nm, respectively.
3.3 In Vitro Since cellular life is chemically distinct from the environment,
Compartmentalization efforts to mimic cells in the laboratory often times exploit w/o
emulsion droplets or vesicles to approximate the compartment of
the living, chemical system [26, 27]. However, even if cellular life
is distinct from the environment, life cannot exist in isolation and
must in some manner interface with the environment to survive
[28]. Since lipid vesicles are semipermeable and more similar to the
types of barriers found in biology, vesicles are better suited than
w/o emulsion droplets for the construction of cellular mimics.
Nevertheless, the encapsulation efficiency of w/o emulsions is
nearly 100 %, whereas encapsulation efficiency in vesicles is at best
30 % [29]. It is for this reason that the screening of compartmen-
talized reactions is carried out with w/o emulsions. Once optimal
conditions are identified, similar vesicle systems are setup.
Cell-free Riboswitch Activity 159
500 ON
200
100 OFF
0
0 100 200 300 400
time (min)
Fig. 3 In vitro theophylline riboswitch activity observed by measuring the expression of the reporter protein
mYPet. The presence of the ligand activates protein expression. A schematic representation of the mRNA in the
ligand-bound ON state and the uncomplexed OFF state are shown. Protein production is inhibited in this case
in the OFF state because the RBS is not available for base-pairing with the ribosome. The data were taken from
a previous in vitro theophylline riboswitch study [18]
3.3.1 Cell-Free The method described here is based on that of Davidson et al. [30]
Expression in w/o but has been scaled down to be compatible with small volume
Emulsion PURE system reactions. Positive displacement pipettes are used to
handle oil samples.
1. The oil phase that will be used for the w/o emulsion is assem-
bled in a 15 mL Falcon tube (see Note 3), in the following
order: 474.75 µL of mineral oil, 22.5 µL of span 80, 2.5 µL of
tween 80, and 0.25 µL of triton X-100.
2. The aqueous phase is first assembled in a microcentrifuge tube
by mixing the PURE system components on ice as described
above for the in vitro reactions. Note that the optimal DNA
template concentration may be different when transcription–
translation is performed in a compartment versus in vitro. We
used 500 ng of plasmid DNA in 25.5 μL of total aqueous vol-
ume (see Note 4) for expression in w/o emulsion droplets.
The theophylline concentration was also increased to 5 mM to
compensate for partitioning into the oil phase (see Note 5).
3. The 15 mL tube containing the oil phase is placed in a 250 mL
beaker filled with ice water on a magnetic stir plate. A teflon
stir bar is inserted in the oil phase and the oil is mixed by stir-
ring at maximum speed for 1 min.
4. The emulsion is formed by the drop-wise addition of the aque-
ous phase containing the PURE system reaction to the oil
phase over 1 min with continuous stirring. The emulsion is
then stirred for an additional 3 min (see Note 6).
5. Finally, the emulsion is transferred to a 2 mL microcentrifuge
tube and incubated at 37 °C for 6 h. 5 μL aliquots are removed
every hour for observation by fluorescence microscopy.
160 Laura Martini and Sheref S. Mansy
4 Notes
Fig. 4 Fluorescence microscopy images of riboswitch activity in compartments. Upper panels (a–c): activity
recorded in emulsion compartments. Middle panels (d–f): activity recorded in liposomes. Lower panels (g, h):
activity due to riboswitch sensing of the environment. Images are all epifluorescence, except for panels a, d, g,
which are bright-field. Panel b, e show the fluorescence resulting in compartments when the ligand is present.
The corresponding controls (i.e., in the absence of ligand) are also shown (c, f). In panel h, the activity of a ribo-
switch as a sensor element is shown. The data are from the characterization of a cell-free theophylline riboswitch
system and panels g and h are reproduced with permission from the Royal Society of Chemistry [18]
References
1. Soukup GA, Breaker RR (1999) Relationship 12. Topp S, Gallivan JP (2007) Guiding bacteria
between internucleotide linkage geometry and with small molecules and RNA. J Am Chem
the stability of RNA. RNA 5:1308–1325 Soc 129:6807–6811
2. Nahvi A, Sudarsan N, Ebert MS, Zou X, Brown 13. Soukup GA, Breaker RR (1999) Engineering
KL, Braker RR (2002) Genetic control by a precision RNA molecular switches. Proc Natl
metabolite-binding mRNA. Chem Biol 9:1043 Acad Sci U S A 96:3584–3589
3. Wakeman CA, Winkler WC (2009) Analysis of 14. Wickiser JK, Winkler WC, Breaker RR,
the RNA backbone: structural analysis of ribo- Crothers DM (2005) The speed of RNA tran-
switches by in-line probing and selective scription and metabolite binding kinetics oper-
2′-hydroxyl acylation and primer extension. In: ate an FMN riboswitch. Mol Cell 18:49–60
Serganov A (ed) Riboswitches. Methods Mol 15. Tawfik DS, Griffiths AD (1998) Man-made
Biol, vol 540, pp 173–191 cell-like compartments for molecular evolu-
4. Ellington AD, Szostak JW (1990) In vitro tion. Nat Biotechnol 16:652–656
selection of RNA molecules that bind specific 16. Sunami T, Matsuura T, Suzuki H, Yomo T
ligands. Nature 346:818–822 (2010) Synthesis of functional proteins within
5. Serganov A, Yuan Y-R, Pikovskaya O, liposomes. Methods Mol Biol 607:243–256
Polonskaia A, Malinina L, Phan AT, Hobartner 17. Lynch SA, Gallivan JP (2009) A flow
C, Micura R, Breaker RR, Patel DJ (2004) cytometry-based screen for synthetic ribo-
Structural basis for discriminative regulation of switches. Nucleic Acids Res 37:184–192
gene expression by adenine- and guanine- 18. Martini L, Mansy SS (2011) Cell-like systems
sensing mRNAs. Chem Biol 11:1729–1741 with riboswitch controlled gene expression.
6. Serganov A, Nudler E (2013) A decade of Chem Commun 47:10734–10736
riboswitches. Cell 152:17–24 19. Lemay J-F, Desnoyers G, Blouin S, Heppell B,
7. Desai SK, Gallivan JP (2004) Genetic screens Bastet L, St-Pierre P, Massé E, Lafontaine DA
and selections for small molecules based on a (2011) Comparative study between transcrip-
synthetic riboswitch that activates protein trans- tionally- and translationally-acting adenine
lation. J Am Chem Soc 126:13247–13254 riboswitches reveals key differences in ribo-
8. Nomura Y, Yokobayashi Y (2007) switch regulatory mechanisms. PLoS Genet
Reengineering a natural riboswitch by dual 7:e1001278
genetic selection. J Am Chem Soc 20. Lentini R, Forlin M, Martini L, Del Bianco C,
129:13814–13815 Spencer AC, Torino D, Mansy SS (2013)
9. Artsimovitch I, Henkin TM (2009) In vitro Fluorescent proteins and in vitro genetic
approaches to analysis of transcription termina- organization for cell-free synthetic biology.
tion. Methods 47:37–43 ACS Synth Biol 2:482–489
10. Mironov A, Epshtein V, Nudler E (2009) 21. Aiba H, Hanamura A, Yamano H (1991)
Transcriptional approaches to riboswitch stud- Transcriptional terminator is a positive regula-
ies. IN: Serganov A (Ed) Riboswitches. tory element in the expression of the Escherichia
Methods Mol Biol, vol 540: 39-51, Humana coli crp gene. J Biol Chem 266:1721–1727
Press, NY, USA 22. Sambrook J, Russel DW (2001) Commonly
11. Muranaka N, Yokobayashi Y (2010) used techniques in molecular cloning. In:
Posttranscriptional signal integration of engi- Molecular cloning. (Ed) Jan Argentine Cold
neered riboswitches yields band-pass output. Spring Harbor Laboratory Press, NY, USA
Angew Chem Int Ed 49:4653–4655
164 Laura Martini and Sheref S. Mansy
23. Sambrook J, Russell DW (2006) Standard eth- liposomes: efficiency depends on interaction
anol precipitation of DNA in microcentrifuge between protein and phospholipid bilayer.
tubes. Cold Spring Harb Protoc. doi:10.1101/ BMC Biotechnol 2:9
pdb.prot4456 30. Davidson EA, Dlugosz PJ, Levy M, Ellington
24. Shimizu Y, Inoue A, Tomari Y, Suzuki T, AD (2009) Directed evolution of proteins
Yokogawa T, Nishikawa K, Ueda T (2001) in vitro using compartmentalization in
Cell-free translation reconstituted with purified emulsions. Curr Protoc Mol Biol. doi:
components. Nat Biotechnol 19:752–755 10.1002/0471142727.mb2406s87
25. Zhou Y, Asahara H, Gaucher EA, Chong S 31. Lazzerini-Ospri L, Stano P, Luisi P, Marangoni
(2012) Reconstitution of translation from R (2012) Characterization of the emergent
Thermus thermophilus reveals a minimal set of properties of a synthetic quasi-cellular system.
components sufficient for protein synthesis at BMC Bioinformat 13(Suppl 4):S9
high temperatures and functional conservation 32. Nguyen AW, Daugherty PS (2005)
of modern and ancient translation compo- Evolutionary optimization of fluorescent pro-
nents. Nucleic Acids Res 40:7932–7945 teins for intracellular FRET. Nat Biotechnol
26. Forlin M, Lentini R, Mansy SS (2012) Cellular 23:355–360
imitations. Curr Opin Chem Biol 16:586–592 33. Donoso P, O'Neill SC, Dilly KW, Negretti N,
27. Ichihashi N, Matsuura T, Kita H, Sunami T, Eisner DA (1994) Comparison of the effects of
Suzuki H, Yomo T (2010) Constructing par- caffeine and other methylxanthines on [Ca2+]i
tial models of cells. Cold Spring Harb Perspect in rat ventricular myocytes. Br J Pharmacol
Biol. doi:10.1101/cshperspect.a004945 111:455–458
28. Bianco CD, Mansy SS (2012) Non replicating 34. Pereira de Souza T, Stano P, Luisi PL (2009) The
protocells. Acc Chem Res 45:2125–2130 minimal size of liposome-based model cells brings
29. Colletier J-P, Chaize B, Winterhalter M, about a remarkably enhanced entrapment and
Fournier D (2002) Protein encapsulation in protein synthesis. Chembiochem 10:1056–1063
Chapter 12
Abstract
Riboswitches are composed of two regions: one for binding to the ligand (the aptamer domain) and the
other for regulating the expression of the gene (the expression platform). In most riboswitches (both natu-
ral and artificial), a part of the aptamer domain required for ligand binding is directly involved in the regu-
lation of expression, so that it is difficult to design other ligand-responsive riboswitches based on these
riboswitches even by using artificial aptamers obtained through in vitro selection. This chapter describes a
method for rationally constructing a foundational ON-riboswitch, which is easily available for the design
of other ligand-dependent riboswitches, by introducing a new region (a modulator sequence: MS) in addi-
tion to the two basic regions. A facile method for preparing arbitrary molecule-dependent riboswitches
based on the foundational riboswitch is also presented.
Key words Riboswitch, IRES, Aptamer, Gene regulation, Biosensor, Cell-free translation
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_12, © Springer Science+Business Media New York 2014
165
166 Atsushi Ogawa
Fig. 1 An example of natural riboswitches: add adenine ON-riboswitch in Vibrio vulnificus. In the absence of
the ligand (adenine), the aTI hybridizes to the TI (the RBS and the start codon) to prevent the bacterial 30S
ribosome from binding to the RBS (OFF state, above). In the presence of the ligand, it leaves the TI to bind to
the aaTI and the ligand (dashed thin line), so that the ribosome can access the RBS to start translation (ON
state, below). Bold and thin lines represent mRNA and base-pair interactions, respectively
2 Materials
2.2 Preparation 1. DNA template with the IRES sequence chosen as described in
of DNA Templates Subheading 3.1.1 (see Note 6).
2.2.1 Amplification 2. Plasmid encoding the luciferase gene (see Note 7).
of DNA Templates 3. PrimeSTAR MAX DNA polymerase including dNTPs (Takara Bio,
Shiga, Japan) or other high-fidelity DNA polymerase with dNTPs.
4. Primers for the upstream part: various forward primers includ-
ing a variety of regulator sequences (see Note 8) and a reverse
primer with a unique restriction enzyme site (RES).
5. Primers for the downstream part: a forward primer with the
same RES as in the reverse primer for the upstream part and a
reverse primer (see Note 9).
6. Forward primer including the T7 promoter (5′-TAATACGA
CTCACTATA-3′) for the final PCR.
2.7 Equipment 1. Thermal cycler for PCR, digestion, ligation, transcription, and
translation.
2. Agarose gel electrophoresis apparatus.
3. UV transilluminator for excitation of ethidium bromide bound
to DNA.
4. Incubator for dissolving of the gel slices at 50–60 °C.
5. Deep freezer.
6. UV spectrometer.
7. Microplate luminometer.
8. Centrifuge.
170 Atsushi Ogawa
3 Methods
This methods section is divided into two subsections: one is for the
design of riboswitches, and the other is for the actual experiments,
such as the construction of DNA templates, transcription, transla-
tion, and reporter protein assays. Experimental procedures are
common in any design step, and thus see the experimental subsec-
tion (Subheading 3.2) for preparing mRNAs and measuring their
riboswitch activities in each design step.
3.1 Design Methods An IRES that has been much studied (especially regarding its sec-
ondary and tertiary structure) and that works with higher transla-
3.1.1 Choice of an IRES
tion efficiency is a better choice than a less-studied IRES. One of
and a Reporter Gene
the best candidates is the intergenic IRES of the Plautia stali intes-
Downstream of the IRES
tine virus (PSIV), which meets these conditions and functions in
many types of eukaryotic translation systems [10–12] (see Note
15). Other IRESs could be used depending on the user’s circum-
stances. For a reporter gene, the firefly luciferase gene is recom-
mended due to its low background, low detection limit, and linear
response over a wide range (see Note 16).
3.1.2 General Design The following parts are required for mRNAs (in this order): a stable
of mRNAs stem-loop (5′-SL), a mimic start codon (optional), regulators (vari-
able), the IRES (see Note 17), the reporter gene (see Note 18), and
a 3′ untranslated region (UTR) (Fig. 2). The stable stem-loop
structure in the 5′-terminus is required for inhibiting 5′ terminus-
mediated translation of the reporter gene [13] (see Note 19). An
8-base stem with an ACAAC pentaloop (e.g., 5′-GGGAGAC
CACAACGGUUUCCC-3′) is sufficient for inhibition, but it
should start from 5′ G for efficient transcription by T7 RNA poly-
merase. A mimic gene in the 5′ UTR is also usable for the inhibi-
tion, because the eukaryotic ribosome tends to start translation at
the start codon that it encounters first (see Note 20). Regulators are
a set of several sequences involved in regulation of gene expression:
a modulator sequence (MS), an anti-anti-IRES (aaIRES), an
aptamer, and an anti-IRES (aIRES), the details of which are
described below. More than 250 nucleotides (nt) of the 3′ UTR are
needed to suppress mRNA degradation from the 3′ end during
translation.
Fig. 2 General design of mRNAs. SL stem-loop, mAUG mimic start codon, UTR
untranslated region, PK pseudoknot, I-dORF a 5′ part of the original downstream
ORF of the chosen IRES. The existence or nonexistence of parts in parentheses
depends on the circumstances of the experiment. The eukaryotic 40S ribosome
preferably binds to the IRES (not to the 5′ terminus) due to the 5′-SL
Fig. 4 Design of IRES-based ON-riboswitches. In the absence of the ligand, since the MS hybridizes to the 3’
end of the aaIRES (and the 5’ end of the aptamer), the aIRES binds to the IRES to inhibit IRES-mediated transla-
tion (OFF state, above). In the presence of the ligand, it releases the aptamer (due to aptamer–ligand complex
formation) and then leaves the aaIRES (due to destabilization of the MS-SL), so that the aIRES binds not to the
IRES but to the aaIRES to promote IRES-mediated translation (ON state, below)
3.1.7 Rational Design of Since the MS-SL in the foundational riboswitch with the highest
Other Ligand-Dependent switching efficiency has the almost-minimum free energy necessary
ON-Riboswitches for inhibiting IRES-mediated translation in the OFF state, any
riboswitch whose MS-SL has similar stability is expected to work
well as a riboswitch. Therefore, arbitrary ligand-dependent
ON-riboswitches can be easily designed as follows:
1. Calculate the free energy (delta G value) of the cropped MS-SL
part in the foundational riboswitch by using the RNAstructure
software [15].
2. On a computer, replace the aptamer and the 5′ part of the MS
in the foundational riboswitch with an aptamer for the ligand
of interest and various lengths of the sequence complementary
to the 5′ end of the chosen aptamer, respectively (see Note 26).
3. Calculate free energies of their MS-SL part and determine
which has the nearest value to the free energy of the MS-SL in
the foundational riboswitch (see Note 32).
4. Prepare actual mRNA with the most promising MS-SL (and
mRNAs with the second and third most promising MS-SL if
necessary), and confirm its switching efficiency in the presence
or absence of the ligand (see Notes 29 and 31).
3.2 Experimental DNA templates are constructed by preparing the upstream part
Methods including the IRES and the downstream part including the
luciferase gene separately and then joining them together
3.2.1 Construction of
(Fig. 5) (see Note 33).
DNA Templates for mRNA
Rational Design of Riboswitches 175
Fig. 5 General procedures of the construction of DNA templates and their transcripts (mRNA)
3.2.2 Preparation of The following protocol is for the preparation of 5′ cap- and 3′
mRNA (In Vitro poly(A)-free mRNAs. Both the 5′ cap and poyl(A) are not necessarily
Transcription) required for both efficient 5′ terminus- and IRES-mediated transla-
tion in the wheat germ extract described here (see Notes 19 and 35)
1. Transcribe the DNA templates with the MEGAscript T7 Kit
according to the manufacturer’s protocol. Typically, incubate
10 µL of the reaction solution containing 2–4 µL of the final
PCR products at 37 °C for 2–4 h.
2. Add 0.5 µL of DNase attached to the kit to the solution, and
then further incubate the mixture at 37 °C for 15 min.
3. Purify the transcribed mRNA with the RNeasy MinElute
Cleanup Kit. To concentrate mRNA sufficiently, use 14 μL of
nuclease-free water to elute mRNA in the final step. Typically,
approx. 50 μg of mRNA can be obtained.
4. Quantify the purified mRNA with a UV spectrometer and
adjust the concentration to 3 pmol/µL with nuclease-free
water.
5. Aliquot the mRNA into smaller volumes (e.g., 3.4 μL for three
times’ translation) and store them at less than −80 °C until use.
3.2.4 Luciferase Assay To use the Luciferase Assay Reagent prepared as described in
Subheading 2.5 immediately after translation, start to thaw the ali-
quot on ice while protecting it from light right after starting the
translation in Subheading 3.2.3, step 7.
1. Add 100 µL of water to 10 µL of each translation solution to
dilute them 11-fold.
2. Put 5 µL of each diluted solution in a row (12 wells) of a
96-well microplate.
3. Aliquot 1 mL of the Luciferase Assay Reagent into 12 aliquots
(~80 µL) in new PCR tubes.
4. Add 75 µL of the reagent to each diluted translation solution
in the plate by using a 12-channel pipette, and mix them well
by pipetting three times.
5. Immediately after mixing, measure the luminescence values of
the mixtures with a luminometer.
4 Notes
20. It should be noted that the ribosome tends to skip the start
codon that is too near to the 5′ terminus. To avoid this prob-
lem, a mimic start codon can also be inserted between regula-
tors or just before the IRES. The chosen IRES may originally
have a start codon(s) available as the mimic codon(s).
Incidentally, the PSIV-IRES has two mimic genes in itself.
Moreover, IRES-mediated translation via the PSIV-IRES can
start at a non-AUG codon, so that 5′ terminus-mediated
translation of the reporter gene should be completely sup-
pressed even if the ribosome skips the mimic start codons or
remains on mRNA after translation of the mimic genes. In this
case, the start codon of reporter gene must absolutely be
deleted (see Note 18).
21. If the inhibition effect is inadequate, strengthen the stem of
the 5′-SL by extending the length, or insert a mimic start
codon(s).
22. In the case of the PSIV-IRES, an aIRES to a 5′ side of the
pseudoknot III (PK-III) is the best [7].
23. In the latter case, aIRESs with the 3′ side shortened (or prefer-
ably replaced with the same length of nucleotides uninvolved
with the aIRES/IRES duplex) should be investigated.
Incidentally, for the PSIV-IRES, the former and the latter are
8 nt and 4 nt, respectively [7].
24. If the restoration efficiency is much lower, longer aaIRES may
work well, but a too-long aaIRES would lead to lower ON/
OFF switching efficiencies.
25. In terms of the PSIV-IRES, an 8-mer aaIRES for the comple-
mentary 8-mer aIRES shows an 80 % recovery of IRES-
mediated translation.
26. If an aptamer other than the theophylline aptamer is used, it
must be minimized beforehand, because it is required that the
ligand–aptamer interaction induces a release of the MS from
the aptamer.
27. Longer ones inhibit IRES-mediated translation even when the
aaIRES releases the MS and hybridizes to the aIRES in the
ON state, whereas shorter ones cannot prevent aaIRES/
aIRES duplex formation even in the OFF state.
28. If this loop length is very short (less than 3 or 4 nt), some
bases can be inserted between the MS and the aaIRES to sta-
bilize the stem-loop structure.
29. Use the maximum concentration of the ligand that does not
affect IRES-mediated translation, which should be investi-
gated beforehand with the regulator-free optimal mRNA
(see Subheading 3.1.3). To examine the ligand dependency,
use various concentrations of the ligand.
180 Atsushi Ogawa
Acknowledgments
References
1. Winkler W, Nahvi A, Breaker RR (2002) 9. Madin K, Sawasaki T, Ogasawara T, Endo Y
Thiamine derivatives bind messenger RNAs (2000) A highly efficient and robust cell-free
directly to regulate bacterial gene expression. protein synthesis system prepared from wheat
Nature 419:952–956 embryos: plants apparently contain a suicide
2. Mironov AS, Gusarov I, Rafikov R et al (2002) system directed at ribosomes. Proc Natl Acad
Sensing small molecules by nascent RNA: a Sci U S A 97:559–564
mechanism to control transcription in bacteria. 10. Sasaki J, Nakashima N (1999) Translation initia-
Cell 111:747–756 tion at the CUU codon in mediated by the
3. Blount KF, Breaker RR (2006) Riboswitches as internal ribosome entry site of an insect picorna-
antibacterial drug targets. Nat Biotechnol like virus in vitro. J Virol 73:1219–1226
24:1558–1564 11. Shibuya N, Nishiyama T, Kanamori Y et al
4. Nudler E, Mironov AS (2004) The riboswitch (2003) Conditional rather than absolute
control of bacterial metabolism. Trends requirements of the capsid coding sequence for
Biochem Sci 29:11–17 initiation of methionine-independent transla-
5. Schwalbe H, Buck J, Fürtig B et al (2007) tion in Plautia stali intestine virus. J Virol
Structures of RNA switches: insight into 77:12002–12010
molecular recognition and tertiary structure. 12. Pfingsten JS, Costantino DA, Kieft JS (2006)
Angew Chem Int Ed 46:1212–1219 Structural basis for ribosome recruitment and
6. Serganov A, Yuan Y-R, Pikovskaya O et al manipulation by a viral IRES RNA. Science
(2004) Structural basis for discriminative regu- 314:1450–1454
lation of gene expression by adenine- and 13. Ogawa A (2009) Biofunction-assisted sensors
guanine-sensing mRNAs. Chem Biol based on a new method for converting apta-
11:1729–1741 zyme activity into reporter protein expression
7. Ogawa A (2011) Rational design of artificial with high efficiency in wheat germ extract.
riboswitches based on ligand-dependent mod- ChemBioChem 10:2465–2468
ulation of internal ribosome entry in wheat 14. Jenison RD, Gill SC, Pardi A, Polisky B (1994)
germ extract and their applications as label-free High-resolution molecular discrimination by
biosensors. RNA 17:478–488 RNA. Science 263:1425–1429
8. Ogawa A (2011) Rational construction of 15. Mathews DH, Disney MD, Childs JL et al
eukaryotic OFF-riboswitches that downregu- (2004) Incorporating chemical modification
late internal ribosome entry site-mediated constraints into a dynamic programming algo-
translation in response to their ligands. Bioorg rithm for prediction of RNA secondary structure.
Med Chem Lett 22:1639–1642 Proc Natl Acad Sci U S A 101:7287–7292
Chapter 13
Abstract
Engineering of translation provides an alternative regulatory layer for controlling transgene expression in
addition to transcriptional regulation. Synthetic mRNA switches that modulate translation of a target gene
of interest in response to an intracellular protein could be a key regulator to construct a genetic circuit.
Insertion of a protein binding RNA sequence in the 5′ UTR of mRNA would allow for the generation of
a protein-responsive RNA switch. Here we describe the design principle of the switch and methods for
tuning and analyzing its translational activity in mammalian cells.
Key words Riboswitch, Mammalian cell, Transfection, Reporter assay, RNA–protein interaction,
RNP, Synthetic biology, Translation, RNA module
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_13, © Springer Science+Business Media New York 2014
183
184 Kei Endo and Hirohide Saito
2 Materials
2.2 Cell Culture 1. A kit for purification of transfection-grade plasmid DNA. In our
and DNA Transfection case, we use Plasmid Midi Kit (QIAGEN).
2. Mammalian cultured cell lines. We use HeLa cells in this
protocol.
3. Dulbecco’s modified Eagle medium (DMEM) supplemented
with 10 % fetal bovine serum and 1 % antibiotic antimycotic
solution (Sigma-Aldrich).
4. Phosphate buffered saline (PBS).
5. 0.25 % Trypsin–EDTA.
6. Equipment for cell counting. We use Countess Automated
Cell Counter.
7. Opti-MEM.
8. Lipofectamine 2000.
2.3 Flow Cytometry 1. Polystyrene tube with 35 μm cell strainer cap (BD Biosciences).
Analysis 2. A flow cytometer, such as BD Accuri C6 (BD Biosciences) and
FACSAria (BD Biosciences).
2.4 Apoptosis Assay Following reagents are also provided as a package (Pacific Blue
AnnexinV/SYTOX AADvanced Apoptosis kit, for flow cytometry).
1. Pacific Blue-conjugated Annexin V.
2. Annexin V binding buffer.
3. SYTOX AADvance.
3 Methods
3.1.1 Switch Vector Figure 1b shows the modular design of a switch vector. The 5′
Expressing OFF Switch UTR of the switch has four specific sites of restriction enzymes.
mRNA Nhe I is placed just downstream of the CMV promoter and Age I
186 Kei Endo and Hirohide Saito
3.1.2 Trigger Vector The level of a transgene transiently expressed from a plasmid varies
Expressing an Input Protein among the transfected cells. Therefore, an indicator for transfec-
tion efficiency must be required to evaluate the activity of a syn-
thetic switch. To monitor the efficiency, a reporter protein
expressed by IRES in the same mRNA as the input protein can be
used as a control (Fig. 1d), because the ratio of gene expression
from two co-transfected plasmids is highly correlated in a popula-
tion (see Note 4). The trigger vector is derived from pIRES
Bicistronic Expression Vector.
3.2 Design and Both the position and the number of the aptamer motifs on the 5′
Tuning of OFF Switch UTR of mRNA are critical elements to control the activity of a
protein-responsive RNA switch in mammalian cells. Shortening
the distance from the 5′ end of mRNA to the aptamer and repeat-
ing the aptamer sequences in the 5′ UTR enhance the activity of
the OFF switch by the input protein [16]. In other words, engi-
neering mRNAs by modifying these features can tune the activity
of the switch composed of the same RNP pair (Fig. 2). This section
provides a strategy for tuning protein-responsive switches.
3.2.1 Spacing Between An aptamer located closer to the 5′ terminus of mRNA has more
the RNA Motif and the impact on repressing translation. The activity of the OFF switch
5′ End decreases as the distance of the aptamer from the 5′ end increases
[16]. Thus, spacing them could modify the activity in a continuous
manner. It is notable that a spacer sequence should not include
ATG triplets, because it is going to be inserted upstream of the
ORF. The following protocol describes a procedure for insertion
of a spacer DNA fragment upstream of an aptamer (Nhe I/BamH I)
in the switch vector (Fig. 1b). It is also possible to reduce the dis-
tance via PCR-based deletion methods [17, 18].
188 Kei Endo and Hirohide Saito
Fig. 2 Tuning the activity of the switch by modification of the copy number and
the position of the aptamer. (a) Schematic illustration of tuning strategy.
Increasing the distance of the aptamer from the 5′ end (l) and the number of the
aptamer (n) in the switch continuously decrease and discretely increase the
activity of the switch, respectively. (b) Tuning of the L7Ae-responsive switch
composed of a K-loop RNA motif (Kl) as an aptamer. Shown is the translational
efficiency (see also Subheading 3.3.4) of switches that contains one to four
times repeated Kl, the first one of which was placed at 18th, 67th, 120th, and
164th nucleotides (nt; l) from the 5′ end
3.2.2 Multiplying Tandem insertion of multiple RNA motifs into the 5′ UTR of
the RNA Motifs mRNA increases the activity of the OFF switch in a step-wise man-
ner. A DNA fragment, which contains multiple motifs, can be used
to replace an aptamer in a switch vector. Alternatively, it is possible
Protein-Responsive mRNA Switch 189
3.3.2 Cotransfection of 1. Mix 120 ng of a switch vector and 600 ng of a trigger vector
Plasmids into HeLa Cells in 60 μL of Opti-MEM.
2. Add 1.2 μL of Lipofectamine 2000 to 58.8 μL of
Opti-MEM.
3. Mix the two solutions above and incubate at room tempera-
ture for 20 min.
4. Apply 100 μL of the mixture per well and gently shake the
plate to mix.
5. Incubate cells at 37 °C for 4 h.
6. Replace the medium by aspiration and add 500 μL of fresh
DMEM containing serum and antibiotics.
a b c
Fig. 3 Plots from flow cytometry analysis. (a) Raw data of cells expressing either EGFP (upper; reporter) or
DsRed (lower; control) used for the fluorescence compensation (gray). Mock (red/dark gray) indicates untrans-
fected cells. Fluorescence of EGFP and DsRed is detected in GFP and PerCP channel, respectively. Fitting curve
and obtained coefficient are also shown. (b) Plots of compensated signals from the same experiments as
shown in (a). (c) Plots of cells expressing L7Ae-responsive RNA switch that outputs EGFP. DsRed was used as
a control. The upper and lower panel shows the output in the absence (w/o input) and presence (w/input) of
L7Ae, respectively
[PerCP - A ] = coef 1 [EGFP ] + [DsRed ] + [auto ]PerCP -A
Protein-Responsive mRNA Switch 191
[GFP - A ] = [EGFP ] + coef 2 [DsRed ] + [auto ]GFP - A
4. Invert the spillover matrix (S) to obtain the compensation
matrix (C).
æ 1 coef 2 ö -1
Here, S = ç ÷ ,C = S
è coef 1 1 ø
From the two equations in steps 2 and 3,
æ [GFP - A ] ö æ [EGFP ] ö
çç ÷÷ = S çç ÷÷
è [PerCP - A ] ø è [ DsRed ] ø
æ [EGFP ] ö æ [GFP - A ] ö
Therefore, ç
ç [ DsRed ] ÷÷ = C çç [PerCP - A ] ÷÷
è ø è ø
5. Apply the compensation matrix (C) to all the other data to
obtain compensated signals from the reporter (EGFP) and the
control (DsRed).
3.3.5 Calculation of Cross comparing the activity of the switch with different configura-
Translational Efficiency tions or in different conditions requires some representative values.
It can be considered as the ratio of the translational efficiency of the
riboswitch in the presence and in the absence of the input protein.
When calculating the ratio, effects of the background fluorescence
and the efficiency of transfection should be taken into consider-
ation. Here it is shown one definition that represents translational
efficiency of the riboswitch as the average of the output from spe-
cific cells determined by the control signal (Fig. 4).
1. Collect cells expressing a specific range of the control fluores-
cent protein (e.g., 1,000 ± 100 a.u. of DsRed) (Fig. 4a).
2. Calculate the mean level of the reporter protein in the col-
lected cells.
3. Divide the mean value from cells in the presence of the input
protein by that in the absence of the input to obtain transla-
tional efficiency (Fig. 4b). Confirm that the choice of the spe-
cific range selected for the above steps does not significantly
change translational efficiency by varying the specific range as
shown in Fig. 4b.
a b
Fig. 5 Cell death induced by a switch. (a) Schematic illustration of the system.
Bcl-xL protein produced from the switch blocks apoptosis induced by Bim-EL. In
the presence of input protein, the switch represses Bcl-xL expression and
induces apoptosis. (b) Induction of apoptosis was observed in an input protein
specific manner
Protein-Responsive mRNA Switch 193
induction of cell death [19]. In this case the switch which outputs
to an anti-apoptotic gene, Bcl-xL, is transferred into mammalian
cells, together with an apoptosis inducible gene, Bim-EL. Bcl-xL
interacts with Bim-EL and therefore blocks the function of
Bim-EL and cell death; in the presence of the input protein, the
expression of Bcl-xL is repressed and the function of Bim-EL
recovers, resulting in the induction of apoptosis.
A procedure for evaluating the induction of apoptosis in trans-
fected cells is as follows.
1. Prepare a switch vector with Bcl-xL as the output instead of
the reporter fluorescent protein described previously.
2. Cotransfect both the switch and trigger vectors as well as an
additional vector, which expresses Bim-EL (see Note 8).
3. Change the medium 4 h after the transfection.
4. Transfer media including detached cells from the well into a
1.5 mL-microcentrifuge tube 24 h after the transfection.
5. Add 100 μL of Trypsin–EDTA and incubate at 37 °C for
3 min.
6. Add 200 μL of medium and gently rock the dish.
7. Transfer the cell suspension to the same tube as in step 4.
8. Spin the suspension at 200 × g for 5 min at room temperature.
9. Aspirate supernatant and add 500 μL of PBS.
10. Spin again at 200 × g for 5 min at room temperature and aspi-
rate supernatant again.
11. Suspend cells with 53 μL of premixed staining reagent (Pacific
Blue Annexin V, 2.5 μL; SYTOX AADvance, 0.5 μL; binding
buffer, 50 μL).
12. Incubate samples in the dark for 30 min at room temperature.
13. Add 200 μL of binding buffer and keep on ice until analysis.
14. Perform the flow cytometry analysis.
15. Gate out untransfected cells based on the fluorescence of the
control protein.
16. Evaluate the ratio of Annexin V positive cells in populations
(Fig. 5b).
a b
Fig. 6 Simultaneous and specific control of two outputs by a single input. (a) Schematic illustration of the
system. (b) Translational efficiencies of co-transfected two distinct L7Ae-responsive switches. dKt defective
sequence of Kt
one switch did not affect the translational efficiency of the other
switch in the same cell (Fig. 6) [16].
1. Prepare another switch vector composed of the same aptamer
but a different reporter protein.
2. Transfect two switch vectors together with a trigger vector
(Fig. 6a).
3. Perform flow cytometry analysis with three fluorescent pro-
teins (two reporter and one control).
4. Evaluate translational efficiency of each switch (Fig. 6b).
Fig. 7 Autofeedback regulation composed of protein-responsive switch. (a) Genetic configuration of the auto-
regulation. A GOI in a switch vector is fused with an input protein from a trigger vector. (b) Schematic illustra-
tion of translational negative feedback loop
4 Notes
References
1. Isaacs FJ, Dwyer DJ, Collins JJ (2006) RNA 12. Paraskeva E, Atzberger A, Hentze MW (1998)
synthetic biology. Nat Biotechnol 24: A translational repression assay procedure
545–554 (TRAP) for RNA-protein interactions in vivo.
2. Wieland M, Fussenegger M (2010) Ligand- Proc Natl Acad Sci U S A 95:951–956
dependent regulatory RNA parts for synthetic 13. Nie M, Htun H (2006) Different modes and
biology in eukaryotes. Curr Opin Biotechnol potencies of translational repression by
21:760–765 sequence-specific RNA-protein interaction at
3. Chang AL, Wolf JJ, Smolke CD (2012) the 5′-UTR. Nucleic Acids Res 34:5528–5540
Synthetic RNA switches as a tool for temporal 14. Saito H, Kobayashi T, Hara T et al (2010)
and spatial control over gene expression. Curr Synthetic translational regulation by an L7Ae-
Opin Biotechnol 23:679–688 kink-turn RNP switch. Nat Chem Biol 6:71–78
4. Nandagopal N, Elowitz MB (2011) Synthetic 15. Werstuck G, Green MR (1998) Controlling
biology: integrated gene circuits. Science gene expression in living cells through small mol-
333:1244–1248 ecule-RNA interactions. Science 282:296–298
5. Ruder WC, Lu T, Collins JJ (2011) Synthetic 16. Endo K, Stapleton JA, Hayashi K et al (2013)
biology moving into the clinic. Science Quantitative and simultaneous translational
333:1248–1252 control of distinct mammalian mRNAs. Nucleic
6. Wieland M, Fussenegger M (2012) Engineering Acids Res. 41:e135
molecular circuits using synthetic biology in 17. Imai Y, Matsushima Y, Sugimura T et al (1991)
mammalian cells. Annu Rev Chem Biomol Eng A simple and rapid method for generating a
3:209–234 deletion by PCR. Nucleic Acids Res 19:2785
7. Ausländer S, Ausländer D, Müller M et al 18. Hansson MD, Rzeznicka K, Rosenbäck M et al
(2012) Programmable single-cell mammalian (2008) PCR-mediated deletion of plasmid
biocomputers. Nature 487:123–127 DNA. Anal Biochem 375:373–375
8. Jackson RJ, Hellen CU, Pestova TV (2010) 19. Saito H, Fujita Y, Kashida S et al (2011)
The mechanism of eukaryotic translation initia- Synthetic human cell fate regulation by protein-
tion and principles of its regulation. Nat Rev driven RNA switches. Nat Commun 2:160
Mol Cell Biol 11:113–127 20. Smith AM, Fuchs RT, Grundy FJ et al (2010)
9. Sonenberg N, Hinnebusch AG (2009) Riboswitch RNAs: regulation of gene expres-
Regulation of translation initiation in eukary- sion by direct monitoring of a physiological
otes: mechanisms and biological targets. Cell signal. RNA Biol 7:104–110
136:731–745 21. Eisenstein RS (2000) Iron regulatory proteins
10. Hentze MW, Kühn LC (1996) Molecular con- and the molecular control of mammalian iron
trol of vertebrate iron metabolism: mRNA- metabolism. Annu Rev Nutr 20:627–662
based regulatory circuits operated by iron, 22. Stapleton JA, Endo K, Fujita Y et al (2012)
nitric oxide, and oxidative stress. Proc Natl Feedback control of protein expression in
Acad Sci U S A 93:8175–8182 mammalian cells by tunable synthetic transla-
11. Stripecke R, Oliveira CC, McCarthy JE et al tional inhibition. ACS Synth Biol 1:83–88
(1994) Proteins binding to 5′ untranslated 23. Suzuki Y, Ishihara D, Sasaki M et al (2000)
region sites: a general mechanism for transla- Statistical analysis of the 5′ untranslated region
tional regulation of mRNAs in human and of human mRNA using ‘Oligo-Capped’ cDNA
yeast cells. Mol Cell Biol 14:5898–5909 libraries. Genomics 64:286–297
Chapter 14
Abstract
Regulation of gene expression by short oligonucleotides (antisense oligonucleotides), which can modulate
RNA structures and inhibit subsequent associations with the translation machinery, is a potential approach
for gene therapy. This chapter describes an alternative antisense strategy using guanine-tethered antisense
oligonucleotides (G-ASs) to introduce a DNA–RNA heteroquadruplex structure at a designated sequence
on RNA targets. The feasibility of using G-ASs to modulate RNA conformation may allow control of RNA
function by inducing biologically important quadruplex structures. This approach to manipulate quadru-
plex structures using G-ASs may expand the strategies for regulating RNA structures and the functions of
short oligonucleotide riboregulators.
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_14, © Springer Science+Business Media New York 2014
197
198 Masaki Hagihara
Fig. 1 Illustrations of the induced hybrid guanine quadruplex structure on mRNA templates by guanine-tethered
antisense oligonucleotides. A guanine-tethered antisense (G-AS, shown in gray ) hybridizes to a target RNA,
and contiguous guanines in both the RNA and the G-AS then associate to form a quadruplex. The drawing
shown in (a) and (c) show a 1:2 split G-quadruplex mode for the number of GGG sequences, and the drawing
shown in (b) uses a 2:2 split G-quadruplex mode
a metal ion, such as potassium and sodium, inside the tetrad [5, 6].
Recent reports have shown that quadruplex formation of mRNAs
plays a significant role in biological processes, including translation
and translocation [7, 8]. Thus, controlled introduction of quadru-
plex structures into mRNAs may provide opportunities for regula-
tion of biological processes.
Antisense oligonucleotides are relatively short single-stranded
DNAs designed to abrogate protein function by hybridizing
directly with oligonucleotides to exposed regions of the targeted
mRNA [9, 10]. A recently reported novel antisense strategy using
guanine-tethered antisense (G-AS) oligonucleotides confers an
aberrant DNA–RNA heteroquadruplex structure [11]. G-ASs
comprise two functionally independent domains; one is an anti-
sense domain that binds specifically to the target RNA in the same
way as conventional antisense oligonucleotides, and the other
involves contiguous guanine runs tethered at the 5′-end of the
antisense domain that is designed to assemble into heteroquadru-
plex structures together with a contiguous guanine run in the tar-
get RNAs (Fig. 1). The key concept of G-ASs is based on the
cooperative formation of DNA–RNA heteroquadruplex structures
with the assistance of hybridization of antisense sequences. The
combination of antisense recognition and self-assembly of contigu-
ous guanines affords predictable and sequence-specific DNA–RNA
heteroquadruplex structures, which effectively induce structural
changes in RNA templates. The induced quadruplex structures
produced effectively inhibit reverse transcription on the native
RNA sequences.
A reverse transcriptase-mediated enzymatic analysis (RTase
stop assay) [12] together with circular dichroism (CD) spectros-
copy [13, 14] and UV thermal melting experiments [15–17] using
the model assembly of RNA and G-AS oligonucleotides provides
information about the induced guanine quadruplex structures on
RNA templates in association with G-ASs. Detailed investigation
of DNA–RNA hybrid heteroquadruplex formation reveals that at
least one GGG sequence in the target RNA is the minimal sequence
for the introduction of a stable heteroquadruplex by G-ASs. This
feasible and effective approach to controlling RNA structures by
Guanine-Tethered Antisense Riboregulators 199
2 Materials
3 Methods
Fig. 2 Guanine-tethered antisense sequences inhibit reverse transcription of an RNA template bearing two
runs of the GGG sequence. An RNA–AS heteroduplex sequence is shown in the figure. The bold line indicates
the AS hybridization site. The lane markers U, G, C, and A indicate the bases on the template RNA. A partial
genomic RNA sequence from the central polypurine tract of HIV-1 (nucleotides 4581–4756), which is involved
in nuclear import of HIV-1, was chosen as the target sequence
Fig. 3 Biophysical properties of a model assembly of RNA–DNA hybrids modified with runs of contiguous
guanines. (a) Oligonucleotide sequences used in these analyses. Guanine-tethered sequences are shown in
italic letters. (b) CD spectra of rnaG0 and dnaG0 (black line) and rnaG2 and dnaG2 (dotted line). (c, d) UV-melting
profile of DNA–RNA hybrids of rnaG2 and dnaG2 measured at (c) 260 nm and (d) 295 nm
4 Notes
Acknowledgment
References
1. Roth A, Breaker RR (2009) The structural and transcriptase. J Am Chem Soc
functional diversity of metabolite-binding ribo- 132:11171–11178
switches. Annu Rev Biochem 78:305–334 12. Hagihara M, Yoneda K, Yabuuchi H et al
2. Serganov A, Patel DJ (2007) Ribozymes, ribo- (2010) A reverse transcriptase stop assay
switches and beyond: regulation of gene revealed diverse quadruplex formations in
expression without proteins. Nat Rev Genet UTRs in mRNA. Bioorg Med Chem Lett
8:776–790 20:2350–2353
3. Mandal M, Breaker RR (2004) Gene regula- 13. Maurizot JC (2000) Circular dichroism of
tion by riboswitches. Nat Rev Mol Cell Biol nucleic acids: nonclassical conformations and
5:451–463 modified oligonucleotides. In: Berova N,
4. Link KH, Breaker RR (2009) Engineering Nakanishi K, Woody RW (eds) Circular dichro-
ligand-responsive gene-control elements: les- ism: principles and applications, 2nd edn.
sons learned from natural riboswitches. Gene Wiley-VCH, New York, pp 719–739
Ther 16:1189–1201 14. Porumb H, Monnot M, Fermandjian S (2002)
5. Burge S, Parkinson GN, Hazel P et al (2006) Circular dichroism signatures of features simul-
Quadruplex DNA: sequence, topology and taneously present in structured guanine-rich
structure. Nucleic Acids Res 34:5402–5415 oligonucleotides: a combined spectroscopic
6. Lane AN, Chaires JB, Gray RD et al (2008) and electrophoretic approach. Electrophoresis
Stability and kinetics of G-quadruplex struc- 23:1013–1020
tures. Nucleic Acids Res 36:5482–5515 15. Sun XG, Cao EH, He YJ et al (1999)
7. Kumari S, Bugaut A, Huppert JL et al (2007) Spectroscopic comparison of different DNA
An RNA G-quadruplex in the 5′ UTR of the structures formed by oligonucleotides.
NRAS proto-oncogene modulates translation. J Biomol Struct Dyn 16:863–872
Nat Chem Biol 3:218–221 16. Mergny JL, Phan AT, Lacroix L (1998)
8. Arora A, Dutkiewicz M, Scaria V et al (2008) Following G-quartet formation by
Inhibition of translation in living eukaryotic UV-spectroscopy. FEBS Lett 435:74–78
cells by an RNA G-quadruplex motif. RNA 17. Mergny JL, Lacroix L (2009) UV melting of
14:1–7 G-Quadruplexes. In: Egli M, Herdewijn P,
9. Stein CA, Cheng YC (1993) Antisense oligo- Matusda A, Yogesh S (eds) Curr Protoc Nucleic
nucleotides as therapeutic agents—is the bullet Acid Chem, Wiley-VCH, New York, Chapter
really magical? Science 261:1004–1012 17: Unit 17.1
10. Eckstein F (2007) The versatility of oligonu- 18. Xiao Y, Pavlov V, Niazov T et al (2004)
cleotides as potential therapeutics. Expert Opin Catalytic beacons for the detection of DNA
Biol Ther 7:1021–1034 and telomerase activity. J Am Chem Soc
11. Hagihara M, Yamauchi L, Seo A et al (2010) 126:7430–7431
Antisense-induced Guanine Quadruplexes 19. Deng M, Zhang D, Zhou Y et al (2008) Highly
inhibit reverse transcription by HIV-1 reverse effective colorimetric and visual detection of
Guanine-Tethered Antisense Riboregulators 207
nucleic acids using an asymmetrically split per- and triplexes containing A-T or A-U base pairs.
oxidase DNAzyme. J Am Chem Soc Nucleic Acids Res 14:10071–10090
130:13095–13102 23. Jin R, Gaffney BL, Wang C et al (1992)
20. De Cian A, Guittat L, Kaiser M et al (2007) Thermodynamics and structure of a DNA tet-
Fluorescence-based melting assays for studying raplex: a spectroscopic and calorimetric study
quadruplex ligands. Methods 42:183–195 of the tetramolecular complexes of d(TG3T)
21. Fukada H, Takahashi K (1998) Enthalpy and and d(TG3T2G3T). Proc Natl Acad Sci U S A
heat capacity changes for the proton dissocia- 89:8832–8836
tion of various buffer components in 0.1 M 24. Benz A, Hartig JS (2008) Redesigned tetrads
potassium chloride. Proteins 33:159–166 with altered hydrogen bonding patterns enable
22. Steely HT Jr, Gray DM, Ratliff RL (1986) CD programming of quadruplex topologies. Chem
of homopolymer DNA-RNA hybrid duplexes Commun (Camb) 34:4010–4012
Chapter 15
Abstract
Recently, a number of study have shown the ligand-dependent allosteric ribozymes can be harnessed as
biosensors, high-throughput screening, and agents for the control of gene expression in vivo, called artifi-
cial riboswitches. In this chapter, we describe how in vitro selection can be used to create an allosteric
ribozyme that senses bacterial second messenger cyclic-di-GMP (c-di-GMP). A hammerhead ribozyme
was joined to a natural c-di-GMP class I riboswitch aptamer via communication modules. Both c-di-GMP-
activating and -inhibiting ribozyme can be obtained by this approach.
Key words Allosteric ribozyme, Aptamer, Hammerhead ribozyme, In vitro selection, Cyclic
di-GMP
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_15, © Springer Science+Business Media New York 2014
209
210 Kazuhiro Furukawa et al.
2 Materials
Fig. 1 (a) Constructs used for the selection of allosteric ribozymes responding to c-di-GMP. The ribozyme
consists of a hammerhead ribozyme (HHR) joined to a class I c-di-GMP aptamer via communication module
(CM) consisting of random sequence (N) nucleotides. The three stems that form the HHR are designated I, II,
and III and the three stems that form the aptamer are labeled P1, P2, and P3. An arrowhead identifies the site
of hammerhead-mediated cleavage. (b) Modified construct for the selection of c-di-GMP-activated ribozyme
CGTTTCGNNNNNNNG-TAACCCCGCTACCTACCATG
TCTTCTGGTTTAG-3′. Underlined nucleotides are comple-
mentary each other at their 3′ ends. T7 promoter sequences are
shown in italic.
3. 5× reverse transcription buffer: 250 mM Tris–HCl, pH 8.3 at
23 °C, 375 mM KCl, 15 mM MgCl2.
4. 100 mM dithiothreitol (DTT).
5. 10× deoxynucleoside 5′-triphosphate (dNTP) mix: 2 mM each
of deoxyguanosine 5′-triphosphate (dGTP), deoxyadenosine
5′-triphosphate (dATP), deoxythymidine 5′-triphosphate
(dTTP), deoxycytidine 5′-triphosphate (dCTP).
6. Reverse transcriptase (e.g., SuperScript II, Invitrogen).
7. 10× transcription buffer: 500 mM HEPES, pH 7.5 at 23 °C,
150 mM MgCl2, 20 mM spermidine and 50 mM DTT.
8. 3 M sodium acetate, pH 5.2.
9. 100 % ethanol.
10. 5× nucleoside 5′-triphosphate (NTP) mix: 12.5 mM each of
guanosine 5′-triphosphate (GTP), adenosine 5′-triphos-
phate (ATP), thymidine 5′-triphosphate (TTP), cytidine
5′-triphosphate (CTP).
212 Kazuhiro Furukawa et al.
3 Methods
3.1 General The construct used for the selection of allosteric ribozymes that
Selection Strategy respond to c-di-GMP was created by linking the P1 stem of the
c-di-GMP class I aptamer derived from Vibrio cholera [16] to stem
II of the hammerhead ribozyme via a communication module
(CM) (Fig. 1). The P1 stem of the aptamer was chosen for CM
since the stability of P1 stem is highly involved in the affinity
between the aptamer and the ligand. Also, there is an important
tertiary interaction between stems P2 and P3 [20, 21], so that
using these stems for CM might disrupt key structural contacts
Allosteric Ribozymes for c-di-GMP 213
Fig. 2 The selection scheme for the c-di-GMP-activated allosteric ribozyme. (A)
Two synthetic oligonucleotides are extended with reverse transcriptase to gener-
ate double-strand DNA encoding randomized sequence. (B) Transcription with T7
RNA polymerase. (C) The full-length RNAs (precursor) are purified by PAGE, and
subject to negative selection in the absence of c-di-GMP. The cleavage products
are removed by PAGE. (D) The precursor are subjected to positive selection in the
presence of c-di-GMP. (E) The PAGE-purified cleavage products are reverse tran-
scribed and amplified by PCR
3.4 Selection of Selection of ribozymes that undergo c-di-GMP induction was con-
Allosterically Inducted ducted using identical method as allosterically inhibited ribozymes
Ribozymes (Subheading 3.3), except that c-di-GMP was included in the posi-
tive selection reaction, but was excluded in both the transcription
and the negative selection reactions (see Note 14).
3.5 Reverse 1. Add 10 pmol of the 3′ primer (primer A or A′) to the resus-
Transcription and pended cleaved products.
Polymerase Chain 2. Reverse transcribe the recovered RNAs in 100 µL of 1× reverse
Reaction transcribe buffer supplemented with 10 mM DTT, 200 µM of
the four dNTPs, and 8 U/µL reverse transcriptase.
3. Incubate at 37 °C for 2 h.
4. Prepare PCR mixture by adding 5–10 µL of the reverse tran-
scription reaction and 20–50 pmol each of primers (primers A
and B for allosterically inhibited ribozymes, primers A′ and B′ for
allosterically inducted ribozymes) to a 100 µL PCR reaction
containing 1× PCR buffer, 200 µM each of the four dNTPs, and
1 U/µL Taq DNA polymerase (see Note 15).
5. Amplify full-length double-stranded DNAs using the following
PCR cycling parameters: segment 1: 94 °C for 1 min; segment
2: 94 °C for 30 s, 48 °C for 30 s, 72 °C for 30 s; segment 3:
72 °C for 5 min (see Note 16).
6. Analyze a small aliquot of the PCR reaction on a 2 % agarose gel
to check the amount and size of the PCR products.
7. Use the amplified DNAs as templates to transcribe the RNA
pool for the next round of selection, as in Subheading 3.2,
step 5. Save a portion of the PCR products.
8. Sequence the individual clones after several rounds of selection
(see Notes 17 and 18).
4 Notes
Acknowledgements
References
1. Breaker RR (2004) Natural and engineered 10. Win MN, Smolke CD (2007) A modular and
nucleic acids as tools to explore biology. Nature extensible RNA-based gene-regulatory plat-
432:838–845 form for engineering cellular function. Proc
2. Famulok M (2005) Allosteric aptamers and Natl Acad Sci U S A 104:14283–14288
aptazymes as probes for screening approaches. 11. Ogawa A, Maeda M (2008) An artificial
Annu Rev Microbiol 59:487–517 aptazyme-based riboswitch and its cascading
3. Tang J, Breaker RR (1997) Rational design of system in E. coli. Chembiochem 9:206–209
allosteric ribozymes. Chem Biol 4:453–459 12. Robertson MP, Ellington AD (1999) In vitro
4. Tang J, Breaker RR (1998) Mechanism for selection of an allosteric ribozyme that trans-
allosteric inhibition of an ATP-sensitive ribo- duces analytes to amplicons. Nat Biotechnol
zyme. Nucleic Acid Res 26:4214–4221 17:62–66
5. Soukup GA, Breaker RR (1999) Design of 13. Najafi-Shoushtari SH, Famulok M (2005)
allosteric hammerhead ribozymes activated by Competitive regulation of modular allosteric
ligand-induced structure stabilization. aptazymes by a small molecule and oligonucle-
Structure 7:783–791 otide effector. RNA 11:1514–1520
6. Soukup GA, Breaker RR (1999) Engineering 14. Lee ER, Baker JL, Weinberg Z, Sudarsan N,
precision RNA molecular switches. Proc Natl Breaker RR (2010) An allosteric self-splicing
Acad Sci U S A 96:3584–3589 ribozyme triggered by a bacterial second mes-
7. Koizumi M, Soukup GA, Kerr JNQ, Breaker senger. Science 329:845–848
RR (1999) Allosteric selection of ribozymes 15. Hengge R (2009) Principles of c-di-GMP sig-
that respond to the second messengers cGMP naling in bacteria. Nat Rev Microbiol
and cAMP. Nat Struct Biol 6:1062–1071 7:263–273
8. Gu H, Furukawa K, Breaker RR (2012) 16. Sudarsan N, Lee ER, Weinberg Z, Moy RH,
Engineered allosteric ribozymes that sense the Kim JN, Link KH, Breaker RR (2008)
bacterial second messenger cyclic diguanosyl Riboswitches in eubacteria sense the second
5′-monophosphate. Anal Chem messenger cyclic di-GMP. Science
84:4935–4941 321:411–413
9. Wieland M, Benz A, Klauser B, Hartig JS 17. Mayer G, Famulok M (2006) High-
(2009) Artificial ribozyme switches containing throughput-compatible assay for glmS ribo-
natural riboswitch aptamer domains. Angew switch metabolite dependence. Chembiochem
Chem Int Ed 48:2715–2718 7:602–604
220 Kazuhiro Furukawa et al.
18. Lünse CE, Schmidt MS, Wittman V, Mayer G 21. Kulshina N, Baird NJ, Ferré-D’Amaré AR
(2011) Carba-sugars activate the glmS- (2009) Recognition of the bacterial second
riboswitch of Staphylococcus aureus. ACS Chem messenger cyclic diguanylate by its cognate
Biol 6:675–678 riboswitch. Nat Struct Mol Biol
19. Furukawa K, Gu H, Sudarsan N, Hayakawa Y, 16:1212–1217
Hyodo M, Breaker RR (2012) Identification of 22. Forster AC, Symons RH (1987) Self-
ligand analogues that control c-di-GMP ribo- cleavage of virusoid RNA is performed by
switches. ACS Chem Biol 7:1436–1443 the proposed 55-nucleotide active site. Cell
20. Smith KD, Lipchock SV, Ames TD, Wang J, 50:9–16
Breaker RR, Strobel SA (2009) Structural basis 23. Fedor MJ, Uhlenbeck OC (1992) Kinetics of
of ligand binding by a c-di-GMP riboswitch. intermolecular cleavage by hammerhead ribo-
Nat Struct Mol Biol 16:1218–1223 zymes. Biochemistry 31:12042–12054
Chapter 16
Abstract
Both synthetic biology and metabolic engineering are aided by the development of genetic control parts.
One class of riboswitch parts that has great potential for sensing and regulation of protein levels is aptamer-
coupled ribozymes (aptazymes). These devices are comprised of an aptamer domain selected to bind a
particular ligand, a ribozyme domain, and a communication module that regulates the ribozyme activity
based on the state of the aptamer. We describe a broadly applicable method for coupling a novel, newly
selected aptamer to a ribozyme to generate functional aptazymes via in vitro and in vivo selection. To
illustrate this approach, we describe experimental procedures for selecting aptazymes assembled from
aptamers that bind p-amino-phenylalanine and a hammerhead ribozyme. Because this method uses selec-
tion, it does not rely on sequence-specific design and thus should be generalizable for the generation of in
vivo operational aptazymes that respond to any targeted molecules.
Key words In vitro selection, RNA aptamer, Ribozyme, Aptazyme, Riboswitch, Synthetic biology
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_16, © Springer Science+Business Media New York 2014
221
222 Jonathan A. Goler et al.
Fig. 1 Dual-selection for ligand-dependent self-cleaving aptazymes. (a) To generate aptazymes, a combinato-
rial sequence library is constructed from individual aptamer and ribozyme domains. In the illustration, a
sequence library comprises the pAF-R1-1 aptamer (kd = 19 μm) and S. mansoni hammerhead ribozyme
(kobs = 1.3 min−1), shown in secondary structure schematic, was constructed. 14 fully randomized bases within
stem I of the hammerhead ribozyme are colored and marked N1, N2, and N3. Aptazyme self-cleavage takes
place at the site indicated by the red triangle. PAGE is used to enrich the library pool for aptazymes with fast
kobs+ by isolating RNA that cleaves rapidly in the presence of ligand (dashed box at left) and for aptazymes with
slow kobs− by isolating RNA that does not cleave in the absence of ligand (dashed box at right). The complete
cycle is repeated 4–6 times. (b) Candidate aptazymes from the in vitro selection are subcloned into a genetic
device for aptazyme-regulated expression screening. A subsequent screen for ligand-dependent regulation of
reporter gene expression identifies aptazymes that catalyze self-cleavage reactions inside the cell
2 Materials
3 Methods
3.1 PAGE Gel Assemble the glass with spacers, prepare gel by adding acrylamide,
urea, and TBE (5 mL of 10× TBE, 12 mL of 20 % acrylamide, 24 g
of urea and water to 50 mL); add 50 μL of TEMED and 500 mL
of APS last and swirl to initiate the polymerization reaction
(see Notes 2 and 3).
3.2 Running 1. Pre-run the gels using constant power (18–22 W) for
PAGE Gels 20–30 min to warm the gel a bit, the gel should be approxi-
mately 40–50 °C (see Note 4).
2. After the pre-run, clean the urea from the wells by utilizing a
sterile syringe. Withdraw buffer and expel firmly to flush out
the wells, you should see the urea being blown out.
3. Add loading buffer (95 % formamide, 5 % 0.5 M EDTA) to the
sample in a 1:1 ratio and gently mix.
4. Load the samples into the gel (see Note 5), actuate the power
and run the gel for 1–1.25 h.
5. Once the gel is complete, separate the gel from the glass plates
for UV shadowing using a fluorescent PEI-Cellulose TLC
plate (see Note 6).
Dual Selection of Aptazyme Parts 227
3.3 Stage One: In this illustration, we used the pAF-binding aptamer pAF-R1-1
In Vitro Aptazyme (kd = 30 μM at 2.5 mM Mg2+ [16], which was connected to the
Selection S. mansoni ribozyme via randomized regions as in Link et al. [18]
to construct the starting combinatorial sequence library. Oligos
were as follows (Integrated DNA Technologies): LSW-11F
5′-GCT AA TAC GAC TCA CTA TAG GCG AAA GCC GGC
GCG TCC TGG ATT CCA CNN NNN CAT GTC CCT ACC
ATA CGG GAT TGC CCA GCT TCG GCT GCC ATG CCG
GC, LSW-11Rc: 5′-AGC GCG TTT CGT CCT ATT TGG GAC
TCA TCA GCN NNN TGT ACC NNN NNC GTA GGC CGG
TTA CCG TTT GGC CGG CAT GGC AGC CG and primers
LSW-11Fp: 5′-GCT AAT ACG ACT CAC TAT AGG CGA AAG
CCG GCG CGT CCT GGA TTC CAC and LSW-11Rp: 5′-AGC
GCG TTT CGT CCT ATT TGG G (also see Note 7).
3.3.1 PAGE Purification The DNA oligos are PAGE-purified, primer-extended, and tran-
of Oligo DNA scribed in vitro, as detailed below.
1. First, solubilize the oligos in ultrapure water, add 1:1 2× load-
ing buffer and vortex.
2. Purify oligos on a 10–12 % PAGE gel, running gels as above.
3. Illuminate gels with UV light against a PEI-Cellulose TLC
plate, mark and excise bands with a razor blade.
4. Elute gel slices (Whatman Elutrap System) at 300 V for 30 min
to 2 h into a final volume between 200 and 400 μL (see Note 8).
5. Ethanol precipitate the eluate with 1/10 volume of 3 M KCl
and 2.5 volumes of 95 % ethanol, chill for 10 min on dry ice
(solution should become slightly whiter and more viscous),
spin for 15 min at 24,000 × g in a refrigerated centrifuge. Decant
using a pipette and air dry at room temperature or 37 °C.
6. Quantify DNA using a NanoDrop or other spectrophotometer.
3.3.2 In Vitro Selection 1. Prepare transcription mix according to the materials section
(see Note 9).
2. Add the template and top strand oligos (if needed) to the tran-
scription mix.
3. Run transcription at 20–25 °C for 15 min to 2 h.
228 Jonathan A. Goler et al.
3.4 Stage Two: In After four to six rounds of in vitro aptazyme selection, the apta-
Vivo Aptazyme Screen zyme parts are screened for ligand-dependent self-cleavage in vivo.
1. The enriched, surviving, sequences are PCR amplified using
cloning adapters (For the pAF aptazyme selections these are
Adapter4A.F: 5′-GAT ATC ATC TCT AGA GGC GAA AGC
CGG CGC GTC CTG GAT TCC AC, Adapter4AR: 5′-GAC
CTT CTC GAG AGC GCG TTT CGT CCT ATT TGG G),
and cloned into prRED-T7-ref (Fig. 1), which harbors the
kanamycin (Kan) resistance gene, a T7 promoter, an XbaI-
BglII cloning site, and the gene encoding the dsRedExpress
Red Fluorescent Protein (RFP) [15] (see Note 11).
2. The resulting plasmids are then transformed initially into E. coli
TOP 10 chemically competent cells.
3. For the transformation, add a 2–3 μL of the enriched library
DNA to the cells.
4. Incubate on ice for 10 min.
5. Place aliquots in a 42 °C water bath for 15 s, then back on ice
for 2 min.
6. Incubate aliquots at 37 °C, shaking, for 75 min.
7. Plate cultures on LB-Kan plates and incubate overnight.
8. The next day, pick individual colonies into test tubes, each with
5 mL of LB medium containing 50 μg/mL kanamycin
(LB-Kan).
Dual Selection of Aptazyme Parts 229
Fig. 2 Winner from the example pAF aptazyme selection, W24. Secondary struc-
ture of the minimum free energy fold, as computed with Vienna RNAfold with
default settings, for the best performing pAF aptazyme from the example selec-
tion is shown with number and color coding as in Fig. 1
Dual Selection of Aptazyme Parts 231
a 1.0 b
1.0
0.9
0.8
Fraction Uncleaved RNA
0.7
0.6
0.4
0.5
0.2
0.4
0.0
0.3
0 5 10 15 0 5 10 15
Time (Minutes) Time (Minutes)
Fig. 3 Measuring the kinetics of aptazyme cleavage in vitro. The kinetics of ligand-dependent cleavage are
assayed for aptazymes showing function in vivo, using RNA transcribed in vitro from the isolated sequences.
In this figure, results for two isolates from the pAF aptazyme selections are shown. The fraction of self-cleaved
aptazyme in the presence (open triangles) and absence (solid triangles) of the target ligand is plotted as a
function of time. The data are fitted to a first-order decay curve, overlaid
4 Notes
1.5
1.4
(relative to 0 mM pAF)
Fluorescence change
1.3
1.2
1.1
1.0
0 2 4 6 8 10
pAF concentration (mM)
References
1. Vitreschak AG, Rodionov DA, Mironov AA, 14. Soukup GA, Breaker RR (1999) Engineering
Gelfand MS (2004) Riboswitches: the oldest precision RNA molecular switches. Proc Natl
mechanism for the regulation of gene expres- Acad Sci U S A 96:3584–3589
sion. Trends Genet 20:44–50 15. Carothers JM, Goler JA, Juminaga D, Keasling
2. Stormo GD, Ji Y (2001) Do mRNAs act as JD (2011) Model-driven engineering of RNA
direct sensors of small molecules to control devices to quantitatively program gene expres-
their expression? Proc Natl Acad Sci U S A sion. Science 334:1716–1719
98:9465–9467 16. Carothers JM, Goler JA, Kapoor R, Lara LD,
3. Patel DJ, Suri AK, Jiang F et al (1997) Keasling JD (2010) Selecting RNA aptamers
Structure, recognition and adaptive binding in for synthetic biology: investigating magnesium
RNA aptamer complexes. J Mol Biol 272: dependence and predicting binding affinity.
645–664 Nucleic Acids Res 38:2736–2747
4. Tuerk C (1997) Using the SELEX combinato- 17. Thompson KM, Syrett HA, Knudsen SM,
rial chemistry process to find high affinity Ellington AD (2002) Group I aptazymes as
nucleic acid ligands to target molecules. genetic regulatory switches. BMC Biotechnol
Methods Mol Biol 67:219–230 2:21
5. Klug SJ, Hüttenhofer A, Famulok M (2000) In 18. Link KH, Guo L, Ames TD et al (2007)
vitro selection of RNA aptamers that bind spe- Engineering high-speed allosteric hammerhead
cial elongation factor SelB, a protein with mul- ribozymes. Biol Chem 388:779–786
tiple RNA-binding sites, reveals one major 19. Jenison RD, Gill SC, Pardi A, Polisky B (1994)
interaction domain at the carboxyl terminus. High-resolution molecular discrimination by
RNA 5:1180–1190 RNA. Science 263:1425–1429
6. Lorsch JR, Szostak JW (1994) In vitro selec- 20. Dower WJ, Cwirla SE (1992) Guide to electro-
tion of RNA aptamers specific for cyanocobala- poration and electrofusion. Academic, San Diego
min. Biochemistry 33:973–982 21. Isambert H, Siggia E (2000) Modeling RNA
7. Winkler WC, Breaker RR (2003) Genetic con- folding paths with pseudo-knots: application to
trol by metabolite-binding riboswitches. hepatitis delta virus ribozyme. Proc Natl Acad
Chembiochem 4:1024–1032 Sci U S A 97:6515–6520
8. Nahvi A, Sudarsan N, Ebert MS et al (2002) 22. Xayaphoummine A, Viasnoff V, Harlepp S,
Genetic control by a metabolite binding Isambert H (2007) Encoding folding paths of
mRNA. Chem Biol 9:1043–1049 RNA switches. Nucleic Acids Res 35:614–622
9. Nahvi A, Barrick JE, Breaker RR (2004) 23. Deana A, Celesnik H, Belasco JG (2008) The
Coenzyme B 12 riboswitches are widespread bacterial enzyme RppH triggers messenger
genetic control elements in prokaryotes. RNA degradation by 5-prime pyrophosphate
Nucleic Acids Res 32:143–150 removal. Nature 451:355–358
10. Liu CC, Arkin AP (2010) The case for RNA. 24. Bayer TS, Smolke CD (2005) Programmable
Science 330:1185–1186 ligand-controlled riboregulators of eukaryotic
11. Hall B, Hesselberth JR, Ellington AD (2007) gene expression. Nat Biotechnol 23:337–343
Computational selection of nucleic acid bio- 25. Wieland M, Hartig JS (2008) Improved apta-
sensors via a slip structure model. Biosens zyme design and in vivo screening enable ribo-
Bioelectron 22:1939–1947 switching in bacteria. Angew Chem Int Ed
12. Lynch SA, Desai SK, Sajja HK, Gallivan JP 47:2604–2607
(2007) A high-throughput screen for synthetic 26. Weigand J, Sanchez M, Gunnesch EB et al
riboswitches reveals mechanistic insights into (2008) Screening for engineered neomycin
their function. Chem Biol 14:173–184 riboswitches that control translation initiation.
13. Suess B, Fink B, Berens C et al (2004) A the- RNA 14:89–97
ophylline responsive riboswitch based on helix 27. Hillson NJ, Rosengarten RD, Keasling JD
slipping controls gene expression in vivo. (2011) j5 DNA assembly design automation
Nucleic Acids Res 32:1610–1614 software. ACS Synth Biol 1:14–21
Chapter 17
Abstract
In many synthetic biology applications, modular and easily accessible tools for controlling gene expression
are required. In addition, in vivo biosensors and diagnostic devices will become more important in the
future to allow for noninvasive determination of protein, ion, or small molecule metabolite levels. In recent
years synthetic RNA-based switches have been developed to act as signal transducers to convert a binding
event of a small molecule (input) into a detectable output. Their modular design allows the development
of a variety of molecular switches to be used in biochemical assays or inside living cells. RNA switches
developed by our group are based on the Schistosoma mansoni hammerhead ribozyme, a self-cleaving RNA
sequence that can be inserted into any RNA of interest. Connection to an aptamer sensing a small mole-
cule renders the cleavage reaction ligand-dependent. In the past we have successfully designed and applied
such hammerhead aptazymes for the allosteric control of both bacterial and eukaryotic gene expression by
affecting transcription elongation, translation initiation, or mRNA stability. In order to yield functional
switches optimization of the connecting sequence between the aptamer and the HHR needs to be carried
out. We have therefore developed an in vivo screening protocol detailed in this chapter that allows the
identification of functional aptazymes in bacteria.
Key words Riboswitch, Allosteric ribozyme, Biosensor, Hammerhead aptazyme, In vivo screening,
Aptamer, Gene expression, Regulation
1 Introduction
Aptamers are short nucleic acids motifs which can specifically rec-
ognize small molecule interaction partners or even ions with high
specificity and affinity [1]. They were originally obtained by sys-
tematic evolution of ligands by exponential enrichment (SELEX)
[2, 3]. However, Breaker and coworkers later proofed the exis-
tence of RNA aptamers in the 5′ untranslated regions (5′ UTR) of
many bacterial messenger RNAs (mRNAs). There they form selec-
tive binding pockets for a respective metabolite and serve as sen-
sors of intracellular metabolite concentrations in regulatory RNA
elements called riboswitches [4–6]. Metabolite binding to the
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_17, © Springer Science+Business Media New York 2014
237
238 Charlotte Rehm and Jörg S. Hartig
Fig. 1 The hammerhead ribozyme as catalytically active domain. (a) Reaction mechanism of the intramolecular
strand-cleavage reaction by in-line nucleophilic attack of the 2′ hydroxyl group. (b) Example of a hammerhead
aptazyme inserted into the 5′ untranslated region of a bacterial mRNA. The RBS is incorporated into stem I and
blocked from access by the ribosome, an aptamer is attached at stem III. Stabilizing tertiary interactions
between stem I and II are shown. The cleavage site is marked by the arrowhead. Variability of the randomiza-
tion site allows screening for activating as well as inactivating aptazymes
In Vivo Screening for Aptazyme-Based Bacterial Riboswitches 239
2 Materials
2.2 Cell Culture and 1. 96-deep-well plate and 96-well plate incubator (e.g., Heidolph
Screening Inkubator 1000 and Titramax 1000) (see Note 3).
2. Air-permeable adhesive seals for 96-well-plates.
3. 1,000× carbenicillin stock solution (100 mg/mL): Dissolve
1 g carbenicillin in 10 mL 50 % (v/v) ethanol. Can be stored
at −20 °C for several weeks.
4. Electro-competent E. coli (e.g., E. coli BL21 (DE3) gold,
Stratagene) (see Note 4). 80 μL aliquots frozen at −80 °C.
5. Electroporator (e.g., Eppendorf Electroporator 2510) and
electroporation cuvettes (e.g., Electroporation cuvettes,
0.1 cm (Bio-Rad)).
6. Fluorescence plate reader (e.g., TECAN M200).
7. LB-Carb-medium: For LB (Lennox) dissolve 10 g tryptone,
5 g yeast extract, 5 g NaCl in 1 L H2O, adjust to pH 7.0, if
necessary, and autoclave. Supplement with 1 mg/L carbenicil-
lin before use. Can be stored at 4 °C for up to 4 weeks.
8. LB-Carb-agar plates: Dissolve 10 g tryptone, 5 g yeast extract,
5 g NaCl, and 10 g agar-agar in 1 L H2O and autoclave. Let
cool until hand-warm, then supplement with 1 mg/L carbeni-
cillin. Pour plates in 15 cm petri dishes. Plates can be stored at
4 °C for up to 6 weeks.
9. Kit for plasmid isolation (e.g., Zyppy Plasmid Miniprep Kit).
10. SOC medium: Dissolve 20 g tryptone, 5 g yeast extract, 0.6 g
NaCl, 0.2 g KCl in 900 mL H2O. Adjust pH to 6.8–7.0 using
NaOH. Adjust volume to 960 mL and autoclave. Add 10 mL 1 M
MgCl2, 10 mL 1 M MgSO4, and 20 mL 1 M glucose from sterile
filtered stocks. Store 1 mL aliquots at −20 °C for several months.
11. Toothpicks sterilized by autoclaving.
12. Flat, transparent 96-well plates.
3 Methods
3.1 Construction of New aptamer sequences are incorporated into the HHR using
Randomized whole plasmid PCR as done for site-directed mutagenesis (Fig. 2).
Riboswitch Libraries Randomization of the connecting sequence is achieved by using
primers with unbiased random positions generated during solid
phase DNA synthesis using a 1:1:1:1 mixtures of nucleoside phos-
phoramidites. For successful ligation of the PCR product phos-
phorylation of one primer at the 5′ end is required. We recommend
purification of the long primers (≥45 nts) by denaturing PAGE, as
described by Sambrook and Russel in Molecular Cloning (see:
Preparation of Denaturing Polyacrylamide Gels and Purification of
Synthetic Oligonucleotides by Polyacrylamide Gel Electrophoresis).
In Vivo Screening for Aptazyme-Based Bacterial Riboswitches 243
Fig. 2 Library construction. Whole plasmid PCR is used for incorporation of new aptamer sequences using
primers carrying randomized sequences that will constitute the connecting sequence. After PCR the template
plasmid is degraded by DpnI digest. The purified PCR product is ligated yielding a library of randomized
riboswitches
2. Use the following conditions for the PCR with Phusion Hot
Start II High-Fidelity DNA polymerase (see Note 5).
3. Pool the PCR reaction mix into one 1.5 mL reaction tube. For
ethanol precipitation add 5 μL of 3 M Na-acetate, pH 5.2,
followed by 165 μL 99 % ethanol and mix by inverting.
4. Place samples at −80 °C for 20 min or −20 °C for a minimum
of 2 h or overnight.
5. Centrifuge samples in a tabletop centrifuge at 12,000 × g for
15 min at room temperature. Discard supernatant and wash
once with 70 % ethanol. Resuspend the pellet in 44 μL H2O.
6. For digestion of the template vector add 5 μL NEB buffer 4
and 1 μL DpnI and mix thoroughly. Incubate at 37 °C for
50 min followed by 10 min incubation at 80 °C for heat inac-
tivation of the enzyme.
7. In the meantime, pour a 0.8 % (w/v) TBE-agarose gel and let
it solidify. Once the sample is ready, add 10 μL of 6× agarose
gel loading buffer and mix. Load the sample in a long well and
load 2.5 μL DNA size standard in a small well to run along-
side. Run the gel at 10 V/cm for 75–90 min.
8. After the run, transfer the gel into ethidium bromide staining
solution for 10 min, then place the gel in destaining solution
for 10 min for improving contrast. As exposure to UV light
may introduce mutations to the DNA, cut the gel vertically
excising the marker and a small part of the band with your
sample. Visualize the DNA bands over a UV light table.
9. Excise the appropriate band in the correct size, piece the gel
back together and excise the corresponding band in the gel
that has not been exposed to UV light and purify the DNA
fragment using a DNA gel extraction kit according to manu-
facturer’s protocol.
10. Determine the DNA concentration by measuring A260.
11. For ligation of the PCR product combine 50 ng of the puri-
fied PCR product in 9 μL of water with 10 μL 2× Quick
Ligation Reaction buffer and add 1 μL Quick Ligase, mix well,
and briefly centrifuge samples. Incubate at 25 °C for 15 min.
In Vivo Screening for Aptazyme-Based Bacterial Riboswitches 245
Fig. 3 Screening for functional riboswitches. The plasmid library of randomized riboswitches is transformed
into bacteria. Individual clones are picked to construct mutant libraries, which are screened for eGFP expres-
sion in the presence (white) or absence (dark grey) of the aptamer ligand. Controls for background fluores-
cence correction (B), positive control (+) and negative control (−) are included in the screening. Identified hits
are further characterized
4 Notes
References
1. Liu J, Cao Z, Lu Y (2009) Functional nucleic damental biochemical pathways in Bacillus sub-
acid sensors. Chem Rev 109:1948–1998 tilis and other bacteria. Cell 113:577–586
2. Tuerk C, Gold L (1990) Systematic evolution 7. Vinkenborg JL, Karnowski N, Famulok M
of ligands by exponential enrichment: RNA (2011) Aptamers for allosteric regulation. Nat
ligands to bacteriophage T4 DNA polymerase. Chem Biol 7:519–527
Science 249:505–510 8. Paige JS, Nguyen-Duc T, Song W, Jaffrey SR
3. Ellington AD, Szostak JW (1990) In vitro (2012) Fluorescence imaging of cellular
selection of RNA molecules that bind specific metabolites with RNA. Science 335:1194
ligands. Nature 346:818–822 9. Breaker RR (2002) Engineered allosteric ribo-
4. Winkler W, Nahvi A, Breaker RR (2002) zymes as biosensor components. Curr Opin
Thiamine derivatives bind messenger RNAs Biotechnol 13:31–39
directly to regulate bacterial gene expression. 10. Blount KF, Uhlenbeck OC (2002) The ham-
Nature 419:952–956 merhead ribozyme. Biochem Soc Trans
5. Winkler WC, Nahvi A, Sudarsan N, Barrick JE, 30:1119–1122
Breaker RR (2003) An mRNA structure that 11. Fedor MJ (2009) Comparative enzymology
controls gene expression by binding and structural biology of RNA self-cleavage.
S-adenosylmethionine. Nat Struct Biol Annu Rev Biophys 38:271–299
10:701–707 12. Khvorova A, Lescoute A, Westhof E, Jayasena
6. Mandal M, Boese B, Barrick JE, Winkler WC, SD (2003) Sequence elements outside the
Breaker RR (2003) Riboswitches control fun- hammerhead ribozyme catalytic core enable
In Vivo Screening for Aptazyme-Based Bacterial Riboswitches 249
intracellular activity. Nat Struct Biol switch for controlling mammalian gene expres-
10:708–712 sion. Mol BioSyst 6:807–814
13. Tang J, Breaker RR (1997) Rational design of 22. Ketzer P, Haas SF, Engelhardt S, Hartig JS,
allosteric ribozymes. Chem Biol 4:453–459 Nettelbeck DM (2012) Synthetic riboswitches
14. Wieland M, Hartig JS (2008) Improved apta- for external regulation of genes transferred by
zyme design and in vivo screening enable ribo- replication-deficient and oncolytic adenovi-
switching in bacteria. Angew Chem ruses. Nucleic Acids Res 40:e167
47:2604–2607 23. Berschneider B, Wieland M, Rubini M, Hartig
15. Wieland M, Benz A, Klauser B, Hartig JS JS (2009) Small-molecule-dependent regula-
(2009) Artificial ribozyme switches containing tion of transfer RNA in bacteria. Angew Chem
natural riboswitch aptamer domains. Angew 48:7564–7567
Chem 48:2715–2718 24. Wieland M, Berschneider B, Erlacher MD,
16. Ogawa A, Maeda M (2008) An artificial Hartig JS (2010) Aptazyme-mediated regula-
aptazyme-based riboswitch and its cascading tion of 16S ribosomal RNA. Chem Biol
system in E. coli. Chembiochem 9:206–209 17:236–242
17. Win MN, Smolke CD (2007) A modular and 25. Kumar D, Annna CI, Yokobayashi Y (2009)
extensible RNA-based gene-regulatory plat- Conditional RNA interference mediated by
form for engineering cellular function. Proc allosteric ribozyme. J Am Chem Soc
Natl Acad Sci U S A 104:14283–14288 131:13906–13907
18. Win MN, Smolke CD (2008) Higher-order 26. Saragliadis A, Krajewski SS, Rehm C,
cellular information processing with synthetic Narberhaus F, Hartig JS (2013) Thermozymes:
RNA devices. Science 322:456–460 Synthetic RNA thermometers based on ribo-
19. Wieland M, Gfell M, Hartig JS (2009) Expanded zyme activity. RNA Biol 10(6):1010–6
hammerhead ribozymes containing addressable 27. Klauser B, Hartig JS (2013) An engineered
three-way junctions. RNA 15:968–976 small RNA-mediated genetic switch based on a
20. Klauser B, Saragliadis A, Auslander S, Wieland ribozyme expression platform. Nucleic Acids
M, Berthold MR, Hartig JS (2012) Post- Res 41(10):5542–52
transcriptional Boolean computation by com- 28. Soukup GA, Breaker RR (1999) Engineering
bining aptazymes controlling mRNA precision RNA molecular switches. Proc Natl
translation initiation and tRNA activation. Mol Acad Sci U S A 96:3584–3589
BioSyst 8:2242–2248 29. Reetz MT, Kahakeaw D, Lohmer R (2008)
21. Auslander S, Ketzer P, Hartig JS (2010) A Addressing the numbers problem in directed
ligand-dependent hammerhead ribozyme evolution. Chembiochem 9:1797–1804
Chapter 18
Abstract
Antibiotic resistance (AR), the ability of a microorganism to withstand the effects of antibiotics, is a growing
and increasingly serious global public health problem. Enzymatic activation of antibiotics though the
production of β-lactamase is one of the main mechanisms causing AR. Synthetic riboswitch containing
aptazyme is constructed in E. coli to regulate the expression of β-lactamase through small molecule–
aptamer interactions, which sharply reduces the antibiotic resistance of the engineered bacteria.
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_18, © Springer Science+Business Media New York 2014
251
252 Libing Liu and Shu Wang
2 Materials
2.1 Construction of 1. 0.5 mg/mL pBV220 solution in water (see Note 1).
Plasmid pBV220-Apt 2. AvrII (NEB) (see Note 2).
3. XhoI (NEB) (see Note 2).
4. DNA purification kit (Tiangen).
5. 10 μM oligonucleotides of aptazyme template and primers in
ultrapure water (see Note 3). Aptazyme template: GCTGTCA
CCGGATGTGCTTTCCGGTCTGATGAGTCCGTGTTC
CT GATACCAGCATCGTCTTGATGCCCTTGGCAGCAG
TGGACGAGGACGAAACAGC.
Primer 1: ATCCTAGGGCTGTCACCGGATGTGC.
Primer 2: ACCTCGAGGCTGTTTCG TCCTCG. Store at
−20 °C.
6. dNTPs (TaKaRa) (see Note 4).
7. Pfu DNA polymerase (Tiangen) (see Note 5).
8. T4 DNA ligase (TaKaRa) (see Note 5).
9. DH5α competent cells (Tiangen) (see Note 6).
Aptazyme for Reducing Antibiotic Resistance 253
2.2 Growth Curves of 1. 100 mM theophylline solution in ultrapure water (see Note 9).
E. coli TOP10 Cells Store at 4 °C.
2.3 Assay for 1. 100 mM nitrocefin solution in ultrapure water (see Note 9).
β-Lactamase Activity Store at 4 °C.
3 Methods
Fig. 1 Construction map of pBV220-Apt containing the aptazyme sequence and its electrophoretic analysis
(Line 2) and restriction enzymes (AvrII and XhoI) cutting (Line 1) (reproduced from ref. 6 with permission from
the Royal Society of Chemistry)
3.2 Growth Curves 1. Transfer a colony of E. coli TOP10 cells harboring the plasmid
of E. coli TOP10 Cell pBV220-Apt from the LB/agar plate containing 50 μg/mL
ampicillin into culture tubes containing 5 mL of LB media
supplemented with 50 μg/mL ampicillin. Then, incubate them
at 37 °C overnight in the shaking incubator (see Note 15).
2. Add 15 μL of the overnight bacteria liquid into culture tubes
containing 5 mL of fresh LB media supplemented with 50 μg/
mL ampicillin and appropriate concentrations of theophylline,
and shake the culture tubes at 37 °C with a speed of 200 rpm
in the shaking incubator (see Note 16).
3. Record the optical density at 600 nm for each concentration of
theophylline by using a plate-reading spectrophotometer at
1 h intervals over 12 h (Fig. 2) (see Note 17).
3.3 Assay for 1. Transfer two separate colonies of E. coli TOP10 cells harboring
β-Lactamase Activity pBV220 and pBV220-Apt plasmids into separate culture tubes
containing 5 mL of LB media. Then, incubate them at 37 °C
overnight in the shaking incubator (see Note 18).
2. Add 15 μL of the overnight culture into culture tubes contain-
ing 5 mL of fresh LB media supplemented with 50 μg/mL
ampicillin and appropriate concentrations (e.g., 0, 0.2, 0.4,
0.6, 0.8, 1.0 mM) of theophylline (see Note 19).
3. After the optical density at 600 nm reaches to the range of
0.5–0.6, collect 1 mL of bacterial cells by centrifugation with
3,000 × g for 4 min at 4 °C.
4. Resuspend the precipitation with 100 μL of pre-cooling puri-
fied water (see Note 20).
5. Freeze and thaw the suspension for 10 times by using liquid
nitrogen and 37 °C water.
6. Centrifuge it with 10,000 × g for 10 min.
Aptazyme for Reducing Antibiotic Resistance 255
4 Notes
11. Several reasons can account for PCR failure. First, PCR is very
sensitive to DNA contamination, which gives rise to nonspe-
cific DNA amplification. Exogenous contamination DNA can
be addressed by optimizing experimental protocols and proce-
dures. It is necessary to set up a separate PCR area for the
analysis or purification of PCR products, to use disposable
plasticware, and to thoroughly clean the work surface between
reaction facilities. Second, primer-design techniques are highly
important for improving PCR product yield and avoiding the
formation of false positive products. The use of alternative buf-
fer components or polymerase enzymes is beneficial.
12. Constant temperature is very critical for maintaining enzyme
activity. Volume of enzyme in the reaction cannot exceed 1/10
of the total. Digestion time can be extended approximately to
2 h if the ligation product cannot be obtained in the step 5 of
this section.
13. T4 DNA ligase easily loses the enzyme activity, so the ligation
reaction solution is usually kept at 4 °C. Ligation reaction
should be carried out at 4 °C for 16 h to obtain a ligation
product. We find that it is optimal to supplement additional
1 μl of T4 DNA ligase after 8 h.
14. It is necessary to incubate the competent E. coli cells for heat-
shocking at 42 °C for exact 90 s. This procedure can tran-
siently result in the formation of pores on the cell membrane,
which facilitates the entry process of exogenous DNA
(plasmid).
15. E. coli colonies on the LB plate contain both pBV220 and
pBV220-Apt. Therefore, we pick several single colonies and
send them to BGI Company for sequence to determine which
colony carries the contracted plasmid pBV220-Apt.
16. It is necessary to keep a proper shaking speed for E. coli growth.
We find the range of 150–220 rounds per minute suitable.
17. The optical density at 600 nm represents solution turbidity,
which reflects the bacterial growth condition.
18. Plasmid in E. coli TOP10 generally loses after cultivation of
generations because of unknown reasons. During experiments,
we find β-lactamase expresses higher in E. coli TOP10 than in
E. coli DH5α. Therefore, we use DH5α to store and amplify
the plasmid pBV220-Apt and use TOP10 to measure data.
19. The overnight culture is regarded as the bacterial seed for the
subsequent amplification. Mix it well before seeding.
20. Enzyme is a kind of protein in living cells and is easily digested.
Thus, the common operation is to keep the solution on ice or
low temperature after this step.
21. One unit of β-lactamase is defined as the amount which pro-
duces 1.0 mmol of product per minute.
258 Libing Liu and Shu Wang
Acknowledgements
References
1. Winkler WC, Nahvi A, Roth A, Collins JA, resistant Escherichia coli isolates from foods,
Breaker RR (2004) Control of gene expression humans, and healthy animals. Antimicrob
by a natural metabolite-responsive ribozyme. Agents Chemother 46:3156–3163
Nature 428:281–286 6. Feng X, Liu L, Duan X, Wang S (2011) An
2. Sharma V, Nomura Y, Yokobayashi Y (2008) engineered riboswitch as a potential gene-
Engineering complex riboswitch regulation by regulatory platform for reducing antibacterial
dual genetic selection. J Am Chem Soc 130: drug resistance. Chem Commun 47:
16310–16315 173–175
3. Bayer TS, Smolke CD (2005) Programmable 7. Win MN, Smolke CD (2007) A modular and
ligand-controlled riboregulators of eukaryotic extensible RNA-based gene-regulatory plat-
gene expression. Nat Biotechnol 23:337–343 form for engineering cellular function. Proc
4. Desai SK, Gallivan JP (2004) Genetic screens Natl Acad Sci U S A 104:14283–14288
and selections for small molecules based on a 8. Hedberg M, Lindqvist L, Bergman T, Nord CE
synthetic riboswitch that activates protein trans- (1995) Purification and characterization of a
lation. J Am Chem Soc 126:13247–13254 new β-lactamase from Bacteroides uniformis.
5. Brinas L, Zarazaga M, Saenz Y, Ruiz-Larrea F, Antimicrob Agents Chemother 39:
Torres C (2002) β-Lactamases in ampicillin- 1458–1461
Chapter 19
Abstract
MicroRNAs (miRNAs) offer powerful tools for targeted gene silencing in almost all eukaryotes.
These tools have received considerable attention for their utility in both fundamental genetic studies and
as therapeutic agents. Rendering individual microRNAs responsive to endogenous or exogenously applied
molecules (or ligands) can improve the stringency of silencing and can mediate autonomous control. This
chapter describes the construction of ligand-responsive miRNAs that undergo reduced processing and
subsequent gene silencing when bound by the recognized ligand. Following a simple set of rules, the
engineered microRNAs can be readily modified to target different sequences and to bind different ligands.
Individual miRNAs also can be incorporated into the same transcript for tunable, multi-gene silencing.
Key words Aptamers, Drosha, Gene regulation, Mammalian cells, MicroRNA, Riboswitch, RNA
interference
1 Introduction
MiRNAs are small regulatory RNAs that carry out gene silencing
in most eukaryotes [1]. In mammalian cells, transcribed miRNAs
undergo sequential processing by the endoribonucleases Drosha
and Dicer to form a ~22-nucleotide “mature” miRNA [2]. The
mature miRNA serves as a guide to recruit the RNA-induced
silencing complex (RISC) to complementary mRNAs, which are
silenced by cleavage, inhibition of translation, or sequestration in
inaccessible intracellular bodies [3]. Many of the known targets of
natural miRNAs are involved in development, angiogenesis, and
cancer formation, underscoring the widespread importance of
these RNA regulators [4].
MiRNAs have also received considerable attention as tools for
targeted gene silencing [5, 6]. The mature miRNA sequence can be
altered to specifically silence different genes within the host organ-
ism or to access regulatory networks of endogenous miRNAs.
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_19, © Springer Science+Business Media New York 2014
259
260 Chase L. Beisel et al.
2 Materials
3 Methods
a b
1 2
5’ G U A AU
A
A U
A A A UAU
A G U GU
GUCCUG CAGUG GCG GCACAAGCUGGAGUACAACUAUA-GUG A GGCACGU ACC U
CGGGAC GUCAU CGC CGUGUUCGACCUCAUGUUGAUAU CACCG CCGUGCA UGG U U
G C C C G A GC
G C U G
3’ AU A C A A
2
1
NO YES
UAU GA CG
GGU AC
U G U
A A A G C GU AC CCA UG
AGCUG CC GCAUC UC UGGAAAUCAGAGAAGAUAAAA CA UG
AC U G
UAU
UCGAC GG CGUAG U U ACCUUUAGUCUCUUCUAUUUU GC
G C U C U
A UC
Fig. 1 Design of ligand-responsive miRNAs. (a) MiRNAs undergo sequential processing by Drosha (circle 1) and
Dicer (circle 2), where one of the two final strands, the mature miRNA, is retained by RISC for the silencing of
complementary mRNAs. The design builds on miRNA-30a through the integration of a mature miRNA sequence
(white letters, dark gray box) and the binding domain of an aptamer (bolded letters, light gray box). The binding
domain of the aptamer is inserted below the lower stem of the miRNA. In the absence of ligand, the aptamer
will be unstructured, allowing Drosha processing. In the presence of ligand, the bound aptamer will be struc-
tured, inhibiting Drosha processing. The displayed design depicts the insertion of a mature miRNA targeting
the La gene in place of a mature miRNA targeting the GFP gene, and the insertion of the binding domain of the
theophylline aptamer in place of a large bulge. (b) The aptamer core must be of sufficient size to allow Drosha
processing in the absence of ligand. In cases where a small aptamer, such as the hypoxanthine aptamer as
shown, is selected, the aptamer binding domain and part of the loop can be integrated. Note that the binding
domain is placed adjacent to the lower stem to ensure that ligand binding inhibits Drosha processing
a
bla U6 promoter
CMV promoter
pUC ori
pCS1815 CD19
SV40 polyA 8712 bp
AgeI
neoR ClaI
BGH polyA
SV40 early promoter
CMV promoter
BGH polyA
DsRed-Express
b
AgeI AsiSI
AATAACCGGTGCGATCGCGAACGGGTCCTGATACCAGCGTGAGCGAGCACAAGCTGGAGTACAACTATAGTGAAGCCA ...
TTATTGGCCACGCTAGCGCTTGCCCAGGACTATGGTCGCACTCGCTCGTGTTCGACCTCATGTTGATATCACTTCGGT
... CAGATGTATAGTTGTACTCCAGCTTGTGCCCGCCTACGCCCTTGGCAGCAGGGCCCGTTTTTAATTAAATCGATTATT
GTCTACATATCAACATGAGGTCGAACACGGGCGGATGCGGGAACCGTCGTCCCGGGCAAAAATTAATTTAGCTAATAA
PacI ClaI
c
AgeI AsiSI
AATAACCGGTGCGATCGCGA ... CCCGTTTaaaaaaaaaaaaaaaaaaccaaaaaaaaaaTTAATTATATT
TTATTGGCCACGCTAGCGCT GGGCAAAttttttttttttttttttggttttttttttAATTAATATAA
PacI
AsiSI
AATACGATCGCaaaaaaaaaaaaaaaaaaccaaaaaaaaaaGAACGGG ... TTTTAATTAAATCGATTATT
TTATGCTAGCGttttttttttttttttttggttttttttttCTTGCCC AAAATTAATTTAGCTAATAA
PacI ClaI
Fig. 2 Cloning designed miRNAs into the pCS1815 expression plasmid. (a) Plasmid map of pCS1815. Designed
miRNAs are cloned into the AgeI/ClaI restriction sites downstream of the gene encoding the CD19 surface
marker. (b) Sequence of linear double-stranded DNA encoding the designed miRNA. The DNA includes four
restriction sites for the insertion of the double-stranded DNA into the expression plasmid. The boxed nucleotides
indicate the oligonucleotide sequences needed to PCR amplify the full-length DNA. (c) Sequences for the
sequential insertion of multiple miRNAs. The A-rich spacer (lowercase letters) helps ensure processing of each
miRNA. The top sequence can be cleaved with AgeI/PacI and inserted into AgeI/AsiSI upstream of a miRNA in
pCS1815. The bottom sequence can be cleaved with AsiSI/ClaI and inserted downstream of a miRNA in pCS1815
3.3 Measurement of 1. The cloned plasmid first is isolated from the JM110 cells. The
Ligand-Responsive purity of the DNA sample is a major factor in transfection effi-
MiRNA Activity in Cell ciency, with highly pure plasmid DNA lacking endotoxins
Culture resulting in the most reproducible, high efficiency transfec-
tions (see Note 7). The original pCS1815 plasmid or the same
plasmid containing a miRNA with a scrambled mature miRNA
sequence should be isolated; this plasmid will serve as a con-
trol when assessing the silencing of GFP expression.
2. HEK 293 cells stably expressing GFP are seeded in 24 well
plates at a density of 100,000 cells/well in Dulbecco’s
Modified Eagle Medium (DMEM) on Day 1. An exogenous
ligand can be added to the medium at this time or during
the transfection. HEK 293 cells lacking GFP can be addi-
tionally seeded to serve as a “DsRed-only” control for
color-compensation.
3. On Day 2, Fugene HD and Opti-MEM are allowed to equili-
brate to room temperature. Then purified plasmid (500 μg) is
mixed with 25 μL of Opti-MEM and 1.5 μL of Fugene HD.
This mixture is incubated at room temperature for 30 min
then added to a single well of cells and mixed with the medium
Ligand-Responsive miRNAs 265
1 - % Knockdown ( +ligand )
Fold change =
1 - % Knockdown ( -ligand )
4 Notes
Acknowledgments
References
1. Bartel DP (2009) MicroRNAs: target recognition 10. Jenison RD, Gill SC, Pardi A, Polisky B (1994)
and regulatory functions. Cell 136:215–233 High-resolution molecular discrimination by
2. Bartel DP (2004) MicroRNAs: genomics, bio- RNA. Science 263:1425–1429
genesis, mechanism, and function. Cell 11. Han J, Lee Y, Yeom KH, Nam JW, Heo I,
116:281–297 Rhee JK, Sohn SY, Cho Y, Zhang BT, Kim VN
3. Gu S, Kay MA (2010) How do miRNAs medi- (2006) Molecular basis for the recognition of
ate translational repression? Silence 1:11 primary microRNAs by the Drosha-DGCR8
4. Alvarez-Garcia I, Miska EA (2005) MicroRNA complex. Cell 125:887–901
functions in animal development and human 12. Zeng Y, Cullen BR (2005) Efficient processing
disease. Development 132:4653–4662 of primary microRNA hairpins by Drosha
5. Leisner M, Bleris L, Lohmueller J, Xie Z, requires flanking nonstructured RNA
Benenson Y (2010) Rationally designed logic sequences. J Biol Chem 280:27595–27603
integration of regulatory signals in mammalian 13. Zeng Y, Yi R, Cullen BR (2005) Recognition
cells. Nat Nanotechnol 5:666–670 and cleavage of primary microRNA precursors
6. Dickins RA, Hemann MT, Zilfou JT, Simpson by the nuclear processing enzyme Drosha.
DR, Ibarra I, Hannon GJ, Lowe SW (2005) EMBO J 24:138–148
Probing tumor phenotypes using stable and 14. Beisel CL, Chen YY, Culler SJ, Hoff KG,
regulated synthetic microRNA precursors. Nat Smolke CD (2011) Design of small molecule-
Genet 37:1289–1295 responsive microRNAs based on structural
7. Tuerk C, Gold L (1990) Systematic evolution requirements for Drosha processing. Nucleic
of ligands by exponential enrichment: RNA Acids Res 39:2981–2994
ligands to bacteriophage T4 DNA polymerase. 15. Petri S, Meister G (2013) siRNA design prin-
Science 249:505–510 ciples and off-target effects. Methods Mol Biol
8. Ellington AD, Szostak JW (1990) In vitro 986:59–71
selection of RNA molecules that bind specific 16. An CI, Trinh VB, Yokobayashi Y (2006)
ligands. Nature 346:818–822 Artificial control of gene expression in mam-
9. Hermann T, Patel DJ (2000) Adaptive recog- malian cells by modulating RNA interference
nition by nucleic acid aptamers. Science through aptamer-small molecule interaction.
287:820–825 RNA 12:710–716
Chapter 20
Abstract
We have recently developed synthetic short hairpin RNA (shRNA) switches that respond to intracellular
proteins and control the expression of target genes in mammalian cells (Kashida et al. Nucleic Acids Res
40:9369–9378, 2012; Saito et al. Nat Commun 2:160, 2011). Here, we describe a method for the three-
dimensional (3D) design of a protein-responsive shRNA switch that employs modeling software and
known 3D structures of RNA–protein complexes. We were able to predict the effect of steric hindrance
between the Dicer enzyme and shRNA-binding protein in silico by superimposing the 3D model of the
shRNA switch on Dicer. The function of the designed switch can be evaluated in vitro and in living cells.
Our expertise will help utilize the 3D structure of biomacromolecular complexes for the design of func-
tional genetic switches.
Key words 3D design, Riboswitch, RNP, shRNA, Dicer, Steric hindrance, Prediction of switch
efficiency, Synthetic biology, RNAi
1 Introduction
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6_20, © Springer Science+Business Media New York 2014
269
270 Shunnichi Kashida and Hirohide Saito
Fig. 1 The 3D design and construction of protein-responsive shRNA switches. The function of the in silico-
designed RNA switches can be evaluated using biochemical assays in vitro and gene reporter assays in mam-
malian cultured cells
Fig. 2 Schematic representation of the protein-responsive shRNA device mechanism and its bound protein
orientation. (a) In the absence of the trigger protein, Dicer can access and process the designed shRNA, and
the processed shRNA induces the knockdown of its target gene through RNAi (left). Dicer is inaccessible to
shRNA in the presence of the trigger protein that binds to the loop region of the shRNA because the RNP inter-
action faces Dicer and inhibits its access (right). The prevention of Dicer’s function causes the de-repression
of the gene knockdown. (b) Protein-responsive shRNA switches are designed in silico by connecting an RNP
motif to the corresponding dsRNA and Dicer. By specifying the position of the devices with reference to the
Dicer cleavage sites and by comparing the bound proteins’ locations, we observed that with a 1-bp (base pair)
insertion, the bound trigger protein on the shRNA switch is located approximately 2.6 Å more distant from the
Dicer cleavage sites and rotates approximately 30° in an anticlockwise direction around the axis of the dsRNA
272 Shunnichi Kashida and Hirohide Saito
2 Materials
2.2 Software Discovery Studio (Accelrys) ver. 2.5-3.1 can be used for the mod-
and Operating eling. The operation of the software should be conducted in accor-
Environment for dance with the instructions of the software. In addition, more than
Macromolecular 6 GB of cache memory capacity is recommended to avoid stalling
Design the program during modeling.
2.3 Inhibition Assay For in vitro transcription, we used T7 RNA polymerase and buffer
of Human Dicer prepared in-house or used a MEGAshortscript™ T7 Kit (AM1354,
Cleavage for shRNA Invitrogen) with slight modifications (see Note 1).
In Vitro 1. Single-stranded template DNA (purchase from an oligonucle-
2.3.1 In Vitro otide synthesis company): 5′-(complementary strand of the
Transcription and shRNA switch) TATAGTGAGTCGTATTAGC-3′ (see also
Purification of shRNA Subheading 3.3 for the sequence design of the shRNA switch).
Switches 2. T7 annealing primer: 5′-GCTAATACGACTCACTATA-3′.
3. Transcription reaction mixture: 0.75 μM single-stranded tem-
plate DNA, 0.75 μM T7 annealing primer, 7.1 μg/mL Pyro
phosphatase (Roche), 50 μg/mL bovine serum albumin,
40 mM HEPES–KOH, 20 mM MgCl2, 5 mM DTT, 2 mM
Table 1
Features of the RNP structures used for shRNA switches
2.3.2 Expression and 1. Competent cell: BL21 (DE3) pLysS (Promega) or chemically
Purification of the Trigger competent E. coli cells suitable for transformation and protein
RNA Binding Proteins expression.
Using FPLC 2. Phosphate buffer A: 20 mM NaH2PO4, 20 mM Na2HPO4,
500 mM NaCl.
3. Phosphate buffer B: 20 mM NaH2PO4, 20 mM Na2HPO4,
500 mM NaCl, 1 M imidazole.
4. FPLC: AKTApurifier (GE Healthcare).
5. Prepacked column: HisTrap HP column (5 mL, GE
Healthcare).
6. 6× Sample buffer: Sample buffer solution with reducing
reagent (6×) for SDS-PAGE (Nacalai).
7. 2× dialysis buffer: 40 mM HEPES–KOH, pH 7.8, 230 mM
NaCl, 10.8 mM KCl, 1.6 mM MgCl2.
8. Dialysis cassette: Slide-A-Lyzer Dialysis Cassettes (Thermo
Scientific).
9. Sample concentrator: Vivaspin 6 concentrator (GE Healthcare).
2.3.3 Inhibition Assay of 1. Recombinant human Dicer: Recombinant human Dicer can be
Human Dicer Cleavage for purchased from Genlantis.inc for the experiments. Human
shRNA In Vitro Dicer purified in the laboratory can also be used for the
experiments.
2. Dicer reaction mixture: 1 μL of 10 mM ATP, 0.5 μL of 50 mM
MgCl2, 4 μL of Dicer Reaction Buffer (Genlantis), 1 μL of 0.5
unit/μL Recombinant Human Dicer Enzyme (Genlantis).
3. Binding buffer: Opti-MEM I (Life Technologies).
4. Gel staining buffer: 1× SYBR Green I and SYBR Green II in
0.5× TBE buffer.
274 Shunnichi Kashida and Hirohide Saito
2.4.2 Trigger Protein pcDNA3.1, 4 and 5 (Life Technologies) or other expression plas-
Expression Plasmid mids suitable for expression in mammalian cells can be used.
2.4.3 EGFP Expression 1. Human cultured cells: Maintain 293FT cells (Life Technologies)
Control of the Designed using Dulbecco’s modified Eagle’s medium (Nacalai) contain-
shRNA Switches in ing 10 % fetal bovine serum (Cambrex), 2 mM L-glutamine
Cultured Mammalian Cells (Life Technologies), 0.1 mM MEM Nonessential Amino Acids
(Life Technologies), 1 mM sodium pyruvate (Sigma), 0.5 %
Penicillin-Streptomycin (Life Technologies) and 500 μg/mL
G418 sulfate at 37 °C under a 5 % CO2 concentration. HeLa
cells, which were also used, and other human cultured cells
successfully transfected with plasmids can also be used for the
experiment.
2. Lipofection reagent: Lipofectamine 2000 (Life Technologies).
3. Fluorescent microscopy: To observe EGFP expression in cells,
we used an IX-81 inverted microscope system (Olympus), a
470- to 495-nm filter for excitation and a 510- to 550-nm fil-
ter for emission.
4. Flow cytometry: To detect and quantify EGFP fluorescence,
we used a flow cytometer (FACS Aria, BD Bioscience), a 488-
nm semiconductor laser beam, and a 530/30-nm emission
detector.
3 Methods
Fig. 3 Overview: 3D molecular design and assessment of the protein-responsive shRNA switches
3.2 dsRNA Sequence Obtain the dsRNA sequence that induces knockdown of the target
Targeting Specific gene through RNAi from literature reports or databases. Sequences
mRNA that start with “G” are preferable because the first “C” in the DNA
template is better suited for the efficient transcription of T7
RNA polymerase in vitro and RNA polymerase III in human cells.
If there are no known dsRNA sequences, construct and assess the
276 Shunnichi Kashida and Hirohide Saito
3.3 shRNA Switch Obtain the 3D molecular structure of the RNP motif composed of
Design on the the trigger protein and its binding RNA motif from the PDB. If
Secondary Structure the 3D structure is not available, the 3D molecular design step
(Fig. 3, Step 1) can be skipped. In this case, the shRNA switches
can be constructed based on the secondary structure as follows
(Fig. 4).
1. Choose the appropriate RNA motifs. Omit the unnecessary
sequences (sequences not having a positive effect on binding
affinity) and extract minimal RNA motifs.
2. Design defective RNA motifs that do not bind to the trigger
proteins by mutating or deleting the corresponding nucleo-
tides of the motif.
3. Connect the RNA motif with the corresponding length of
dsRNA (21–31 bp, also see Note 3) designed to code the tar-
get mRNA sequence to form the stem-loop structure of the
RNA (Fig. 4, 1–3).
4. Add two nucleotides (“AG” or “UU”) at the 3′ end of the
RNA (Fig. 4, 4) to help Dicer recognize the target dsRNA.
3D Molecular Design for shRNA Switches 277
3.5 Prediction of The entire structure of human Dicer has not been solved. For our
Steric Hindrance 3D model of Dicer, we used the RNase III catalytic domain and
Between Dicer and the peripheral structure of Giardia Dicer (PDB ID: 2QVW, 326–385,
Trigger Protein Bound and 401 C-terminal a.a.; see Note 6). In this section, we demon-
on the shRNA Switch strate how to display the catalytic domain of Dicer, highlight the
catalytic sites and superimpose the RNP on Dicer.
3.5.1 Displaying and 1. Download the PDB file of the crystal structure of Giardia
Modifying the Catalytic Dicer (PDB ID: 2QVW). The crystal structure has four sym-
Domain and Peripheral metric groups in the crystal lattice.
Structure of Giardia Dicer 2. Exclude three groups and focus on one group.
3. Trim the a.a. sequence of the protein without 326–385 and
401 of the C-terminal a.a by unchecking the checked boxes of
the nucleotides.
4. Change the display style of the Dicer protein and the four
metal ions from “None” to “CPK,” and highlight the metal
ions and 9 a.a. (E336, E340, D404, D407, E649, D653,
E684, D720, and E723) by changing the color of the atoms.
3.5.2 Superimpose the 1. Copy and paste the 3D molecular model of the protein-
Protein-Responsive shRNA responsive shRNA switch variants and bound trigger protein
Switch and Bound Trigger (designed in Subheading 3.4) on the modified Dicer structure
Protein on Dicer displayed in the window.
2. Arrange the positions and orientations of the switch and Dicer
three-dimensionally in reference to the two phosphates of the
22nd nucleotides, the highlighted metal ions and the 9 a.a. of
Dicer.
3. Choose the modified Dicer and add surfaces to the proteins.
4. Display the modified Dicer and each variant of the shRNA
switch with the trigger protein and observe the degree of ste-
ric hindrance between them. You can predict which trigger
protein of the variants will most strongly collide against Dicer
(see Note 7).
3.6.2 Expression and The use of an affinity column and FPLC, such as the AKTA (GE
Purification of the Trigger Healthcare) system, is recommended for purification of the trigger
RNA Binding Proteins RNA binding proteins. For U1A protein purification, we modified
Using FPLC a protein expression and purification protocol that has been
reported previously [22].
1. Pre-culture competent cells transformed with the His-tagged
trigger protein expression plasmid with 10 mL of LB (+antibiotic
for selection) at 37 °C and 200 rpm (linear shake) overnight.
3D Molecular Design for shRNA Switches 281
3.6.4 The Efficiency 1. Mix 0.5 μL of in vitro-transcribed shRNA switch with the Dicer
of In Vitro Human Dicer reaction mixture and water (up to 10 μL). Do not chill the
Cleavage for the shRNA mixture during assembly because this may cause precipitation.
Switches
2. Incubate the mixture at 37 °C for 15 h and stop the incubation
by adding 1 μL of Dicer Stop Solution to the reaction.
3. Conduct the same procedure detailed in steps 3–8 in
Subheading 3.6.3 on the incubated reaction.
4. Quantify the total fluorescent intensity of the band of cleaved
dsRNA and confirm that the cleavage efficiency by human
Dicer is the same among the base-pair variants and their nega-
tive controls (defective RNA motif) for the shRNA switches.
3.6.5 Inhibition Assay 1. Mix 0.5 μM in vitro-transcribed shRNA switch with the corre-
of Human Dicer Cleavage sponding trigger protein (The concentration of the protein was
for shRNA In Vitro determined at step 10 in Subheading 3.6.3 to form the RNP
complex). The total volume of the mixture is up to 3.5 μL.
2. (Optional) Incubate at room temperature for approximately
30 min. The incubation time will depend on the affinity
between the RNP complexes.
3D Molecular Design for shRNA Switches 283
3.7 EGFP Expression The ShRNA cloning Kit can be used to construct the shRNA-
Control of the switch expressing plasmids. The plasmids should be constructed
Designed shRNA using the kit, and required DNA sequences should be synthesized
Switches in Cultured in accordance with the kit manual. The annealing steps in the kit
Mammalian Cells manual are modified as bellow:
3.7.1 Construction 1. Mix 200 μM of ShRNA top strand and 200 μM of ShRNA
of shRNA Expressing bottom strand in Oligo Annealing Buffer of the cloning kit.
Plasmids 2. Incubate the mixture using a thermal cycler: 95 °C for 4 min
and incubate at 80 °C for 1 min. Decreased 0.1 °C/s to the
melting temperature of the duplex of the top strand and bottom
strand. And incubate the mixture at the melting temperature for
5 min, followed by room temperature incubation for 10 min.
Prepare the negative control plasmid that contains a defective
RNA motif using the same procedure as indicated.
3.7.2 Trigger Protein The trigger protein expression plasmid is prepared as follows.
Expression Plasmid
1. Design the set of PCR primers for amplifying the region of the
gene that codes the trigger protein, whose amino acid (a.a.)
sequence corresponds with that of the protein in the RNP
complex derived from the PDB. Add each sequence of PCR
primer to the cutting site of a selected different restriction
enzyme.
2. Amplify the gene by PCR from the total cDNA or plasmid
using the designed primers.
3. Clone the gene into destination plasmids between the two dif-
ferent restriction enzyme sites to construct the trigger protein
expression plasmid.
4. Verify the sequence.
5. Prepare the negative control plasmid that contains a noncog-
nate protein sequence using the same procedure as indicated.
284 Shunnichi Kashida and Hirohide Saito
3.7.3 EGFP Expression 1. Seed 293FT cells (1.5 × 105 per well) or other chosen cultured
Control of the Designed cells on a 24-well plate and incubate at 37 °C in a CO2 incuba-
shRNA Switches in tor for 1 day.
Cultured Mammalian Cells
2. Co-transfect cells with the following three types of plasmids
using 2 μL of the Lipofection reagent: 0.3 μg of the bp variant
of protein-shRNA switches expressing plasmids or that of the
binding-defective control, 0.3 μg of the trigger protein expres-
sion plasmid or that of the binding-defective control and
0.3 μg of the EGFP expression plasmid.
3. Twenty-four hours after transfection, observe and image the
transfected cells using a fluorescent microscope (Fig. 3, Step 3).
4. Collect the medium of the 24-well plate and transfer to new
tubes.
5. Add 200 μL of 0.25 % trypsin-EDTA (1×) phenol red to each
well and incubate at 37 °C in a CO2 incubator for 2–5 min.
6. Add the collected medium to each well, resuspend in the
medium and filter the cell suspension with a 12 × 75 mm tube
and a cell strainer cap.
7. Quantify the mean intensity of the EGFP fluorescence of
30,000 cells per sample using a flow cytometer.
8. To exclude abnormal cells, select the cells whose FCS and SSC
signal scopes are identical to those of untreated cells to calcu-
late the mean intensities.
4 Notes
References
1. Endy D (2005) Foundations for engineering ulation of internal ribosome entry in wheat
biology. Nature 438:449–453 germ extract and their applications as label-free
2. Kashida S, Inoue T, Saito H (2012) Three- biosensors. RNA 17:478–488
dimensionally designed protein-responsive 16. Sinha J, Reyes SJ, Gallivan JP (2010)
RNA devices for cell signaling regulation. Reprogramming bacteria to seek and destroy
Nucleic Acids Res 40:9369–9378 an herbicide. Nat Chem Biol 6:464–470
3. Saito H, Fujita Y, Kashida S et al (2011) 17. Muranaka N, Sharma V, Nomura Y et al (2009)
Synthetic human cell fate regulation by protein- An efficient platform for genetic selection and
driven RNA switches. Nat Commun 2:160 screening of gene switches in Escherichia coli.
4. Xie Z, Wroblewska L, Prochazka L et al (2011) Nucleic Acids Res 37:e39
Multi-input RNAi-based logic circuit for iden- 18. Liang JC, Bloom RJ, Smolke CD (2011)
tification of specific cancer cells. Science 333: Engineering biological systems with synthetic
1307–1311 RNA molecules. Mol Cell 43:915–926
5. Culler SJ, Hoff KG, Smolke CD (2010) 19. Oubridge C, Ito N, Evans PR et al (1994)
Reprogramming cellular behavior with RNA Crystal structure at 1.92 A resolution of the
controllers responsive to endogenous proteins. RNA-binding domain of the U1A spliceosomal
Science 330:1251–1255 protein complexed with an RNA hairpin.
6. Park J-E, Heo I, Tian Y et al (2011) Dicer rec- Nature 372:432–438
ognizes the 5′ end of RNA for efficient and 20. Galardi S, Mercatelli N, Farace MG et al (2011)
accurate processing. Nature 475:201–205 NF-kB and c-Jun induce the expression of the
7. Vermeulen A, Behlen L, Reynolds A et al oncogenic miR-221 and miR-222 in prostate
(2005) The contributions of dsRNA structure carcinoma and glioblastoma cells. Nucleic
to Dicer specificity and efficiency. RNA 11: Acids Res 39:3892–3902
674–682 21. Ben-Neriah Y, Karin M (2011) Inflammation
8. Tsutsumi A, Kawamata T, Izumi N et al (2011) meets cancer, with NF-κB as the matchmaker.
Recognition of the pre-miRNA structure by Nat Immunol 12:715–723
Drosophila Dicer-1. Nat Struct Mol Biol 22. Endoh T, Sisido M, Ohtsuki T (2008) Cellular
18:1153–1158 siRNA delivery mediated by a cell-permeant
9. An C-I, Trinh VB, Yokobayashi Y (2006) RNA-binding protein and photoinduced RNA
Artificial control of gene expression in mam- interference. Bioconjug Chem 19:1017–1024
malian cells by modulating RNA interference 23. Poeck H, Besch R, Maihoefer C et al (2008)
through aptamer–small molecule interaction. 5′-Triphosphate-siRNA: turning gene silencing
RNA 12:710–716 and Rig-I activation against melanoma. Nat
10. Beisel CL, Bayer TS, Hoff KG et al (2008) Med 14:1256–1263
Model-guided design of ligand-regulated 24. Kim D-H, Longo M, Han Y et al (2004)
RNAi for programmable control of gene Interferon induction by siRNAs and ssRNAs
expression. Mol Syst Biol 4:224 synthesized by phage polymerase. Nat
11. Rybak A, Fuchs H, Smirnova L et al (2008) A Biotechnol 22:321–325
feedback loop comprising lin-28 and let-7 con- 25. Sutcliffe MJ (1993) Representing an ensemble
trols pre-let-7 maturation during neural stem- of NMR-derived protein structures by a single
cell commitment. Nat Cell Biol 10:987–993 structure. Protein Sci 2:936–944
12. Nam Y, Chen C, Gregory RI et al (2011) 26. Takeshita D, Zenno S, Lee WC et al (2007)
Molecular basis for interaction of let-7 microR- Homodimeric structure and double-stranded
NAs with Lin28. Cell 147:1080–1091 RNA cleavage activity of the C-terminal RNase
13. Beisel CL, Smolke CD (2009) Design princi- III domain of human dicer. J Mol Biol 374:
ples for riboswitch function. PLoS Comput 106–120
Biol 5:e1000363 27. Macrae IJ, Li F, Zhou K et al (2006) Structure
14. Salis HM, Mirsky EA, Voigt CA (2009) of Dicer and mechanistic implications for
Automated design of synthetic ribosome bind- RNAi. Cold Spring Harb Symp Quant Biol 71:
ing sites to control protein expression. Nat 73–80
Biotechnol 27:946–950 28. Macrae IJ, Zhou K, Li F et al (2006) Structural
15. Ogawa A (2011) Rational design of artificial basis for double-stranded RNA processing by
riboswitches based on ligand-dependent mod- Dicer. Science 311:195–198
INDEX
Atsushi Ogawa (ed.), Artificial Riboswitches: Methods and Protocols, Methods in Molecular Biology, vol. 1111,
DOI 10.1007/978-1-62703-755-6, © Springer Science+Business Media New York 2014
287
ARTIFICIAL RIBOSWITCHES: METHODS AND PROTOCOLS
288 Index
Fluorometer. See Spectrofluorometer Phosphorimager ................ 44, 49, 50, 55, 212, 215, 226, 231
FPLC. See Fast protein liquid chromatography (FPLC) Plate reader ...................................................20, 82, 111, 133,
Free energy .................................. 5, 6, 99, 128, 174, 180, 230 145, 226, 242, 253
FRET. See Fluorescence resonance energy transfer (FRET) Polymerase chain reaction (PCR) .................................. 1, 18,
32, 43, 81, 95, 113, 133, 142, 156, 168, 184, 200,
G 212, 228, 240, 253, 263, 283
Gel purification Promoters
agarose gel ............................................ 83, 168–169, 180 CMV promoter .................................. 185–186, 262, 266
polyacrylamide gel ....................... 215, 227, 228, 242, 280 lac promoter ........................... 94, 111, 112, 115, 119, 126
Lux promoter...................................................... 142, 149
I Rha promoter .........................................................84, 88
tac promoter .................................................................81
Internal ribosomal entry site (IRES) ........ 168, 170–180, 187
T7 promoter .........................................22, 25, 30–31, 42,
In vitro compartmentalization ..................................158–160
43, 122, 156, 157, 168, 176, 180, 204, 210–211,
In vitro selection
228, 229
aptamers ......................................... 17–27, 29–39, 41–55
Trc promoter............................................................... 144
aptazymes ........................................... 209–219, 223–228
U6 promoter ....................................................... 262, 266
In vitro transcription ..................................23, 42, 45, 47, 54,
Pull-down assay ................................................ 43–44, 49–50
72, 113–114, 122, 123, 126, 127, 155–156, 158,
PURE system ................................................... 155, 158–161
169, 176, 217, 222, 225, 234, 272–273, 276,
279–280, 282 Q
In vitro translation .................................................... 158, 176
In vivo screening. See In vivo selection Quadruplex ............................................... 197–199, 201–206
In vivo selection
R
aptazymes ........................................................... 224, 225
riboswitches ............................................... 57–74, 77–90, Rational design ...................................................... 52, 93, 94,
93–104, 107–128, 131–140, 237–248 108, 141, 165–181, 240
IPTG. See Isopropyl β-D-1-thiogalactopyranoside (IPTG) RBS. See Ribosome binding site (RBS)
IRES. See Internal ribosomal entry site (IRES) Real-time PCR............................................. 20, 26, 104, 161
Isopropyl β-D-1-thiogalactopyranoside Reporter protein
(IPTG) ...............................94, 95, 101, 102, 104, chloramphenicol acetyltransferase (CAT) ..................110
112, 115, 117, 119, 120, 226, 229, 281 fluorescent protein (FP)
Isothermal titration calorimetry (ITC) ............ 110, 114–115, CFP variant ..........................................................194
123–125, 127, 128 GFP variant ............................ 78, 110, 132, 147, 157,
ITC. See Isothermal titration calorimetry (ITC) 191, 246, 261, 274
RFP variant .......................................... 191, 228, 262
L YFP variant..................................................... 78, 157
Ligand molecule. See Aptamers; Aptazymes β-galactosidase (lacZ) ............................................ 99, 154
Liposomes ................................................ 156, 160, 162–163 LacI ..............................................................................94
β-lactamase ........................................................... 81, 252
M luciferase ..................................................................... 168
RpoS .............................................................................94
Mammalian cells ....................................... 64, 183–196, 221,
tobacco etch virus (TEV) protease .........................78–79
259–261, 270, 274, 277, 283–284
Reverse transcriptase-mediated enzymatic
MicroRNA (miRNA)............................... 239, 259–266, 270
analysis (RTase stop assay) ..................... 198–201,
Mutagenesis............................................72, 80, 82, 107–128,
204, 205
143–144, 149
Reverse transcription-PCR (RT-PCR) ....................... 22, 24,
N 26, 27, 30, 34–35, 38, 39, 42, 46–48, 52, 54–55,
198, 201, 211–213, 216, 226, 228
Negative selection ............................................ 19, 32–34, 42, Ribonucleoprotein (RNP) ....................................... 184, 187,
45–46, 67, 141, 213–218, 224, 228 194, 270–272, 274–277, 279, 282, 283, 285
Riboregulators .................................................. 197–206, 240
P
Ribosome binding site (RBS) ................................ 62, 72, 73,
PCR. See Polymerase chain reaction (PCR) 79, 90, 96, 97, 100, 101, 139, 149, 151, 153–154,
Phenol-chloroform extraction ...........122–123, 157, 178, 279 156, 157, 159, 161, 165, 166, 177, 238
ARTIFICIAL RIBOSWITCHES: METHODS AND PROTOCOLS
Index
289
Riboswitches Rosetta............................................................................4
in bacteria .....................................57–74, 77–90, 93–104, siExplorer ...................................................................276
107–128, 131–140, 221–234, 237–248, 251–258 Vienna RNA ..........................................................4, 229
in mammalian cells ............................................. 183–196 Spectrofluorometer. .............................................. ....156, 161
in PURE system ................................................. 153–163 Spectrometer .........................................43, 45, 169, 176, 200
in wheat germ extract ......................................... 165–181 SPR. See Surface plasmon resonance (SPR)
RISC. See RNA induced silencing complex (RISC) Surface plasmon resonance (SPR) .................... 30, 36–37, 51
RNAi. See RNA interference (RNAi) Systematic evolution of ligands by exponential enrichment
RNA induced silencing complex (RISC) ................. 259, 262 (SELEX). See In vitro selection, aptamers
RNA interference (RNAi) ....................................... 239, 269,
271, 274–276, 284, 285 T
RNP. See Ribonucleoprotein (RNP) Target molecule. See Aptamers; Aptazymes
RT-PCR. See Reverse transcription-PCR (RT-PCR) Thermal cycler ................................ 20, 43, 95, 113, 133, 146,
169, 184, 200, 253, 283
S Three-dimensional (3D) structure......................... 3–5, 8–11,
13, 222, 272, 274, 276
Secondary structures ............................................ 2–8, 13, 17, Transfection ..............................................185, 187, 189, 191,
19, 29, 80, 99, 127, 135, 201, 203, 222, 223, 230, 193–196, 260, 262, 264–266, 274, 276, 284
234, 240, 261, 270, 276 Transformation ..................................... 25, 58, 59, 65, 68, 79,
Sequencing ....................................... 1, 18, 29, 41, 58, 78, 96, 82, 84, 87, 90, 97, 117, 119, 133, 134, 137, 140,
116, 133, 141, 153, 165, 184, 196, 210, 222, 239, 147, 150, 157, 168, 184, 187, 188, 222, 228, 229,
252, 259, 269 231, 245–248, 264, 273, 280
Short hairpin RNA (shRNA) ...................................269–285 Transilluminator ..............................20, 82, 87, 133, 140, 169
shRNA. See Short hairpin RNA (shRNA)
Software U
AutoDockTools (ADT)................................................12
UV melting analysis...........................198, 200, 202–203, 205
AutoDock Vina ............................................................12
ChemFinder ....................................................... 111, 116 W
Discovery Studio ........................................................272
OpenBabel......................................................................4 Water-in-oil emulsion ......................................................155
RNAstructure ....................................................... 96, 169 Wheat germ extract .................................. 168, 169, 176, 178