Sunteți pe pagina 1din 9

Gene Therapy (2017) 24, 453–461

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 0969-7128/17
www.nature.com/gt

ORIGINAL ARTICLE
Silencing of HIV-1 by AgoshRNA molecules
E Herrera-Carrillo, A Harwig and B Berkhout

RNA interference (RNAi) is a sequence-specific gene silencing mechanism that is triggered by the expression of a short hairpin RNA
(shRNA). shRNA molecules enter the RNAi pathway at the Dicer processing step. Recent studies indicated that the cellular microRNA
miR-451 is not recognized by Dicer, but that it is processed instead by the Argonaute 2 (Ago2) protein. Subsequently, Dicer-
independent shRNAs were described that rely on Ago2 for processing, as well as the subsequent silencing step. We called
these AgoshRNA molecules because they depend on Ago2 both for maturation and activation. Processing of an AgoshRNA yields
only a single active RNA strand, thus reducing the chance of adverse off-target effects induced by the passenger strand of regular
shRNAs. In this study, we converted several anti-HIV-1 shRNAs into AgoshRNAs. Seven of the 21 designed AgoshRNAs were potent
anti-HIV molecules, although their RNAi activity is generally somewhat reduced compared with the matching shRNAs. The
AgoshRNA candidates revealed no cellular toxicity. This may relate to the absence of passenger strand expression, which was
verified for these AgoshRNA candidates. Furthermore, we demonstrate that a toxic shRNA can be converted into a non-toxic
AgoshRNA.

Gene Therapy (2017) 24, 453–461; doi:10.1038/gt.2017.44

INTRODUCTION shift to Ago2-mediated processing (bottom panel). Dicer


The cellular RNA interference (RNAi) mechanism uses small, non- generates the duplex small interfering RNA (siRNA) consisting
coding RNA molecules to regulate gene expression at the of the guide and passenger strands, both with potential
posttranscriptional level.1–4 The canonical microRNA (miRNA) path- silencing activity (marked white-5′ and black-3′). In contrast,
way employs the Drosha endonuclease to liberate the pre-miRNA Ago2 generates a single-guide RNA strand that upon poly(A)-
hairpin from the primary transcript. The pre-miRNA is transported specific ribonuclease processing yields the AgoshTRIM molecule
by Exportin-5 to the cytoplasm, where the Dicer endonuclease (marked gray).
removes the loop of the hairpin to generate an active miRNA There is accumulating evidence that the AgoshRNA pathway is
duplex, which subsequently associates with the Argonaute 2 (Ago2) very similar to that of miR-451. One of the hallmarks of miR-451 is
protein forming the RNA-induced silencing complex to induce the 5′-terminal A residue instead of a more common U, and this A
degradation of complementary target mRNAs.5–7 Designed short remains unpaired at the bottom of the hairpin stem. A recent
hairpin RNA (shRNA) molecules enter the RNAi pathway at the Dicer study indicated that the identity of the bottom bp is also
processing step, thus avoiding the Drosha processing step. important for AgoshRNA activity.13 We subsequently performed a
Recent studies identified Dicer-independent RNAi pathways, more in-depth analysis of the bottom bp, which includes variation
in which the unusually short miR-451 hairpin of just 17 base of the 5′-end nt and thus the transcription start site, to facilitate
pairs (bp) is processed by Ago2 instead of Dicer.8–10 Ago2 binds the design of optimized AgoshRNA molecules.19 We concluded
the duplex and cleaves on the 3′ side between bp 10 and 11 to that AgoshRNA optimization is possible by introduction of a
generate a single, extended ~ 30 nucleotides (nt) guide strand. bottom mismatch with 5′-terminal A or G. The latter purine
In contrast, regular miRNAs generate a duplex of guide and requirement also has a role in optimized transcription by the H1
passenger strand, of which the latter molecule may cause polymerase III promoter.19,20
unwanted side effects. Subsequent 3′-end processing of A clear advantage of the AgoshRNA design over shRNAs is the
miR-451 by poly(A)-specific ribonuclease creates the ~ 22–26 lack of possible off-target effects that are caused by a passenger
nt mature miRNA, but this modification appears non-essential strand. We listed additional advantages of AgoshRNA
for activity.11 Short shRNAs that resemble the unique miR-451 inhibitors,21 for example, the activity in monocytes that express
molecule are also recognized by Ago2 instead of Dicer.12–18 We very little or no Dicer.22 We recently also addressed the
named these molecules AgoshRNA because Ago2 is involved in important mechanistic question whether AgoshRNAs trigger
both the processing and silencing steps.15 Compared with a cleavage or translational suppression of a perfect complemen-
regular shRNA, the silencing activity switches from a 3′ side- tary mRNA.19 Precise cleavage at the predicted site in the target
encoded guide to a single, extended 5′ side-encoded guide mRNA was observed. In fact, the position of AgoshRNA-mediated
strand. Deep sequencing confirmed 3′ side cleavage between mRNA cleavage was more accurate than cleavage by a regular
bp 10 and 11.14 The regular shRNA and the alternative shRNA, which is likely due to imprecise Dicer-mediated proces-
AgoshRNA processing routes are depicted in Figure 1a. Regular sing of the shRNA, a step that is circumvented by the AgoshRNA
shRNAs are processed by Dicer (top panel), but shorter design. We now converted previously validated and potent anti-
AgoshRNA hairpins (⩽18 bp) escape from Dicer cleavage and HIV-1 shRNA molecules into AgoshRNAs. We demonstrate that

Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of
Amsterdam, Amsterdam, The Netherlands. Correspondence: Professor B Berkhout, Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection
and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam Meibergdreef 15, Amsterdam, 1105 AZ Noord-Holland, The Netherlands.
E-mail: b.berkhout@amc.uva.nl
Received 13 January 2017; revised 13 April 2017; accepted 12 May 2017; accepted article preview online 29 May 2017; advance online publication, 29 June 2017
Antiviral AgoshRNA design
E Herrera-Carrillo et al
454

Figure 1. Anti-HIV shRNAs and AgoshRNAs. (a) In the canonical pathway (top) the shRNA stem is cleaved by Dicer into an siRNA duplex of
~ 21 bp with 3′ UU overhang that is loaded into RNA-induced silencing complex. The 5′ passenger (white arrow) is cleaved and degraded and
the 3′ guide strand (black arrow) acts in RNAi silencing. In the non-canonical pathway (bottom), the AgoshRNA is cleaved by Ago2 on the
3′ side between base pairs 10 and 11 into an extended guide of ~ 33 nt (gray arrow). AgoshRNA subsequently may instruct Ago2 for RNAi
silencing or may be trimmed by poly(A)-specific ribonuclease (PARN) to create a ~ 24 nt guide named AgoshTRIM. The predicted Dicer and
Ago2 cleavage sites are marked with black and gray arrows, respectively. (b) Secondary structure of the R/T5 shRNA and AgoshRNA as
predicted by Mfold, with the guide boxed in gray. The 5′-end nucleotide of AgoshRNA constructs and the basepairing partner were replaced
by A C. (c) The HIV-1 genome showing the position of all target sites. (d) HIV-1 target sequences. We designed two or even three AgoshRNAs
for targets with sequences that are highly conserved among HIV-1 isolates.

an active, but toxic shRNA antiviral can be reconfigured into an RESULTS


active and non-toxic AgoshRNA that targets the same HIV-1 RNA Design of anti-HIV AgoshRNA molecules
sequence. We also show that these AgoshRNA inhibitors, unlike Figure 1b depicts the secondary RNA structure of a potent antiviral
shRNA inhibitors, remain active in Dicer-minus monocytic cells shRNA (shR/T5) and the newly designed AgoshRNA (AgoshR/T5) as
that are host cells for HIV-1 infection. These findings will aid in predicted by Mfold. Both molecules encode an identical anti-HIV
the future design of an active and safe AgoshRNA-based gene guide on their 3′ and 5′ side, respectively (marked by a gray box).
therapy against HIV-1 infection. The predicted Dicer and Ago2 cleavage sites are indicated,

Gene Therapy (2017) 453 – 461 © 2017 Macmillan Publishers Limited, part of Springer Nature.
Antiviral AgoshRNA design
E Herrera-Carrillo et al
455
respectively. We replaced the bottom bp in the AgoshRNA
molecule by an unpaired A C for optimal AgoshRNA activity.19
These small RNAs were synthesized from the H1 polymerase III
promoter, which requires a purine at the +1 position for optimal
transcription. We selected A over G because the MID domain of
the human Ago2 protein prefers to load small RNAs with a 5′-end
U or A in the RNA-induced silencing complex.23,24 We avoided
stretches of four or more Us in the AgoshRNA sequence because a
polyU tract acts as a termination signal for RNA polymerase III
transcription. As a favorite delivery vehicle for an AIDS gene
therapy is the HIV-based lentiviral vector, we avoided anti-HIV-1
guide sequences that trigger ‘self’-targeting of the JS1 lentiviral
vector.25,26
Over the years, we designed several sets of anti-HIV shRNAs
based on high conservation of the viral target sequence among
different virus isolates.27,28 Nearly 100 shRNA candidates were
tested for in vitro antiviral activity against the primary HIV-1 LAI
isolate,29 which is the prototype virus strain used in our
laboratory and a few shRNAs have progressed to in vivo tests.30
Four shRNAs (shPol1, shPol47, shR/T5 and shGag5) were
selected because of robust and durable inhibition of HIV-1
replication. However, shGag5 was subsequently reported to
exhibit a negative effect on T cell physiology in a competitive
cell growth assay in vitro and in vivo in the humanized immune
system mouse.30 In this study, we designed a set of AgoshRNA
mimics of this pre-selected set of potent shRNAs. The
conversion of shRNA into AgoshRNA molecules is not likely to
be easy because of the different processing routes of these
molecules. We thus wondered whether potent shRNAs can
indeed be converted into AgoshRNAs without losing antiviral Figure 2. Inhibition of HIV-1 production by AgoshRNA constructs.
potency. We also addressed whether the toxicity of shGag5 is (a) HEK293T cells were co-transfected with 250 ng of the HIV-1 pLAI,
removed in the AgoshRNA design that does not generate a 1 ng of Renilla luciferase plasmid (pRL) and 25 ng of the AgoshRNA
passenger strand. constructs. Two days post-transfection, inhibition of HIV-1 produc-
tion was determined by measuring CA-p24 levels in the culture
In addition, we evaluated AgoshRNAs against critically impor- supernatant. CA-p24 values were normalized to the Renilla luciferase
tant and highly conserved viral sequences that are perhaps not activities. The ratio between the CA-p24 level and the Renilla
allowed to mutate: the primer-binding site and the polypurine luciferase activity in the presence of 25 ng pBS control was set at
tract (PPT) and domains in the untranslated leader region (UTR-A 100%. Bars represent the average values from eight independent
and UTR-B). Early attempts to raise potent shRNAs against such transfections and error bars shows the s.d. (b) Dose-dependent
theoretically ideal targets failed, in part because of repressive RNA inhibition of HIV-1 production by the most potent AgoshRNA
structure.28,31 Figures 1c and d depict all target sites in the HIV-1 constructs. HEK293T cells were co-transfected with HIV-1 pLAI, pRL
and increasing amount of AgoshRNA constructs (1, 5 and 25 ng).
genome and the actual nt sequences, respectively. In total, we pBS control was set at 100%. The mean values and s.d. are based on
designed and tested 21 anti-HIV AgoshRNAs (Supplementary six independent transfections. Statistical analyses (one-way analysis
information). When conservation of the HIV-1 target was extended of variance followed by Tukey’s post-hoc tests) were performed and
into flanking sequences, we designed multiple overlapping differences among groups were considered significant when the
AgoshRNAs. For instance, the Gag target starting at position correspondent P-value was below 0.05.
1364 in HIV-1 LAI allowed the design of three overlapping
AgoshRNAs, AgoshGag4–6 (Figure 1d).
We selected the most potent AgoshRNAs (Gag4-6, Pol1,
Antiviral activity of the AgoshRNA inhibitors in transient assays Pol8, Pol45 and R/T5) for a titration in human embryonic
kidney 293T (HEK293T) cells co-transfected with the
Each AgoshRNA expression construct (25 ng) was co-transfected HIV-1 pLAI molecular clone, the Renilla plasmid and an
in HEK293 T cells with the HIV-1 molecular clone pLAI (250 ng) to increasing amount of the AgoshRNA constructs (1, 5 and
score inhibition of virus production. A fixed amount of Renilla 25 ng, Figure 2b). The results confirm the inhibitory potency of
luciferase plasmid was included to control for variation in the these seven AgoshRNA molecules, which act in a dose-
transfection efficiency. Two days post-transfection, HIV-1 produc- dependent manner.
tion was measured by the CA-p24 level in the culture supernatant, We next compared the AgoshRNA versus shRNA activity on
which was corrected for the Renilla luciferase activity (Figure 2a). matching luciferase reporters with the sense HIV-1 target
Virus production in the presence of 25 ng pBluescript (pBS) control sequence and a control encoding the antisense HIV-1 sequence
plasmid was set at 100%. Seven of the 21 AgoshRNAs showed a (Figure 3a). The HIV-sense reporter detects the activity of the
significant inhibitory activity (Gag4-6, Pol1, Pol8, Pol45 and R/T5) shRNA guide strand (black arrow) and the extended guide
with virus production levels dropping to o 50% versus the pBS strand from the 5′ side of AgoshRNA (gray arrow), whereas the
control (0.0001 ⩽ P o 0.01). The other 14 AgoshRNA candidates HIV-antisense reporter will score any shRNA passenger strand
showed little inhibitory activity (P40.05). Among the inactive activity (white arrow). HEK293T cells were co-transfected using
AgoshRNAs, 10 mimic shRNAs that are potent (Pol2, Pol6, Pol7, 25 ng of inhibitor and the luciferase activity was measured after
Pol9, Pol41, Pol42, Pol44, Pol47, R/T4 and Nef). Four inactive two days in cell lysates. We set the activity measured with the
AgoshRNAs target ideally conserved HIV-1 sequences (PBS, PPT, unrelated shNef construct at 100% (Figure 3b). All seven
UTR-A and UTR-B) that also resisted shRNA attack.28 AgoshRNAs produce 5′ guide strands that target the

© 2017 Macmillan Publishers Limited, part of Springer Nature. Gene Therapy (2017) 453 – 461
Antiviral AgoshRNA design
E Herrera-Carrillo et al
456

Figure 3. Luciferase knockdown assays by AgoshRNAs. (a) Luciferase reporter constructs with the sense and antisense HIV-derived sequences.
The Luc-sense reporter scores canonical shRNA guide activity and AgoshRNA activity. The Luc-antisense reporter can theoretically detect both
shRNA (21 nt) and AgoshRNA passenger strand (10 nt) activity. (b) Luciferase knockdown was determined by co-transfection of the reporters
with the AgoshRNA constructs. HEK293T cells were co-transfected with 100 ng of the respective firefly luciferase reporter plasmid, 1 ng of
Renilla luciferase plasmid and 25 ng of the AgoshRNA constructs, which were compared with the original shRNA constructs with identical
HIV-1 target sequence. An irrelevant shRNA (shNef ) served as negative control, for which the activity was set at 100% luciferase expression.
The Luc-sense results are shown in black bars and the Luc-antisense results in white bars scores shRNA passenger strand activity. The mean
values and s.d. are based on six independent transfections. Statistical analysis (one-way analysis of variance followed by Tukey’s post-hoc tests)
demonstrated that luciferase expression in the presence of antiviral AgoshRNAs differed significantly from luciferase expression measured
with the shNef control (ns P40.05, *P ⩽ 0.05, **P ⩽ 0.01, ***P ⩽ 0.001 and ****P ⩽ 0.0001).

corresponding HIV-sense reporter. No passenger strand activity comparison yielded a mixed outcome. The inhibitory potency
was measured as predicted for the AgoshRNA design. However, was increased for AgoshGag4, AgoshGag6 and AgoshPol8
passenger strand activity was measured for all regular shRNAs versus the corresponding shRNAs, but AgoshGag5 and Agosh-
on the respective HIV-antisense reporters, with relative lucifer- Pol45 were less potent than the matching shRNAs. AgoshPol1
ase expression levels dropping to between 20 and 80% of the and AgoshR/T5 showed similar inhibitory potential as the
uninhibited value. The AgoshGag4–6 set reduced the relative corresponding shRNAs, with luciferase expression levels of the
luciferase expression level to o 30%. The shRNA-AgoshRNA HIV-sense reporter dropping to o 40%.

Gene Therapy (2017) 453 – 461 © 2017 Macmillan Publishers Limited, part of Springer Nature.
Antiviral AgoshRNA design
E Herrera-Carrillo et al
457

Figure 4. Northern blotting of the AgoshRNA processing products.


Processing of the AgoshGag4–6 variants and the corresponding
shRNA set was analyzed by northern blot analysis with (a) the ‘guide’
probe and (b) the ‘passenger’ probe. Ethidium bromide staining of
small rRNAs and tRNAs are shown as loading controls below
the blot.

Intracellular processing of shRNA/AgoshRNA inhibitors


The range of silencing potencies observed for the anti-HIV
AgoshRNAs might be due to altered RNAi activity, but could also
reflect differences in RNA processing by Ago2 or intracellular RNA
stability. Northern blot analysis revealed intracellular processing of
the overlapping AgoshRNA triplet (AgoshGag4-6) and the
corresponding shRNAs set (shGag4-6). Processing of the 3′ guide
strand of regular shRNAs and the 5′ guide strand of AgoshRNAs
variants was analyzed by northern blot analysis with a ‘guide’
probe (Figure 4a), while processing of the 5′ passenger strand of Figure 5. Kinetics of HIV-1 LAI replication in AgoshRNA-expressing
regular shRNAs was detected by the ‘passenger’ probe (Figure 4b). cells. Six parallel infections per AgoshRNA-expressing SupT1 cells
The most prominent ~ 21 nt fragment was observed for shGag5, were performed. The empty lentiviral vector JS1 served as the
followed by shGag4 and shGag6, consistent with the ~ 2-fold negative control in all three panels. CA-p24 antigen was measured
improved luciferase knockdown activity of shGag5. The guide starting at day three post-infection up to day 56.
probe also detected the ~ 30 nt guide strand for the AgoshRNA
constructs and the trimmed products (~24 nt fragments). The We will first describe the results for AgoshGag4. It took around
differential luciferase activity scored for the AgoshGag4-6 variants 21–56 days for viral escape to occur in five of six AgoshGag4
correlated with the amount of guide RNA produced. The cultures (Figure 5, upper panel). We screened for the selection of
AgoshGag6 guide is ~ 2-fold more abundant and consequently truly AgoshRNA-resistant HIV-1 variants by passage of cell-free
more active than the guide strand of AgoshGag4 and AgoshGag5. virus on AgoshRNA-expressing SupT1 cells and control cells
The passenger probe detected a prominent Dicer product of (untransduced). For all five escape cultures, the passaged virus
~ 21 nt fragment exclusively for shGag5, which may relate to the replicated equally well on these cells (data not shown), confirming
toxicity induced by this construct.30 Some striking exceptions that a resistant virus was selected. Next, the 18 nt HIV-1 targets
were also apparent, for example, shGag4 produces much more and flanking regions were analyzed by population-based sequen-
guide RNA than AgoshGag4 (Figure 4a), although the order is cing. Point mutations within the target sequence (A3G, 2 × A6G
reversed in the luciferase assay (Figure 3b). We currently do not and C14U) were detected in four of the five cultures, confirming
fully understand these intricacies. the pressure imposed by the AgoshGag4 inhibitor. It may take
As predicted, none of the AgoshRNAs generated a passenger longer for resistance mutations to become visible in the
strand. The 10 nt product of processed AgoshRNAs is never population-based sequence of the fifth culture.
detected on northern blottings, possibly because it is rapidly AgoshGag5 and AgoshGag6 challenged cultures yielded a CA-
degraded.15 p24-positive supernatant around day 12–28. The phenotype test
indicated that the restricted cells were in fact partially permissive
to replication of the LAI isolate and we scored only wild-type (WT)
Antiviral activity in stably transduced T cells and virus escape
target sequences. We previously described similar breakthrough
options
replication of the WT virus in experiments with poorly active
The anti-HIV-1 AgoshRNAs (Gag4-6, Pol1, Pol8, Pol45 and R/T5) shRNA inhibitors.32 The same phenomenon was observed for
were stably expressed in SupT1 T cells upon transduction with the AgoshPol8 cells (peak infection around day 18–39) and Agosh-
corresponding lentiviral vectors (LV-GFP) to allow HIV-1 replication Pol1/AgoshPol45/AgoshR/T5 cells (peak infection around day 12).
studies. GFP-positive cells were selected and challenged with the Although some delay of virus replication was observed compared
HIV-1 LAI isolate (0.01 ng CA-p24). Six parallel infections were to the JS1 control, the viral phenotype test indicated that
performed for each AgoshRNA to monitor virus evolution, which is restricted cells were permissive to replication of the passaged
a chance process. CA-p24 antigen was measured starting at day 3 virus and only WT target sequences were detected by sequencing
post infection and we followed the spreading infection for (Figure 5, intermediate and bottom panel). We think that no
56 days. The replication curves were grouped by target gene in resistant virus was selected because the AgoshRNAs do not put
three graphs, with JS1—the empty LV—as negative control sufficient selective pressure on WT HIV-1.
(Figure 5). Virus replication was delayed in all AgoshRNA- Many effects were consistently observed among the different
expressing cells compared with JS1 cells, with differential assays, but we also want to mention some discrepancies in
inhibitory activity among the AgoshRNA constructs. AgoshRNA activity. For instance, only AgoshGag4 was potent in

© 2017 Macmillan Publishers Limited, part of Springer Nature. Gene Therapy (2017) 453 – 461
Antiviral AgoshRNA design
E Herrera-Carrillo et al
458

Figure 6. Anti-HIV activity of AgoshGag4 in monocyte-derived cells. Six parallel infections were performed for AgoshGag4 and the
corresponding shGag4 inhibitor in Dicer- minus (THP-1) and Dicer-positive (SupT1) cells. The empty lentiviral vector JS1 served as the negative
control. CA-p24 antigen was measured starting at day 3 post-infection up to day 25.

Figure 7. Competitive cell growth assay. SupT1 T cells were transduced with the lentiviral vector expressing shRNA or AgoshRNA at a
multiplicities of infection of 0.15 or 1.5, yielding a cell population with ~ 20 and 80% GFP-positive cells, respectively. The cells were passaged
and analyzed by fluorescence-activated cell sorting (FACS) measurement twice a week up to 50 days. JS1 represents the empty lentiviral
vector.

the assay of Figure 5, but AgoshGag6 was about twofold better in 5 ng HIV-1 LAI by means of spinoculation in six parallel cultures.
the initial tests in Figures 2 and 3. Likewise, AgoshGag6 is more SupT1 T cells were transduced and GFP-positive cells were
efficient than AgoshGag4 in transient assays, but this advantage selected for subsequent virus challenge with 0.01 ng HIV-1 LAI.
disappears in long-term assays. We cannot provide a full Cells with the empty LV JS1 served as negative control. CA-p24
explanation for these differences, but the most likely explanation antigen was measured starting at day 3 post-infection and we
is differential RNA processing or activity in different cell types. followed the spreading infections for 25 days, after which reduced
cell surface expression of the CD4 receptor occurs in THP-1 cells.37
AgoshRNAs maintain their activity in Dicer-minus cells shGag4 showed antiviral activity in Dicer-positive cells, but lost its
HIV-1 inhibitory potency in Dicer-minus THP-1 cells (Figure 6).
We next wanted to probe sustained antiviral activity of
AgoshGag4 demonstrated equal antiviral activity in Dicer-minus
AgoshGag4 versus shGag4 in cells that do not express Dicer.
THP-1 cells versus Dicer-positive SupT1 T cells, confirming this
The THP-1 cell line originated from a heterogeneous population of
AgoshRNA advantage.
tumor cells that were cultured from the blood of a patient with
acute monocytic leukaemia.33 These cells maintain monocyte
characteristics and do not express Dicer. Differentiation into AgoshRNA toxicity tests in a human T-cell line
mature macrophage-like cells can be induced by phorbol The main theoretical advantage of the AgoshRNA design over a
12-myristate 13-acetate, resulting in Dicer expression.33 This regular shRNA is the absence of a passenger strand that can
would be the ideal Dicer-positive control, but this phenotype is induce adverse effects. This was tested by AgoshRNA-
not stable and cells will undergo dedifferentiation.34 Furthermore, transformation of the shGag5 antiviral, which was previously
although undifferentiated THP-1 cells can be infected with CXCR4- shown to trigger reduced cell growth in vitro and in vivo.30 We
using virus strains, phorbol 12-myristate 13-acetate-differentiated transduced SupT1 cells with the respective LV and the empty JS1
cells are resistant to these viruses.34–38 We therefore used SupT1 vector as negative control. To determine any negative effects on
T cells as Dicer-positive control cells. growth of the transduced cells, we monitored the percentage
THP-1 cells were transduced with the respective LV and GFP- of GFP-positive (and RNA-expressing) cells in the transduced
positive cells were selected for subsequent virus challenge with culture for 50 days (Figure 7). Untransduced cells provide

Gene Therapy (2017) 453 – 461 © 2017 Macmillan Publishers Limited, part of Springer Nature.
Antiviral AgoshRNA design
E Herrera-Carrillo et al
459
boost the silencing activity and to develop better design
Table 1. Competitive cell growth (CCG) assay
algorithms.
Lentiviral vector construct Reduction in GFP+ cells (%)a Recent evidence indicates that one can design shRNAs with a
small loop and short duplex length that avoid Dicer processing
moi:0.15 moi:1.5
and instead are processed by Ago2.12,15,17,18,47 This new
AgoshRNA design has a major hypothetical advantage that only
a single active guide RNA is generated, without a passenger strand
Mean s.d. Mean s.d. that can induce unwanted side effects. We previously listed
JS1 2.3 0.5 0.4 1.6 additional advantages of the AgoshRNA design.21 In this study, we
AgoshGag4 1.7 2.7 1.7 1.6 confirmed the safety advantage by converting a ‘toxic’ shRNA into
AgoshGag5 2.2 1.3 0.0 3.1 a ‘non-toxic’ AgoshRNA. An additional safety aspect relates to the
AgoshGag6 − 0.3 1.6 0.8 3.2 shorter duplex of AgoshRNAs versus shRNAs, which may activate
AgoshPol1 − 0.1 1.0 0.7 1.5 innate immunity sensors such as interferon less efficiently.48
AgoshPol8 2.1 2.1 0.7 1.6 Furthermore, AgoshRNAs like miR-451 are loaded exclusively in
AgoshPol45 − 0.8 1.0 − 0.8 1.0
AgoshRT5 0.3 1.5 − 1.4 1.1
Ago2-containing RNA-induced silencing complex complexes49
shGag5 14.1 1.1 22.2 1.7 (Harwig et al., unpublished results). Avoiding AgoshRNA loading
shRT5 1.4 1.7 0.4 2.0 in Ago1, 3 and 4 will restrict unwanted off-targeting,50 although
the contribution of the individual Ago proteins to siRNA off-target
Abbreviations: GFP, green fluorescent protein; moi, multiplicities of
activity is still unclear.50–53 The AgoshRNA design is ideally suited
infection. aGFP expression was measured up to 50 days.
for specific therapeutic strategies. A restriction for massive
AgoshRNA (and shRNA) use is that siRNA design algorithms
the internal control. The transduction was performed at two cannot be applied, which calls for a difficult trial-and-error design
different multiplicities of infection, 0.15 and 1.5, which obviously process.39–42
yielded a different starting level of GFP-positive cells. We
confirmed the selective loss of GFP-positive cells for shGag5, but
MATERIALS AND METHODS
no cell growth impairment was apparent in the AgoshRNA
cultures (included AgoshGag5) (Figure 7). Similar results were Plasmid construction
obtained in five independent experiments. A significant decrease For the AgoshRNA and shRNA constructs, complementary DNA oligonu-
of GFP-positive cells was observed exclusively for shGag5 cleotides encoding the corresponding sequences were annealed to create
(P o 0.0001) compared with the other shRNAs, all AgoshRNAs sticky BamHI and HindIII sites, and subsequently inserted into the
corresponding restriction sites of the pSUPER vector.54 Firefly luciferase
and the JS1 control (Table 1).
reporter constructs (pGL3; Promega, Madison, WI, USA) were made by
insertion of a 50–70 nt HIV-1 sequence, with the 19 nt target region in the
center, in the EcoRI and PstI sites of the pGL3 plasmid.31 The precise
DISCUSSION
positions of the inserted HIV-1 fragments are as follows: Gag (1341–1399),
We tested 21 novel anti-HIV AgoshRNA candidates by incorporat- Pol1 (1889–1948), Pol8 (4017–4155), Pol45 (4531–4589) and R/T5 (5535–
ing previously proven shRNA-derived guide sequences into the 5583). The luciferase reporters with the sense and antisense target
AgoshRNA design.27,28 Seven of these 21 AgoshRNAs were highly sequences were described previously.15 All constructs were verified by
active in reporter silencing. However, only AgoshPol8 and sequence analysis.
AgoshGag4 exhibited profound HIV-1 inhibition in spreading
infections on a T-cell line. Resistant HIV-1 variants were selected in Cell culture
the AgoshGag4 cultures, but breakthrough replication of the WT HEK293T adherent cells (ATCC CRL-11268, Manassas, VA, USA) were grown
virus was apparent for the other AgoshRNAs. The lack of selection as monolayer in Dulbecco’s modified Eagle’s medium (Life Technologies,
of AgoshRNA-resistant virus variants indicates that these antivirals Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal calf serum,
do not exert strong selective pressure on the HIV-1 RNA minimal essential medium non-essential amino acids, streptomycin
genome.27,32 Although this initial result of a single potent (100 μg ml − 1) and penicillin (100 U ml − 1) in a humidified chamber
AgoshRNA is not encouraging, we stress that only four potent at 37 °C and 5% CO2. SupT1 T cells (ATCC CRL-1942) were grown in
shRNAs were previously selected by us in a large screen of nearly Advanced RPMI (Gibco BRL, Carlsbad, CA, USA) supplemented with
100 shRNA candidates.27 RNAi tools are popular, but it is difficult L-glutamine, 1% fetal calf serum, streptomycin (30 μg ml − 1) and penicillin
to design active shRNAs and AgoshRNAs, as these molecules do (30 U ml − 1) in a humidified chamber at 37 °C and 5% CO2. THP-1 cells
not adopt the known siRNA design rules.39–42 Furthermore, HIV-1 (ATCC TIB-202) were grown in RPMI (Gibco BRL) supplemented with 10%
represents a difficult target mRNA because it adopts stable RNA fetal calf serum, streptomycin (1000 μg ml − 1) and penicillin (100 U ml − 1)
in a humidified chamber at 37 °C and 5% CO2. Regular testing for
structures that inhibit an RNAi attack.43–46 On the positive side, the
mycoplasma contamination was performed.
AgoshRNAs exclusively induced guide-mediated luciferase knock-
down, whereas the shRNA triggered guide and passenger strand
activity. We checked the absence of a passenger strand for the Transient HIV-1 inhibition assays
AgoshRNA candidates by northern blotting, which indeed seems a HEK293T cells were seeded 1 day before transfection in 24-well plates at a
major bonus over regular shRNAs. Furthermore, the AgoshRNA density of 1.2 × 105 cells per well. Co-transfection of 250 ng of the full-
candidates revealed no toxicity in the ultra-sensitive competitive length HIV-1 molecular clone pLAI,29 1 ng of pRL-CMV and 25 ng of
cell growth assay. Most strikingly, AgoshGag5 revealed no toxicity, AgoshRNA construct was performed using Lipofectamine 2000 reagent
(Invitrogen). We added pBS to have an equal DNA concentration per
whereas the corresponding shGag5 inhibitor showed a profound
transfection. Two days post transfection, samples were taken for virus
impact on T-cell growth. We demonstrated another advantage of production, which was determined by CA-p24 enzyme-linked immunosor-
AgoshRNAs: their ability to maintain antiviral activity in a Dicer- bent assay and cells were lysed for Renilla luciferase activity measurement
deficient monocyte cell line that lack Dicer expression. Thus, the with the Renilla Luciferase Assay System (Promega). Relative HIV-1
AgoshRNA design may constitute a new platform for gene production was calculated as the ratio between the CA-p24 level and
silencing that seems competitive with current miRNA and shRNA the Renilla luciferase activity. We performed three independent transfec-
technology. These data indicate that the future for AgoshRNA tions, each in duplicate. Values were corrected for between-session
therapeutics is promising, but further refinements are essential to variation as described previously.55

© 2017 Macmillan Publishers Limited, part of Springer Nature. Gene Therapy (2017) 453 – 461
Antiviral AgoshRNA design
E Herrera-Carrillo et al
460
Luciferase assays the membrane using UV light (1200 μJ × 100). The membranes were
Luciferase tests were performed as previously described.56 In brief, hybridized overnight with locked nucleic acid oligonucleotides. Locked
HEK293T cells were seeded one day before transfection in 24-well plates nucleic acid oligonucleotide probes were 5′-end labeled with the
at a density of 1.2 × 105 cells per well. Cells were co-transfected with the kinaseMax kit (Life Technologies, Invitrogen) in the presence of 1 μl
indicated DNA construct (1, 5 or 25 ng), 100 ng of the firefly luciferase [γ − 32 P] ATP (0.37 MBq μl − 1, Perkin Elmer, Waltham, MA, USA). The
expression plasmid and 1 ng of Renilla luciferase expression plasmid (pRL) probes were purified on Sephadex G-25 columns (Amersham Biosciences
using Lipofectamine 2000 reagent (Invitrogen). We added pBS plasmid to Inc., Sunnyvale, CA, USA) to remove unincorporated nt. We used the
create an equal DNA concentration for each transfection. Cells were lysed following oligonucleotides probes (locked nucleic acid positions under-
48 h post transfection, to measure firefly and Renilla luciferase activities lined) to detect the 5′ and 3′ strand of the siRNA, respectively: guide:
using the Dual-Luciferase Reporter Assay System (Promega). The ratio 5′-GAAGAAATGATGACAGCAT-3′ and passenger: 5′-ATGCTGTCATCATTTC
between firefly and Renilla luciferase activity was used for normalization of TTC-3′. The signals were captured by the Typhoon FLA 9500 (GE Healthcare
experimental variations such as differences in transfection efficiency. The Bio-Sciences AB, Uppsala, Zweden) and quantifying using the Molecular
pBS plasmid and an unrelated shRNA (shNef) served as negative controls. Dynamics ImageQuant (5.2) software package (GE Healthcare, Waukesha,
The luciferase activity scored with shNef activity was set at 100%. We WI, USA).
performed three independent transfections, each in duplicate. Values were
corrected for between-session variation. The resulting six values were used
Competitive cell growth assay
to calculate the s.d. shown as error bar.
SupT1 T cells were transduced at a multiplicities of infection of 0.15 and
1.5, and transduced cells were screened for a negative impact of lentiviral
Lentiviral vector production and transduction integration and/or AgoshRNA expression on cell growth using the
The LV was produced and titrated as described previously.27 LV plasmids competitive cell growth assay.61 In brief, the mixture of transduced (GFP
encoding the AgoshRNAs are derived from the construct JS1 +/AgoshRNA+) and untransduced (GFP − ) cells was analyzed for the GFP
(pRRLcpptpgkgfppreSsin).57 The vector was produced by co-transfection +/ − ratio up to 50 days by fluorescence-activated cell sorting. The impact
of packaging plasmids pSYNGP,58 pRSV-rev and pVSV-g59 and LV plasmid on cell growth was converted as percentage reduction in cell growth.
with Lipofectamine 2000 (Invitrogen). After transfection, the medium was
replaced with OptiMEM (Invitrogen). The LV containing supernatant was
collected, filtered (0.45 μm) and aliquots were stored at − 80 °C. The
Statistical analysis
transduction titer was measured via GFP expression. SupT1 and THP-1 cells Results are presented as mean ± s.d. P-values o0.05 were considered
were transduced at a multiplicities of infection of 0.15. Three days after statistically significant. Comparisons between three or more groups were
transduction, live cells were selected by fluorescence-activated cell sorting analysed by one-way analysis of variance followed by a Tukey’s post-hoc
for GFP expression. test. Statistical analysis was performed using GraphPad Prism 7.02
Transduced SupT1 T cells (1 × 106 cells in 5 ml medium) were challenged (GraphPad Prism, La Jolla, CA, USA).
with HIV-1 LAI (0.01 ng CA-p24). Transduced THP1 cells (0.2 × 106 cells in
96-well plate in 0.2 ml medium) were challenged with HIV-1 LAI (5 ng CA-
p24). Transduced THP-1 infected cells were centrifuged at 1000 g for 2 h at CONFLICT OF INTEREST
18 °C. Each cell pellet was resolved in 2 ml of media and transferred to its The authors declare no conflict of interest.
own well in a six-well plate tissue culture plate. Virus spread was monitored
by measuring CA-p24 production and we scored HIV-induced syncytia
formation twice a week. ACKNOWLEDGEMENTS
This work was supported by the Nederlandse Organisatie voor Wetenschappelijk
Sequencing proviral target regions Onderzoek—Chemische Wetenschappen (NWO-CW, Top Grant) and Zorg Onderzoek
When virus replication was observed after infection with HIV-1 LAI, cellular Nederland—Medische Wetenschappen (ZonMw, Translational Gene Therapy Grant).
DNA of the infected cells with the integrated provirus was isolated as We thank Berend Hooibrink for expertise in cell sorting and maintenance of the flow
previously described.60 Integrated proviral DNA sequences were PCR cytometry facility. We thank Cosimo Cristella for valuable help with the statistical
amplified with the following primer pairs (5′–3′, the position within pLAI is analyses.
indicated): Gag sense (5′-CAGACCATCAATGAGGAAGCTGCAGAATGGGAT;
position 1445-3′) and antisense (5′-CCCTGGCCTTCCCTTGTAGGAAAA
CCAGATCTTCCC-3′; position 2141); Protease, sense (5′-GTCAGAGCAGACCA REFERENCES
GAGCCAACAG-3′; position 2183) and antisense (5′-GATATTTCTCATGTTCAT 1 Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009; 136:
CTTGGGCCTTATCTATTCC-3′; position 2659); Integrase, sense (5′-GGCAA 215–233.
CTAGATTGTACACATTTAGAAGG-3′; position 4499) and antisense (5′-CTCT 2 Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short
TTTTCCTCCATTCTATGGAGA-3′; position 5377) and Tat-Rev sense (5′-ATATC interfering RNAs in mammalian cells. Science 2002; 296: 550–553.
AAGCAGGACATAACAAGG-3′; position 5525) and antisense (5′-TGCTTTAG 3 Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell
CATCTGATGCACAAAATA-3′; position 6458) with 30 cycles (1 min denatura- 2009; 136: 642–655.
tion at 96 °C, 1 min annealing at 62 °C and 2 min extension at 72 °C). The 4 Berkhout B. Toward a durable anti-HIV gene therapy based on RNA interference.
PCR products were sequenced with the Big Dye Terminator Cycle Ann N Y Acad Sci 2009; 1175: 3–14.
Sequencing kit (ABI) using the same primers. 5 Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K
et al. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene
silencing. Nature 2005; 436: 740–744.
Northern blot analyses 6 Jaskiewicz L, Filipowicz W. Role of Dicer in posttranscriptional RNA silencing. Curr
Northern blot experiments were performed as previously described.15,56 Top Microbiol Immunol 2008; 320: 77–97.
For siRNA analyses, 1.5 × 106 HEK293T cells were seeded in T25 flasks. The 7 Siomi H, Siomi MC. Posttranscriptional regulation of microRNA biogenesis in
cells were transfected with 5 μg AgoshRNA and shRNA constructs using animals. Mol Cell 2010; 38: 323–332.
Lipofectamine 2000 reagent. Total cellular RNA was extracted two days 8 Yang JS, Maurin T, Robine N, Rasmussen KD, Jeffrey KL, Chandwani R et al.
post transfection using the mirVana miRNA isolation kit (Life Technologies, Conserved vertebrate mir-451 provides a platform for Dicer-independent, Ago2-
Invitrogen). RNA concentrations were determined on the Nanodrop 1000 mediated microRNA biogenesis. Proc Natl Acad Sci USA 2010; 107: 15163–15168.
machine (Thermo Fisher Scientific, Wilmington, DE, USA). Five micrograms 9 Cifuentes D, Xue H, Taylor DW, Patnode H, Mishima Y, Cheloufi S et al. A novel
of total RNA was electrophoresed in a 15% denaturing polyacrylamide gel miRNA processing pathway independent of Dicer requires Argonaute2 catalytic
(Precast Novex TBU gel, Life Technologies, Invitrogen). [γ − 32P]-labeled activity. Science 2010; 328: 1694–1698.
decade RNA marker (Life Technologies, Invitrogen) was run along-side for 10 Cheloufi S, Dos Santos CO, Chong MM, Hannon GJ. A dicer-independent miRNA
size estimation. To check for equal sample loading, the RNA in the gel was biogenesis pathway that requires Ago catalysis. Nature 2010; 465: 584–589.
stained with 2 μg ml − 1 ethidium bromide and visualized under UV light. 11 Yoda M, Cifuentes D, Izumi N, Sakaguchi Y, Suzuki T, Giraldez AJ et al. Poly(A)-
The RNA was electro-transferred to a positively charged nylon membrane specific ribonuclease mediates 3'-end trimming of Argonaute2-cleaved precursor
(Boehringer Mannheim, GmbH, Mannheim, Germany) and cross-linked to microRNAs. Cell Rep 2013; 5: 715–726.

Gene Therapy (2017) 453 – 461 © 2017 Macmillan Publishers Limited, part of Springer Nature.
Antiviral AgoshRNA design
E Herrera-Carrillo et al
461
12 Herrera-Carrillo E, Harwig A, Liu YP, Berkhout B. Probing the shRNA characteristics 37 Ushijima H, Kunisada T, Ami Y, Tsuchie H, Takahashi I, Schacke H et al. Char-
that hinder Dicer recognition and consequently allow Ago-mediated processing acterization of cells of the myeloid-monocytic lineage (ML-1, HL-60, THP-1, U-937)
and AgoshRNA activity. RNA 2014; 20: 1410–1418. chronically infected with the human immunodeficiency virus-1. Pathobiology
13 Herrera-Carrillo E, Harwig A, Berkhout B. Toward optimization of AgoshRNA 1993; 61: 145–153.
molecules that use a non-canonical RNAi pathway: variations in the top and 38 Su J, Palm A, Wu Y, Sandin S, Hoglund S, Vahlne A. Deletion of the GPG motif in
bottom base pairs. RNA Biol 2015; 12: 447–456. the HIV type 1 V3 loop does not abrogate infection in all cells. AIDS Res Hum
14 Harwig A, Herrera-Carrillo E, Jongejan A, van Kampen AH, Berkhout B. Deep Retroviruses 2000; 16: 37–48.
sequence analysis of AgoshRNA processing reveals 3' A addition and trimming. 39 Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs exhibit
Mol Ther Nucleic Acids 2015; 4: e247. strand bias. Cell 2003; 115: 209–216.
15 Liu YP, Schopman NC, Berkhout B. Dicer-independent processing of short 40 Schwarz DS, Hutvagner G, Du T, Xu Z, Aronin N, Zamore PD. Asymmetry in the
hairpin RNAs. Nucleic Acids Res 2013; 41: 3723–3733. assembly of the RNAi enzyme complex. Cell 2003; 115: 199–208.
16 Dallas A, Ilves H, Ge Q, Kumar P, Shorenstein J, Kazakov SA et al. Right- and left- 41 Reynolds A, Leake D, Boese Q, Scaringe S, Marshall WS, Khvorova A. Rational
loop short shRNAs have distinct and unusual mechanisms of gene silencing. siRNA design for RNA interference. Nat Biotechnol 2004; 22: 326–330.
Nucleic Acids Res 2012; 40: 9255–9271. 42 Tomari Y, Zamore PD. Perspective: machines for RNAi. Genes Dev 2005; 19:
17 Sun G, Yeh SY, Yuan CW, Chiu MJ, Yung BS, Yen Y. Molecular properties, functional 517–529.
mechanisms, and applications of sliced siRNA. Mol Ther Nucleic Acids 2015; 4: 43 Westerhout EM, Ooms M, Vink M, Das AT, Berkhout B. HIV-1 can escape from RNA
e221. interference by evolving an alternative structure in its RNA genome. Nucleic Acids
18 Ma H, Zhang J, Wu H. Designing Ago2-specific siRNA/shRNA to avoid competition Res 2005; 33: 796–804.
with endogenous miRNAs. Mol Ther Nucleic Acids 2014; 3: e176. 44 Westerhout EM, ter Brake O, Berkhout B. The virion-associated incoming HIV-1
19 Herrera-Carrillo E, Zong-liang G, Harwig A, Heemskerk MT, Berkhout B. The RNA genome is not targeted by RNA interference. Retrovirology 2006; 3: 57.
influence of the 5’-terminal nucleotide on AgoshRNA activity and biogenesis: 45 Westerhout EM, Berkhout B. A systematic analysis of the effect of target RNA
importance of the polymerase III transcription initiation site. Nucleic Acids Res structure on RNA interference. Nucleic Acids Res 2007; 35: 4322–4330.
2017; 45: 4036–4050. 46 Low JT, Knoepfel SA, Watts JM, ter Brake O, Berkhout B, Weeks KM. SHAPE-
20 Ma H, Wu Y, Dang Y, Choi JG, Zhang J, Wu H. Pol III promoters to express small directed discovery of potent shRNA inhibitors of HIV-1. Mol Ther 2012; 20:
RNAs: delineation of transcription initiation. Mol Ther Nucleic Acids 2014; 3: e161. 820–828.
21 Berkhout B, Liu YP. Towards improved shRNA and miRNA reagents as inhibitors of 47 Ge Q, Ilves H, Dallas A, Kumar P, Shorenstein J, Kazakov SA et al. Minimal-length
HIV-1 replication. Future Microbiol 2014; 9: 561–571. short hairpin RNAs: the relationship of structure and RNAi activity. RNA 2010; 16:
22 Coley W, Van Duyne R, Carpio L, Guendel I, Kehn-Hall K, Chevalier S et al. Absence 106–117.
of DICER in monocytes and its regulation by HIV-1. J Biol Chem 2010; 285: 48 Bridge AJ, Pebernard S, Ducraux A, Nicoulaz AL, Iggo R. Induction of an interferon
31930–31943. response by RNAi vectors in mammalian cells. Nat Genet 2003; 34: 263–264.
23 Frank F, Sonenberg N, Nagar B. Structural basis for 5'-nucleotide base-specific 49 Dueck A, Ziegler C, Eichner A, Berezikov E, Meister G. microRNAs associated with
recognition of guide RNA by human AGO2. Nature 2010; 465: 818–822. the different human Argonaute proteins. Nucleic Acids Res 2012; 40: 9850–9862.
24 Hu HY, Yan Z, Xu Y, Hu H, Menzel C, Zhou YH et al. Sequence features associated 50 Petri S, Dueck A, Lehmann G, Putz N, Rudel S, Kremmer E et al. Increased siRNA
with microRNA strand selection in humans and flies. BMC Genomics 2009; 10: 413. duplex stability correlates with reduced off-target and elevated on-target effects.
25 Liu YP, Westerink JT, Ter Brake O, Berkhout B. RNAi-inducing lentiviral vectors for RNA 2011; 17: 737–749.
anti-HIV-1 gene therapy. Methods Mol Biol 2011; 721: 293–311. 51 Aleman LM, Doench J, Sharp PA. Comparison of siRNA-induced off-target RNA
26 Liu YP, Vink MA, Westerink JT, Ramirez de Arellano E, Konstantinova P, Ter Brake O and protein effects. RNA 2007; 13: 385–395.
et al. Titers of lentiviral vectors encoding shRNAs and miRNAs are reduced by 52 Wu L, Fan J, Belasco JG. Importance of translation and nonnucleolytic ago pro-
different mechanisms that require distinct repair strategies. RNA 2010; 16: teins for on-target RNA interference. Curr Biol 2008; 18: 1327–1332.
1328–1339. 53 Vickers TA, Lima WF, Wu H, Nichols JG, Linsley PS, Crooke ST. Off-target and a
27 Ter Brake O, Konstantinova P, Ceylan M, Berkhout B. Silencing of HIV-1 with RNA portion of target-specific siRNA mediated mRNA degradation is Ago2 'Slicer'
interference: a multiple shRNA approach. Mol Ther 2006; 14: 883–892. independent and can be mediated by Ago1. Nucleic Acids Res 2009; 37:
28 Das AT, Brummelkamp TR, Westerhout EM, Vink M, Madiredjo M, Bernards R et al. 6927–6941.
Human immunodeficiency virus type 1 escapes from RNA interference-mediated 54 Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific
inhibition. J Virol 2004; 78: 2601–2605. genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature
29 Peden K, Emerman M, Montagnier L. Changes in growth properties on passage in 1998; 391: 806–811.
tissue culture of viruses derived from infectious molecular clones of HIV-1LAI, 55 Ruijter JM, Thygesen HH, Schoneveld OJ, Das AT, Berkhout B, Lamers WH.
HIV-1MAL, and HIV-1ELI. Virology 1991; 185: 661–672. Factor correction as a tool to eliminate between-session variation in replicate
30 Centlivre M, Legrand N, Klamer S, Liu YP, Eije KJ, Bohne M et al. Preclinical in vivo experiments: application to molecular biology and retrovirology. Retrovirology
evaluation of the safety of a multi-shRNA-based gene therapy against HIV-1. Mol 2006; 3: 2.
Ther Nucleic Acids 2013; 2: e120. 56 Herrera-Carrillo E, Gao ZL, Harwig A, Heemskerk MT, Berkhout B. The influence of
31 Westerhout EM, Ooms M, Vink M, Das AT, Berkhout B. HIV-1 can escape from RNA the 5-terminal nucleotide on AgoshRNA activity and biogenesis: importance of
interference by evolving an alternative structure in its RNA genome. Nucleic Acids the polymerase III transcription initiation site. Nucleic Acids Res 2017; 45:
Res 2005; 33: 796–804. 4036–4050.
32 von Eije KJ, Ter Brake O, Berkhout B. Human immunodeficiency virus type 1 57 Seppen J, Rijnberg M, Cooreman MP, Oude Elferink RP. Lentiviral vectors for
escape is restricted when conserved genome sequences are targeted by RNA efficient transduction of isolated primary quiescent hepatocytes. J Hepatol 2002;
interference. J Virol 2008; 82: 2895–2903. 36: 459–465.
33 Auwerx J. The human leukemia cell line, THP-1: a multifacetted model for the 58 Kotsopoulou E, Kim VN, Kingsman AJ, Kingsman SM, Mitrophanous KA. A Rev-
study of monocyte-macrophage differentiation. Experientia 1991; 47: 22–31. independent human immunodeficiency virus type 1 (HIV-1)-based vector that
34 Tsuchiya S, Yamabe M, Yamaguchi Y, Kobayashi Y, Konno T, Tada K. Establishment exploits a codon-optimized HIV-1 gag-pol gene. J Virol 2000; 74: 4839–4852.
and characterization of a human acute monocytic leukemia cell line (THP-1). Int J 59 Zufferey R, Dull T, Mandel RJ, Bukovsky A, Quiroz D, Naldini L et al. Self-
Cancer 1980; 26: 171–176. inactivating lentivirus vector for safe and efficient in vivo gene delivery. J Virol
35 Kitano K, Baldwin GC, Raines MA, Golde DW. Differentiating agents facilitate 1998; 72: 9873–9880.
infection of myeloid leukemia cell lines by monocytotropic HIV-1 strains. Blood 60 Konstantinova P, de Haan P, Das AT, Berkhout B. Hairpin-induced tRNA-mediated
1990; 76: 1980–1988. (HITME) recombination in HIV-1. Nucleic acids Res 2006; 34: 2206–2218.
36 Ushijima H, Dairaku M, Honma H, Yamaguchi K, Shimizu H, Tsuchie H et al. Human 61 Eekels JJ, Pasternak AO, Schut AM, Geerts D, Jeeninga RE, Berkhout B. A com-
immunodeficiency virus infection in cells of myeloid-monocytic lineage. Microbiol petitive cell growth assay for the detection of subtle effects of gene transduction
Immunol 1991; 35: 487–492. on cell proliferation. Gene Ther 2012; 19: 1058–1064.

Supplementary Information accompanies this paper on Gene Therapy website (http://www.nature.com/gt)

© 2017 Macmillan Publishers Limited, part of Springer Nature. Gene Therapy (2017) 453 – 461

S-ar putea să vă placă și