You are on page 1of 7

PROTOCOL

Design and cloning of lentiviral vectors expressing


small interfering RNAs
Gustavo Tiscornia, Oded Singer & Inder M Verma

The Salk Institute for Biological Studies, Laboratory of Genetics, 10010 North Torrey Pines Road, La Jolla, California 92037, USA. Correspondence should be addressed to
I.M.V. (verma@salk.edu).

Published online 27 June 2006; doi: 10.1038/nprot.2006.36

RNA interference (RNAi) has emerged as a powerful technique to downregulate gene expression. The use of polIII promoters to
express small hairpin RNAs (shRNAs), combined with the versatility and robustness of lentiviral vector–mediated gene delivery to
© 2006 Nature Publishing Group http://www.nature.com/natureprotocols

a wide range of cell types offers the possibility of long-term downregulation of specific target genes both in vitro and in vivo. The
use of silencing lentivectors allows for a rapid and convenient way of establishing cell lines (or transgenic mice) that stably express
shRNAs for analysis of phenotypes produced by knockdown of a gene product. Here we present two possible protocols describing the
design and cloning of silencing lentiviral vectors. These protocols can be completed in less than 3 weeks.

INTRODUCTION
The procedures described in this protocol will allow the reader mobilization in the transduced cell6,7. For further description of
to take full advantage of the combination of two powerful tech- the lentiviral vector system, see ref. 1.
niques, lentiviral vector technology for gene transfer and gene
silencing by RNAi. While a comprehensive description of the Overview of RNAi
basic biology underlying these two techniques is beyond the RNAi has forcefully emerged as a pathway constituting a whole
scope of this protocol, a very brief overview of each will be given, new level of control of gene expression8. The pathway has been
along with literature references containing more detailed infor- under intense study, and details of its basic biological mecha-
mation for readers unfamiliar with these technologies. This step- nism are described elsewhere8–13. Briefly, short 21–23 nucleotide
by-step protocol is designed as a comprehensive set of instruc- double-stranded small interfering RNAs (siRNAs) are incorpo-
tions for the researcher with basic molecular skills (but not nec- rated into a multi-component nuclease complex (RISC: RNA–
essarily experience in either lentiviral vectors or RNAi) seeking induced silencing complex) that selects and degrades mRNAs
to clone a lentiviral vector capable of efficient downregulation of that contain a target complementary to the antisense strand of the
a particular target; it is intended to be used in conjunction with siRNA14,15. In mammalian systems, siRNAs can be delivered by
the accompanying protocol1 describing production and purifi- either transient transfection of synthetic siRNAs16,17 or transient
cation of lentiviral vectors. It must be emphasized that because or stable transfection (if viral vectors are used, transduction) of
residual levels of expression (commonly 5–15% of wild-type constructs expressing shRNAs from polIII promoters18–22. More
expression levels) remain even with the most efficient shRNAs, recently, methods have been developed for using polII promoters
the technique produces hypomorphs, not nulls; thus, the suit- for expression of shRNAs in the context of microRNA precur-
ability of the approach will depend on the particular target gene sors23–25. The shRNA is processed by the endogenous microRNA
in question. For example, effective knockdown of a hypothetical pathway, resulting in the production of a mature siRNA and
enzyme for which 5% of wild-type expression levels are enough subsequent degradation of the target mRNAs. For further read-
to preserve wild-type function will not be expected to result in ing on various aspects of shRNA and microRNA biology, see
a phenotype. refs. 12,13,26–31.

Overview of lentiviral vectors Design and cloning of lentiviral vectors expressing shRNAs
Lentiviral vectors derived from HIV-1 are capable of transduc- The two polIII promoters most commonly used for hairpin
ing a wide variety of dividing and non-dividing cells, and they expression are H1 and U6 (both human and mouse); both pro-
integrate stably into the host genome, resulting in long-term moters have efficient, ubiquitous expression and are essentially
expression of the transgene2. The presence of vesicular stoma- equivalent for in vitro experiments, although expression effi-
titis virus glycoprotein (VSVG) in the viral envelope membrane ciency differences in vivo have been reported32. PolIII promoters
confers the viral particle with the ability to transduce a broad are of compact size (less than 400 bp) and (because all sequenc-
range of cell types, including primary cells, stem cells and early es required for promoter function are upstream of the +1 tran-
embryos3–5. An important safety feature is provided by a dele- scriptional start site33) are ideally suited to express shRNAs
tion of the promoter-enhancer region in the 3′LTR (SIN vec- consisting of approximately 60 nucleotides that include a 21–23
tors). During reverse transcription the proviral 5′LTR is copied nucleotide sense sequence that is identical to the target sequence
from the 3′LTR, thus transferring the deletion to the 5′LTR; the in the mRNA to be downregulated, followed by a 9-nucleotide
deleted 5′LTR of the integrated provirus is therefore transcrip- loop and an antisense 21–23 nucleotide sequence. A stretch
tionally inactive, preventing subsequent viral replication or of five Ts provides a polIII transcriptional termination signal.

234 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS


PROTOCOL

BOX 1 VALIDATION OF THE CLONED shRNA CASSETTES


The simplest way to validate the shRNA cassettes is to coexpress a tagged cDNA of the target gene together with the shRNA silencing
cassette in an easily transfected cell line (e.g., 293T HEK cells). This approach will work even with modest transfection efficiencies,
an advantage that is lost if knockdown of the endogenous target is chosen as the validation method. Typically, proceed as described
below:
(i) Seed 105 293T HEK cells in a 24-well plate the day before transfection.
(ii) Mix target cDNA plasmid with the plasmid containing the silencing cassette in 1/5 molar ratio. If comparing different wells,
include a cotransfection control (ideally, a plasmid expressing any cDNA of a size different from the tagged cDNA being knocked
down, but containing the same tag, so as to detect both proteins with the same antibody); aim for a total amount of DNA of
1 µg/well and be sure to equalize the total amount of DNA transfected per well, filling up with nonspecific plasmid DNA if
necessary.
© 2006 Nature Publishing Group http://www.nature.com/natureprotocols

(iii) Transfect the DNA mix into the cells using Fugene or equivalent transfection reagent.
(iv) Perform western blot analysis Harvest protein 48–72 h after transfection. A caveat can arise if you choose to test the silencing
cassette by transfecting the lentiviral silencing vector (as opposed to a plasmid expressing only the shRNA) mixed with a tagged
cDNA driven by a CMV promoter. Since the silencing lentivector plasmid also contains GFP driven by CMV, cotransfecting both
plasmids will result in less expression of the tagged cDNA target simply due to promoter competition. The appropriate control is a
1/5 molar ratio transfection of the target cDNA plasmid and L-CMV-GFP (containing no silencing cassette).

The total length of the silencing cassette is ~350 bp. Expression can begin with any base, but efficiency of expression from a U6
results in a short 21–23 bp hairpin; the loop is cleaved by the promoter depends on the first base transcribed, with G result-
endonuclease Dicer and the resulting siRNA triggers degrada- ing in strong expression and efficiency decreasing with other
tion of the target gene mRNA. bases40. shRNAs can be directed to the 5′UTR, ORF or 3′UTR of
Development and validation of an efficient lentiviral silenc- the target mRNA as desired. Strings of identical bases should be
ing vector involves (i) selection of siRNA target sequences and avoided in order to avoid stalling by the RNA polymerase III (in
design of shRNAs, (ii) cloning and validation of shRNAs and particular 5 Ts constitute a polIII transcriptional termination
(iii) cloning the lentiviral silencing vector. signal). As a loop we generally use the 9 bp sequence (TTC AAG
AGA)18, but a variety of loops have been reported. The design of
Selection of siRNA target sequences and design of shRNAs an shRNA hairpin for a given target is best explained by example
A number of algorithms have been developed to predict effec- (see Step 1).
tive siRNA sequences34–38 and many of them are available online
for free or commercial use (e.g., http://www.ambion.com or Cloning and validation of shRNAs
http://sfold.wadsworth.org). Typically, 4–6 shRNAs need to be The candidate shRNAs are cloned into a plasmid containing
generated and tested for every target gene. In general, the target only the silencing cassette (see Steps 1A and 1B) and tested for
sequence should be 21–23 bases long, but lengths of up to 28 efficiency of downregulation (Box 1).
bases have been reported22. Longer targets should be avoided,
as longer dsRNA molecules can trigger a PKR (protein kinase Cloning lentiviral silencing vectors
activated by double stranded RNA) response39. A database When an efficient shRNA candidate has been identified, it is
search is recommended to filter out candidate targets that are cloned into the lentiviral vector (see Steps 1A and 1B). High-
present in other genes to avoid silencing of these loci. The G/C titer lentiviral suspensions can be prepared and validated for
content should be 40–55%. shRNAs driven by the H1 promoter target downregulation (Box 2)1.

MATERIALS
REAGENTS •NheI restriction enzyme
• Plasmid expressing a tagged cDNA target • SspI restriction enzyme
• Mammalian cells: 293T human embryonic kidney (HEK; Invitrogen cat. no. • Bacterial strains: general cloning-competent E. coli, such as TOP10 (Invitrogen
R700-07) cat. no. 44-0301 or DH5α (Invitrogen cat. no.18265-017).
• Transfection reagent: Fugene (Roche cat. no. 11814443001) FOR STEP 1B ONLY:
• LB-Ampicillin plates and media • Plasmids: pENTR/U6 (Invitrogen cat. no. K4945-00); L-pDest-CMV-GFP
(Verma Lab, Salk Institute for Biological Studies)
FOR STEP 1A ONLY:
• Custom-designed oligonucleotides (O1 and O2, order at 200 µM
• Plasmids: pH1; L-CMV-GFP-NheI (Verma Lab, Salk Institute for Biological
concentration, PAGE purified; for design, see Step 1Bi)
Studies)
• Primers: hU6-F: 5′-GGACTATCATATGCTTACCG-3′, M13-Reverse
• PCR reagents (Advantage GC-2 Polymerase Mix (BD cat. no. 639114)
• BsaI restriction enzyme
• Primers (P1 and P2; for design, see Step 1A)
• NdeI restriction enzyme
• H1-F: 5′-TGGCAGGAAGATGGCTGTGA-3′
• ClaII restriction enzyme
• XbaI restriction enzyme

NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 235


PROTOCOL
• AvaII restriction enzyme • LR clonase (Invitrogen cat. no. 11791-020)
• Bacterial strains: transformation-competent E. coli DB3.1 (Invitrogen cat. no. • 10× annealing buffer: 100 mM Tris-HCl pH 8, 10 mM EDTA, 1M NaCl
11782-018) and E. coli STBL3 (Invitrogen cat. no. C7373-03) EQUIPMENT
• LB-Kanamycin plates and media • PCR machine
• Proteinase K (2 µg/µl) • DNA electrophoresis equipment
• SDS-PAGE electrophoresis equipment

PROCEDURE
Lentiviral vector creation and cloning a
A lentiviral vector carrying GFP as a marker and the
silencing cassette positioned in the 3′LTR (Option A) results
in duplication of the silencing cassette upon proviral
integration. While this maximizes the silencing power of the
© 2006 Nature Publishing Group http://www.nature.com/natureprotocols

vector, it can also lead to lower expression of the marker


because the siRNA target sequence is also present in the
mRNA expressing the marker gene (GFP). To avoid this b
problem, use option B, in which the silencing cassette is
cloned upstream of the marker; in this case, only one copy of c
the silencing cassette will be delivered per viral particle.
(A) Clone the silencing cassette into a unique NheI site d
in the 3′LTR of a lentiviral vector expressing GFP as a
marker (this cloning strategy is summarized in Fig. 1).
(i) Select target and design primers to amplify the
silencing cassette. Select a target within the
e
gene to be silenced (e.g., for human p53: 5′-
GACTCCAGTGGTAATCTAC-3′; ref. 41). The forward primer
(P1) must contain a NheI-compatible site such as
XbaI. The reverse primer (P2) includes sequence
complementary to the entire shRNA cassette (in the
5′-to-3′ orientation of the primer: XbaI site + 5As +
target sense sequence + loop anti-sense sequence
+ target anti-sense sequence) followed by 22 bases
complementary to the last 22 bases upstream of the Figure 1 | Cloning scheme for Option A. The most convenient procedure is
+1 transcriptional start site (sense strand) of the to PCR amplify the silencing cassette42 from a template polIII promoter by
using a 5′ forward primer (P1) upstream of the polIII promoter (containing
polIII promoter (Fig. 1a). For the human p53 target
an NheI-compatible site such as XbaI) and a 3′ reverse primer (P2)
suggested above, design the following 3′ reverse that includes sequence complementary to the entire shRNA (in 5′-to-3′
primer: 5′CTGTCTAGACAAAAAGACTCCAGTGGTAATCTACTCT orientation of the primer: XbaI site + 5As + target sense sequence + loop
CTTGAAGTAGATTACCACTGGAGTcGGGGATCTGTGGTCTCATA anti-sense sequence + target anti-sense sequence) followed by 22 bases
CA3′, where sequence in italic bold is complementary complementary to the last 22 bp upstream of the +1 transcriptional start
to the H1 promoter, XbaI is underlined, the loop is bold site (sense strand) of the polIII promoter (Fig. 1a). The resulting shRNA
expression cassette can be cloned into an A/T sequencing vector
underlined and nucleotide +1 is in small caps. (Fig. 1b), sequenced to confirm hairpin integrity (Fig. 1c), tested for target
(ii) Amplify the silencing cassette. Using P1 and P2 knockdown efficiency as described in Box 1 (Fig. 1d) and subsequently
primers (final primer concentration is 10 µM) subcloned into the lentiviral vector (Fig. 1e).
described above, use 10 ng template pH1 to PCR
amplify the silencing cassette. Use the reaction
conditions described below.
▲ CRITICAL STEP It is essential to add DMSO or other similar agents in order to weaken hydrogen bonding and prevent
formation of hairpin structure. We use Advantage GC-2 polymerase mix (BD) with GC melt additive, or add 7% DMSO to a
regular Taq polymerase reaction to prevent hairpin formation.

Cycle number Denaturation Annealing Polymerization


1 3 min at 94 °C
2–31 30 s at 94 °C 30 s at 55 °C 40 s at 72 °C
32 10 min at 72 °C

236 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS


PROTOCOL
(iii) Clone and sequence the silencing cassette. PCR amplification should result in a ~400 bp fragment. Clone the insert
into an A/T sequencing vector and sequence it, as mutations in the hairpin potentially introduced by PCR can have
considerable impact on silencing efficiency.
(iv) Verify the knockdown efficiency of the silencing cassette as indicated in Box 1.
(v) Transfer the silencing cassette to the lentiviral vector and resequence the hairpin. The insert must be digested with
the XbaI, gel purified, ligated into the lentivector (previously linearized with NheI, dephosphorylated and gel purified)
and transformed. Plasmid DNA from the resulting colonies can be screened by digestion with SspI. The parental vector
should have only one SspI site, whereas the vector containing the insert will acquire an additional SspI site located in
the H1 promoter, resulting in an insert of 1.4 kb or 1.6 kb, depending on orientation (which has no effect on silencing
efficiency). Re-verify the hairpin by sequencing with primer H1-F.
(vi) High titer lentiviral preparations can be prepared as described in ref. 1 and tested for efficiency of target
downregulation (Box 2).
(B) Creation of a lentiviral vector carrying GFP as a marker and cloning the silencing cassette upstream of the GFP
© 2006 Nature Publishing Group http://www.nature.com/natureprotocols

marker. This version of the lentiviral silencing system has been adapted to Gateway cloning technology (Invitrogen),
allowing for a more convenient cloning procedure, as explained in Figure 2.
(i) Select target and design oligonucleotides to generate the hairpin insert. Select a target within the gene to be
silenced (e.g., for human p53: 5′-GACTCCAGTGGTAATCTAC-3′; ref. 41). Cloning of the shRNA will require designing two
complementary oligonucleotides (O1 and O2) and annealing them to generate an insert with BsaI-compatible termini for
subsequent cloning into pENTR/U6, as described below. For example, for the human p53 target sequence given above,
the required oligonucleotides, after annealing, would look as follows, where the sense strand of the hairpin is in italics,
the loop is in bold and the antisense strand of the hairpin is underlined (note the 5′ overhangs present at both ends of
the annealed product).
5′CACCGACTCCAGTGGTAATCTACTTCAAGAGAGTAGATTACCACTGGAGTC 3′
3′CTGAGGTCACCATTAGATGAAGTTCTCTCATCTAATGGTGACCTCAGAAAA5′
(ii) Prepare the hairpin insert. Perform the oligonucleotide annealing procedure by mixing: 5 µl O1 (200 µM), 5 µl O2
(200 µM) and 2 µl 10× annealing buffer, and adjust to 8 µl final volume with H2O. This results in 50 µM final
concentration for each oligonucleotide. Heat the annealing mixture to 94 °C for 5 min and then use a PCR machine to
cool to 25 °C at 0.1 °C/s. Even at 50 µM final concentration, the efficiency of the reaction is only ~50%. Efficiency of
annealing can be ascertained by running an aliquot of the annealed oligonucleotides (5 µωl of a 1/100 dilution of the
annealing mix) on a 4% agarose gel. Single-stranded oligonucleotides will migrate as a hairpin of ~30 bases, whereas
the annealed product will migrate at its predicted size (~60 bp).
? TROUBLESHOOTING
(iii) Ligate the hairpin insert into pENTR/U6. Dilute annealing mix 1:1,000 in H20 and clone into the BsaI linearized pENTR/
U6 vector. A diagram of pENTR/U6 is shown in Figure 2a.
▲ CRITICAL STEP pENTR/U6 has a ccdB toxic gene and must therefore be propagated in tolerant strains such as E. coli
DB3.1. When this plasmid is digested with BsaI (Invitrogen sells it already linearized with BsaI), the vector is left with

BOX 2 VALIDATION OF LENTIVIRAL SILENCING VECTOR SUSPENSIONS


The final validation of high titer viral preparations by transduction of a cell line expressing the target protein, followed by
measuring target expression by RT-PCR, northern or western blot) is crucial. Overexpression of any siRNA can cause some nonspecific
downregulation of gene expression; therefore, inclusion of adequate controls is critical. Appropriate controls are lentiviral vectors
carrying a silencing cassette expressing an shRNA against no known target, a different (nonrelated) target, or lacking a silencing
cassette altogether. Furthermore, any knockdown experiment should show similar knockdown phenotypes using a minimum of two
different shRNAs in order to rule out the possibility that the observed phenotype is due to nonspecific effects. Transduction at
several MOIs should be tested. Precise determination of efficiency of downregulation of the target will require testing homogeneously
transduced cell populations, which can be obtained by transducing at high MOIs or by FACS sorting for GFP-positive cells. The MOI
required to silence a given target will depend on the levels of expression of the target mRNA, siRNA efficiency and the transducibility
of the cell type involved. Typically, proceed as described below:
(i) Seed 105 293T HEK cells in a 24-well plate the day before transduction.
(ii) Add an aliquot of lentiviral silencing vector to the well and mix gently. Return plates to the incubator. GFP fluorescence should
start appearing after 24 h and be very clear by 48 h. Lack of GFP fluorescence indicates a problem during lentiviral production or
that the promoter driving GFP is not efficient in the cell type transduced. The cells may by expanded, passaged and FACS sorted as
required.
(iii) Determine knockdown efficiency by RT-PCR, northern or western blot analysis.

NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 237


PROTOCOL
the following termini, which are compatible with the a Cut pENTR/U6 with BsaI
termini of the insert: BsaI BsaI
XXXXX 3′ 5′ TTTTTXXXXXXX attL1 hU6 ccdB T5 attL2

XXXXXGTGG 5′ 3′ AXXXXXXX BsaI BsaI


T5 attL2
Typically, 50 ng of vector are ligated to 2–3 µl of a attL1 hU6

1/1,000 dilution of the annealed product. Transform


competent bacteria and select on LB-kanamycin plates. b Anneal O1/O2
shRNA
? TROUBLESHOOTING
(iv) Analyze transformants. Resulting plasmids will have
the structure shown in Figure 2c; plasmid DNA from c Clone shRNA into pENTR/U6

colonies can be digested with NdeI-XbaI and run on a attL1 hU6 shRNA T5 attL2

4% agarose gel. Clones containing inserts will show a


band of 127 bp compared to a 76 bp band for clones
© 2006 Nature Publishing Group http://www.nature.com/natureprotocols

d Sequence
lacking an insert. The nucleotide sequence of the
shRNA insert should be determined with hU6-F and e Verify KD efficiency (Box 1)
M13-R primers to check hairpin integrity.
(v) Test the silencing cassettes for silencing efficiency as
f Perform LR clonase reaction
explained in Box 1.
(vi) Perform the LR recombination reaction. The L-CMV-GFP attL1 hU6 shRNA T5 attL2

destination vector is shown in Figure 3e. Initially


set up the LR recombination reaction by mixing RSV attR1 ccdB attR2 cPPT CMV GFP WPRE 3' LTR

100–300 ng of entry vector, 150 ng of destination


vector and 4 µl of 5× LR buffer, and making up to 20 µl RSV attB1 hU6 shRNA T5 attB2 cPPT CMV GFP WPRE 3' LTR

final volume with H2O. Incubate at room temperature


(22 – 26°C) overnight. Add 2 µl of Proteinase K (2
Figure 2 | Cloning scheme for Option B. The pENTR/U6 vector is linearized
µg/µl) mg/ml) and incubate 10 min at 37 °C. Transform with BsaI (Fig. 3a). An shRNA hairpin is created by annealing (Fig. 3b)
STBL3 (or equivalent recombination deficient strain) and cloned into pENTR/U6 (Fig. 3c). This procedure results in a pENTR/u6
competent bacteria with 2 µl of LR reaction mix. plasmid containing a hU6 driven silencing cassette flanked by recombination
▲ CRITICAL STEP Use of nonrecombination-deficient sites from phage lambda (attL1 and attL2). The silencing cassette should
bacteria can result in unwanted recombination events be sequenced to confirm hairpin integrity (Fig. 3d), tested for target
knockdown efficiency as described in Box 1 (Fig. 3e) and subsequently
within the plasmid). Plate on LB-ampicillin plates. transferred to the lentiviral destination vector (L-Dest-CMV-GFP, a lentiviral
? TROUBLESHOOTING vector with a Gateway destination cassette (phage lambda attR1 and attR2
(vii) Analyze resulting clones. The resulting constructs will recombination sites flanking a ccdB toxic gene) cloned upstream of the GFP
have the structure depicted in Figure 2f, and plasmid marker) by performing an LR clonase reaction (Fig. 3f). Further information
DNA can be digested with ClaI-AvaI. Clones containing on the Gateway Cloning system can be found at http://www.invitrogen.com.
the silencing cassette should have a ~700 bp insert,
whereas the unrecombined parental destination vector
will show a 1.9 kb insert. Sequence positive clones with hU6-F primer to check hairpin integrity.
? TROUBLESHOOTING
(viii) High-titer lentiviral preparations can be prepared as described in ref. 1 and tested for silencing efficiency (Box 2).

● TIMING
Option A:
Day 1: select target and design primers
Day 2: amplify silencing cassette
Days 3–9: clone and sequence the silencing cassettes
Days 10–13: verify knockdown efficiency
Days 14–19: transfer silencing cassette to the lentiviral vector and sequence

Option B:
Day 1: select target and design oligonucleotides
Day 2: prepare hairpin insert
Days 3–8: clone and sequence silencing cassettes
Days 9–12: verify knockdown efficiency
Days 13–18: transfer silencing cassette to lentivector and sequence

238 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS


PROTOCOL
? TROUBLESHOOTING
See Table 1.

TABLE 1 | Troubleshooting table.


STEP PROBLEM SOLUTION
1Bii Low oligonucleotide annealing efficiency may be caused Purify the oligonucleotides by ethanol precipitation
by excess salt in the oligonucleotide stock solution
1Biii Absence of kanamycin-1 resistant colonies may be due to Analyze annealing efficiency in Step 1Bii by agarose gel
low ligation efficiency caused by low annealing efficiency electrophoresis
1Bvi Absence of colonies may be due to excess volume of Adhere to the recommended ligation volume (2 µl of ligation
ligation reaction, especially if the PEG-based ligation reaction in 100 µl competent cell suspension) added to the
buffer provided with the LR clonase kit is used transformation
© 2006 Nature Publishing Group http://www.nature.com/natureprotocols

1Bvii Unexpected restriction digest patterns in the ampicillin-


resistant colonies may have two causes:
a) Erroneous use of kanamycin instead of ampicillin, a) Use the correct antibiotic selection (ampicillin)
resulting in growth of colonies harboring pENTR/U6
instead of L-Dest-CMV-GFP
b) Use of bacterial strains that are not recombination b) Adhere to the recommended STBL3 (or equivalent recombination-
deficient deficient) E. coli competent bacteria; if recombination problems
persist, grow the transformation plates and the individual colony DNA
minipreparations at 30 °C, avoiding growth into stationary phase

ANTICIPATED RESULTS a UT L318 L339 b UT L318 L339


The protocols described here are very MOI 0 10 10 MOI 0 10 100 10 100
robust; adherence to the recommended htt
procedures should result in relatively
GFP
problem-free cloning. Lentiviral 28S
silencing vectors described in this
protocol routinely result in a high c
degree of knockdown (>90%) both in
htt
vitro and in vivo. An example of in vitro htt

results is given in Figure 3. p300

Figure 3 | An example of an efficient lentiviral silencing vector. A lentiviral silencing vector was
developed (Option B) to express an shRNA against primate huntingtin (L339) or an shRNA against no
known target (L318). A primate cell line was transduced with MOI of 0 (untransduced control), 10 or 100.
Cells were expanded during six consecutive passages and FACS-sorted for GFP+ cells. Levels of endogenous
huntingtin in all three lines were assessed by (a) imunohistochemistry, (b) northern blot and (c) western
blot. The 28S ribosomal band and p300 are shown as loading controls in b and c, respectively.

ACKNOWLEDGMENTS The authors wish to thank Kenneth Fringpong and Knut 3. Naldini, L. et al. In vivo gene delivery and stable transduction of nondividing
Madden of Invitrogen Corporation for their assistance in development of the L- cells by a lentiviral vector. Science 272, 263–267 (1996).
pDest-cmv-gfp vector. 4. Pfeifer, A., Ikawa, M., Dayn, Y. & Verma, I.M. Transgenesis by lentiviral
vectors: lack of gene silencing in mammalian embryonic stem cells and
COMPETING INTERESTS STATEMENT The authors declare that they have no preimplantation embryos. Proc. Natl. Acad. Sci. USA 99, 2140–2145 (2002).
competing interests. 5. Lois, C., Hong, E.J., Pease, S., Brown, E.J. & Baltimore, D. Germline
transmission and tissue-specific expression of transgenes delivered by
Published online at http://www.natureprotocols.com/ lentiviral vectors. Science 295, 868–872 (2002).
Reprints and permissions information is available online at http://npg.nature. 6. Miyoshi, H., Blomer, U., Takahashi, M., Gage, F.H. & Verma, I.M. Development
com/reprintsandpermissions/ of a self-inactivating lentivirus vector. J. Virol. 72, 8150–8157 (1998).
7. Zufferey, R. et al. Self-inactivating lentivirus vector for safe and efficient in
1. Tiscornia, G., Singer, O. and Verma, I.M. Production and purification of vivo gene delivery. J. Virol. 72, 9873–9880 (1998).
lentiviral vectors. Nat. Protocols 1, 241–245 (2006). 8. Denli, A.M. & Hannon, G.J. RNAi: an ever-growing puzzle. Trends Biochem.
2. Trono, D. Lentiviral Vectors (Berlin-Heidelberg, Springer-Verlag, 2002). Sci. 28, 196–201 (2003).

NATURE PROTOCOLS | VOL.1 NO.1 | 2006 | 239


PROTOCOL
9. Du, T. & Zamore, P.D. microPrimer: the biogenesis and function of microRNA. silencing using micro-RNA designed hairpins. Rna 8, 842–850 (2002).
Development 132, 4645–4652 (2005). 26. Dudley, N.R. & Goldstein, B. RNA interference: silencing in the cytoplasm and
10. Chen, P.Y. & Meister, G. microRNA-guided posttranscriptional gene nucleus. Curr. Opin. Mol. Ther. 5, 113–117 (2003).
regulation. Biol. Chem. 386, 1205–1218 (2005). 27. Pe’ery, T., Mathews, M.B. & Baulcombe, D. RNA interference. Methods 30,
11. Lund, E., Guttinger, S., Calado, A., Dahlberg, J.E. & Kutay, U. Nuclear export 287–288 (2003).
of microRNA precursors. Science 303, 95–98 (2004). 28. Cullen, B.R. Transcription and processing of human microRNA precursors. Mol.
12. Kim, V.N. MicroRNA precursors in motion: exportin-5 mediates their nuclear Cell 16, 861–865 (2004).
export. Trends Cell Biol. 14, 156–159 (2004). 29. Gregory, R.I., Chendrimada, T.P., Cooch, N. & Shiekhattar, R. Human RISC
13. Lee, Y. et al. The nuclear RNase III Drosha initiates microRNA processing. couples microRNA biogenesis and posttranscriptional gene silencing. Cell
Nature 425, 415–419 (2003). 123, 631–640 (2005).
14. Fjose, A., Ellingsen, S., Wargelius, A. & Seo, H.C. RNA interference: 30. Zeng, Y. & Cullen, B.R. Sequence requirements for micro RNA processing and
mechanisms and applications. Biotechnol. Annu. Rev. 7, 31–57 (2001). function in human cells. Rna 9, 112–123 (2003).
15. Hannon, G.J. RNA interference. Nature 418, 244–251 (2002). 31. Zeng, Y. & Cullen, B.R. Structural requirements for pre-microRNA binding and
16. Elbashir, S.M. et al. Duplexes of 21-nucleotide RNAs mediate RNA nuclear export by Exportin 5. Nucleic Acids Res. 32, 4776–4785 (2004).
interference in cultured mammalian cells. Nature 411, 494–498 (2001). 32. Makinen, P.I. et al. Stable RNA interference: comparison of U6 and H1
17. Oliveira, D.M. & Goodell, M.A. Transient RNA interference in hematopoietic promoters in endothelial cells and in mouse brain. J. Gene Med. (2006).
© 2006 Nature Publishing Group http://www.nature.com/natureprotocols

progenitors with functional consequences. Genesis 36, 203–208 (2003). 33. Myslinski, E., Ame, J.C., Krol, A. & Carbon, P. An unusually compact external
18. Brummelkamp, T.R., Bernards, R. & Agami, R. A system for stable expression promoter for RNA polymerase III transcription of the human H1RNA gene.
of short interfering RNAs in mammalian cells. Science 296, 550–553 (2002). Nucleic Acids Res. 29, 2502–2509 (2001).
19. Miyagishi, M. & Taira, K. U6 promoter-driven siRNAs with four uridine 3� 34. Amarzguioui, M. & Prydz, H. An algorithm for selection of functional siRNA
overhangs efficiently suppress targeted gene expression in mammalian cells. sequences. Biochem. Biophys. Res. Commun. 316, 1050–1058 (2004).
Nat. Biotechnol. 20, 497–500 (2002). 35. Jagla, B. et al. Sequence characteristics of functional siRNAs. Rna 11, 864–
20. Rubinson, D.A. et al. A lentivirus-based system to functionally silence 872 (2005).
genes in primary mammalian cells, stem cells and transgenic mice by RNA 36. Reynolds, A. et al. Rational siRNA design for RNA interference. Nat.
interference. Nat. Genet. 33, 401–406 (2003). Biotechnol. 22, 326–330 (2004).
21. Tiscornia, G., Singer, O., Ikawa, M. & Verma, I.M. A general method for gene 37. Santoyo, J., Vaquerizas, J.M. & Dopazo, J. Highly specific and accurate
knockdown in mice by using lentiviral vectors expressing small interfering selection of siRNAs for high-throughput functional assays. Bioinformatics 21,
RNA. Proc. Natl. Acad. Sci. USA 100, 1844–1848 (2003). 1376–1382 (2005).
22. Paddison, P.J., Caudy, A.A., Bernstein, E., Hannon, G.J. & Conklin, D.S. Short 38. Ui-Tei, K. et al. Guidelines for the selection of highly effective siRNA
hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian sequences for mammalian and chick RNA interference. Nucleic Acids Res 32,
cells. Genes Dev. 16, 948–958 (2002). 936–948 (2004).
23. Stegmeier, F., Hu, G., Rickles, R.J., Hannon, G.J. & Elledge, S.J. A lentiviral 39. Clemens, M.J. & Elia, A. The double-stranded RNA-dependent protein kinase
microRNA-based system for single-copy polymerase II–regulated RNA PKR: structure and function. J. Interferon Cytokine Res. 17, 503–524 (1997).
interference in mammalian cells. Proc. Natl. Acad. Sci. USA 102, 13212– 40. Boden, D. et al. Promoter choice affects the potency of HIV-1-specific RNA
13217 (2005). interference. Nucleic Acids Res. 31, 5033–5038 (2003).
24. Vaucheret, H. MicroRNA-dependent trans-acting siRNA production. Sci. STKE 41. Brummelkamp, T.R., Bernards, R. & Agami, R. A system for stable expression
300, pe43 (2005). of short interfering RNAs in mammalian cells. Science 296, 550–553 (2002).
25. McManus, M.T., Petersen, C.P., Haines, B.B., Chen, J. & Sharp, P.A. Gene 42. Castanotto, D., Li, H. & Rossi, J.J. Functional siRNA expression from
transfected PCR products. Rna 8, 1454–1460 (2002).

240 | VOL.1 NO.1 | 2006 | NATURE PROTOCOLS

You might also like