throbber
Downloaded from
`
`
`
`rnajournal.cshlp.org Cold Spring Harbor Laboratory Press on December 26, 2017 - Published by
`
`
`
`
`
`siRNA function in RNAi: A chemical modification analysis
`
`YA-LIN CHIU and TARIQ M. RANA
`Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School,
`Worcester, Massachusetts 01605, USA
`
`ABSTRACT
`Various chemical modifications were created in short-interfering RNAs (siRNAs) to determine the biochemical properties
`required for RNA interference (RNAi). Remarkably, modifications at the 2ⴕ-position of pentose sugars in siRNAs showed the
`2ⴕ-OHs were not required for RNAi, indicating that RNAi machinery does not require the 2ⴕ-OH for recognition of siRNAs and
`catalytic ribonuclease activity of RNA-induced silencing complexes (RISCs) does not involve the 2ⴕ-OH of guide antisense RNA.
`In addition, 2ⴕ modifications predicted to stabilize siRNA increased the persistence of RNAi as compared with wild-type siRNAs.
`RNAi was also induced with chemical modifications that stabilized interactions between A–U base pairs, demonstrating that
`these types of modifications may enhance mRNA targeting efficiency in allele-specific RNAi. Modifications altering the structure
`of the A-form major groove of antisense siRNA–mRNA duplexes abolished RNAi, suggesting that the major groove of these
`duplexes was required for recognition by activated RISC*. Comparative analysis of the stability and RNAi activities of chemically
`modified single-stranded antisense RNA and duplex siRNA suggested that some catalytic mechanism(s) other than siRNA
`stability were linked to RNAi efficiency. Modified or mismatched ribonucleotides incorporated at internal positions in the 5ⴕ or
`3ⴕ half of the siRNA duplex, as defined by the antisense strand, indicated that the integrity of the 5ⴕ and not the 3ⴕ half of the
`siRNA structure was important for RNAi, highlighting the asymmetric nature of siRNA recognition for initiation of unwinding.
`Collectively, this study defines the mechanisms of RNAi in human cells and provides new rules for designing effective and stable
`siRNAs for RNAi-mediated gene-silencing applications.
`Keywords: RNAi; siRNA; human; nucleotide modification; GFP
`
`INTRODUCTION
`
`The evolutionarily conserved phenomenon RNA interfer-
`ence (RNAi), the process by which specific mRNAs are
`targeted for degradation by complementary short-interfer-
`ing RNAs (siRNAs), has increasingly become a powerful
`tool for genetic analysis and is likely to become a potent
`therapeutic approach for gene silencing (for review, see
`Hammond et al. 2001; McManus and Sharp 2002). Conse-
`quently, understanding the mechanism of RNAi has be-
`come critical for developing the most effective RNAi meth-
`odologies for both laboratory and clinical applications. The
`general mechanism of RNAi
`involves the cleavage of
`double-stranded RNA (dsRNA) to short 21–23-nt siRNAs.
`This processing event is catalyzed by Dicer, a highly con-
`served, dsRNA-specific endonuclease that is a member of
`the RNase III family (Hammond et al. 2000; Zamore et al.
`
`Reprint requests to: Tariq M. Rana, Chemical Biology Program, De-
`partment of Biochemistry and Molecular Pharmacology, University of
`Massachusetts Medical School, 364 Plantation Street, Worcester, MA
`01605, USA; e-mail: tariq.rana@umassmed.edu; fax: (508) 856-6696.
`Article and publication are at http://www.rnajournal.org/cgi/doi/
`10.1261/rna.5103703.
`
`2000; Bernstein et al. 2001; Hamilton et al. 2002; Provost et
`al. 2002; Zhang et al. 2002). Processing by Dicer results in
`siRNA duplexes that have 5⬘-phosphate and 3⬘-hydroxyl
`termini, and subsequently, these siRNAs are recognized by
`the RNA-induced silencing complex (RISC; Hammond et
`al. 2000). Active RISC complexes (RISC*) promote the un-
`winding of the siRNA through an ATP-dependent process,
`and the unwound antisense strand guides RISC* to the
`complementary mRNA (Nykanen et al. 2001). The targeted
`mRNA is then cleaved by RISC* at a single site that is
`defined with regard to where the 5⬘-end of the antisense
`strand is bound to the mRNA target sequence (Hammond
`et al. 2000; Elbashir et al. 2001b). For RNAi-mediated
`mRNA cleavage and degradation to be successful, 5⬘-phos-
`phorylation of the antisense strand must occur, and the
`double helix of the antisense-target mRNA duplex must be
`in the A form (Chiu and Rana 2002).
`One highlighted difference between mammalian RNAi
`and RNAi in other eukaryotes is the lack of an amplification
`system for long-term persistence of RNAi in mammalian
`cells. For example, in Drosophila, ∼35 molecules of dsRNA
`can silence ∼1000 copies of the targeted mRNA per cell and
`can persist over the course of many generations (Kennerdell
`
`1034
`
`RNA (2003), 9:1034–1048. Published by Cold Spring Harbor Laboratory Press. Copyright © 2003 RNA Society.
`
`Alnylam Exh. 1044
`
`

`

`
`
`Downloaded from rnajournal.cshlp.org on December 26, 2017 - Published by Cold Spring Harbor Laboratory Press
`
`
`
`
`
`
`
`and Carthew 1998; Zamore 2001). In mammalian cells,
`RNAi only persists effectively for an average of ∼66 h before
`the siRNA is likely diluted out over the course of several cell
`divisions (Chiu and Rana 2002). The amplification that is
`seen in flies and other lower eukaryotes can potentially be
`attributed to three factors. One is that the conversion of
`long trigger dsRNA to smaller 21–23-nt siRNAs by Dicer
`adds a degree of RNAi amplification, whereas in mamma-
`lian cells long trigger dsRNA invokes the interferon re-
`sponse that activates the protein kinase PKR (Stark et al.
`1998). This suggests that only siRNA transfections success-
`fully trigger RNAi in mammalian cells without other side
`effects, and thus, no amplification would take place through
`the processing of longer RNAs. A second factor in amplifi-
`cation is the presence of RNA-dependent RNA polymerase
`(RdRP), which has been found in plants, worms, fungi, and
`flies (Cogoni and Macino 1999; Dalmay et al. 2000; Sijen et
`al. 2001). RdRP has been postulated to amplify target
`mRNA,
`through a random, degradative PCR model
`(Lipardi et al. 2001; Nishikura 2001; Sijen et al. 2001), into
`dsRNA, which can be targeted by Dicer. However, no RdRP
`homologs have been found in mammalian cells, and the
`3⬘-OH that is required for RdRP-dependent degradative
`PCR is not required for RNAi in mammalian cells (Chiu
`and Rana 2002; Schwarz et al. 2002; Stein et al. 2003),
`indicating that PCR-based amplification likely does not oc-
`cur in mammals. A third factor in amplification may be the
`high enzymatic turnover rate of RISC* during the targeting
`and cleavage of mRNA (Hutvagner and Zamore 2002),
`which may add a degree of amplification to RNAi induction
`in all eukaryotes, including mammals. However, as the per-
`sistence of RNAi occurs for only a short period of time,
`finding methods for increasing the longevity of siRNAs in
`human cells will be fundamental for applying RNAi to labo-
`ratory and therapeutic applications.
`To address this issue of siRNA stability for prolonging the
`duration of dsRNA-mediated gene silencing and to further
`dissect the mechanism of RNAi in human cells, various
`chemically modified nucleotides predicted to affect siRNA
`stability were incorporated into siRNAs to study whether
`specific modifications increased or decreased the efficacy
`and persistence of RNAi in vivo. The most important of
`these modifications was to the 2⬘-OH of the ribonucleotide
`that distinguishes RNA from DNA and is required for the
`nucleophilic attack occurring during the hydrolysis of the
`RNA backbone,
`the reaction catalyzed by degradative
`RNases. Our results showed that the 2⬘-OH was not re-
`quired for RNAi,
`indicating that structural rather than
`chemical properties of siRNA–mRNA duplexes were the key
`to inducing RNAi and that RISC* did not require the 2⬘-
`OH for ribonuclease activity. 2⬘-modified siRNAs also in-
`creased the persistence of RNAi in human cells. Modifica-
`tions that stabilized base-pairing interactions were also in-
`corporated into the antisense strand of siRNAs and were
`able to initiate RNAi, signifying that this class of chemical
`
`Chemical modification of siRNAs
`
`modifications could be used to increase the targeting effi-
`ciency of siRNAs for mRNA target sequences and for allele-
`specific inhibition of gene expression.
`Other chemical modifications affected the formation of
`the major groove of the A-form helix of the antisense-
`siRNA–target-mRNA duplex, and potentially disrupted H-
`bonds or sterically hindered protein contacts, most prob-
`ably preventing the RISC* complex from stably interacting
`with the dsRNA duplex. These modifications completely
`abolished RNAi, demonstrating that an intact major groove
`in the A-form helix and stable RNA–protein interactions
`were required for RNAi in human cells. Finally, previous
`observations of psorelan cross-linked siRNAs implied that
`unwinding of siRNA occurred from the 5⬘-end of the an-
`tisense strand and that complete unwinding may not be
`necessary for effective RNAi (Chiu and Rana 2002). By us-
`ing mismatched or chemically modified nucleotides on ei-
`ther the 3⬘ or 5⬘ half of the antisense strand within the
`siRNA duplex, we have shown here that RNAi depended on
`the integrity of the 5⬘, and not the 3⬘, half of the siRNA
`duplex, as defined by the antisense strand. Altogether, these
`results gave insight into the essential biochemical properties
`of functional siRNAs and how specific changes in the siRNA
`structure can affect the efficacy of RNAi. Furthermore, these
`studies present new methodologies for improving the sta-
`bility and utility of siRNAs for future RNAi applications.
`
`RESULTS
`
`2ⴕ-OH is not required for siRNA to enter the
`RNAi pathway
`
`Previous results showed that RNAi effects typically peaked
`between 42 and 54 h posttransfection, and targeted gene
`expression started to be restored by 66 h posttransfection
`(Chiu and Rana 2002). To determine if the duration of
`RNAi could be prolonged by increasing the half-life of
`siRNAs, various chemical modifications were made to
`nucleotides that affected siRNA stability. These modified
`siRNAs were then tested in an improved dual fluorescence
`reporter assay based on the original assay developed previ-
`ously (Chiu and Rana 2002). Briefly, GFP and RFP were
`constitutively expressed from pEGFP-C1 and pDsRed2-N1,
`respectively. EGFP mRNA was targeted for RNAi using a
`21-nt siRNA targeted to nucleotides 238–258 of the EGFP
`mRNA (Fig. 1A). The fluorescence intensity ratio of target
`(GFP) to control (RFP) fluorophores was determined in the
`presence of siRNA duplexes and normalized to that ob-
`served in the mock-treated cells. The sequence of EGFP
`siRNA and EGFP mRNA, the specific mRNA cleavage site,
`plus the structures of the chemically modified nucleotides
`are diagrammed in Figure 1. As outlined previously, the
`cleavage site was defined precisely 11 nt downstream of the
`target position complementary to the 3⬘-most nucleotide of
`
`www.rnajournal.org
`
`1035
`
`

`

`Downloaded from
`
`Chiu and Rana
`
`
`
`rnajournal.cshlp.org Cold Spring Harbor Laboratory Press on December 26, 2017 - Published by
`
`
`
`
`
`Sense strand (SSI
`
`siRNA
`
`21
`5· GCAGCACGACUUCUUCAAGdTdT
`
`Antisense strand (ASI dTdTCGUCGUGCUGAAGAAGUUC 5·
`21
`1
`
`s· m7GpppG,
`t
`Poly A
`/
`,,,.,.-- AAGCAGCACGACUUCUUCAAG---....
`258 ~
`'-----"""
`238
`I Cleavage on mRNA I
`
`A
`
`B
`
`EGFP mRNA
`
`1-~ -
`
`y o
`
`H
`
`2·-0eoxy
`
`2'-0Me
`
`2· -Fluoro-uridine = 2'FU
`
`2· -Fluoro-cytidine = 2'FC
`
`~
`
`o
`
`Y O
`
`OH
`
`5-Bromo-uridine = U[5Br]
`
`5-lodo-uridine = U[51]
`
`!-\:1
`I \:::1
`
`?
`
`O=P- S
`
`OH
`
`Y O
`
`OH
`
`Thioate linkage= P-S
`
`N3-Methyl Uridine = 3MU
`
`2,6-Dlamlnopurine = DAP
`
`and 2⬘-fluoro-cytidine (2⬘-FC), respec-
`tively, which have a fluoro group at the
`2⬘-position in place of the 2⬘-OH (Fig.
`1B). Where these modified 2⬘-FU, 2⬘-FC
`nucleotides reside in the siRNA se-
`quence are highlighted in red in Figure
`2A. Addition of a 2⬘-fluoro group
`should increase the stability of
`the
`siRNA by making the siRNAs less rec-
`ognizable to RNases, thereby providing
`siRNAs protection from degradation
`(see below). When measured in the dual
`fluorescence assay, 2⬘-FU, 2⬘-FC siR-
`NAs, modified only in the sense strand,
`only in the antisense strand, or in both
`strands, all
`showed decreased EGFP
`fluorescence when normalized to non-
`targeted RFP fluorescence that was com-
`parable to the normalized decrease seen
`with wild-type siRNAs (Fig. 2; Table 1,
`rows 1–4). These results indicated that
`the 2⬘-OH was not required for RNAi
`and that nucleotides modified with 2⬘-
`fluoro groups could be used in siRNA
`constructs to successfully induce RNAi-
`mediated gene silencing.
`To support the conclusion that the
`2⬘-OH was not required for RNAi, ad-
`enine and guanine deoxynucleotides
`that inherently have 2⬘-H in place of the
`2⬘-OH (Fig. 1B) were incorporated into
`the sense, antisense, or both strands of
`2⬘-FU, 2⬘-FC-modified EGFP siRNAs to
`determine their effect on RNAi (Fig. 2A;
`green nucleotides). When 2⬘-FU, 2⬘-FC
`nucleotides were incorporated into the
`EGFP siRNA antisense strand with gua-
`nine and adenine deoxynucleotides at
`positions 9, 10, and 13, which base pair
`with nucleotides lining the cleavage site
`(Fig. 2A), EGFP RNAi effects were al-
`most indistinguishable from wild-type
`levels (Fig. 2B; Table 1, row 5). This
`same antisense construct base-paired to
`2⬘-FU, 2⬘-FC-modified sense strands also showed consider-
`able EGFP silencing at ∼64% (Table 1, row 6). In addition,
`siRNAs that had the entire antisense strand replaced with
`2⬘-FU, 2⬘-FC, dATP, and dGTP nucleotides still showed
`moderate levels of RNAi activity at ∼42%, or ∼44% if the
`sense strand was also modified with 2⬘-FU, 2⬘-FC (Table 1,
`rows 7,8). All together, these results demonstrated that a
`2⬘-OH group was not required for RNAi-mediated degra-
`dation and, even more specifically, was not required for
`nucleotides base-paired with nucleotides lining the mRNA
`cleavage site. There was, however, a limit on the extent to
`
`FIGURE 1. Structures of EGFP siRNA and chemical modifications. (A) Graphical represen-
`tation of dsRNAs used for targeting EGFP mRNA. EGFP was encoded by the pEGFP-C1
`reporter plasmid. siRNAs were synthesized with 2-nt deoxythymidine overhangs at the 3⬘-end.
`The position of the first nucleotide of the mRNA target site is indicated relative to the start
`codon of EGFP mRNA. The sequence of the antisense strand of siRNA is exactly complemen-
`tary to the mRNA target site. (B) Structure and nomenclature of chemical modifications.
`
`the antisense guide siRNA (Elbashir et al. 2001a). The spe-
`cific chemical modifications, the particular siRNA strand(s)
`where modifications were made, and the effect of the
`chemically modified siRNA on RNAi activity are summa-
`rized in Table 1. The RNAi activity of siRNAs was evaluated
`with eight different siRNA concentrations (ranging from 1
`to 200 nM). Each experiment was completed in duplicate
`and repeated twice.
`The effects of modifying the 2⬘-OH of nucleotides on
`RNAi were studied by replacing uridine and cytidine in the
`antisense strand of siRNA with 2⬘-fluoro-uridine (2⬘-FU)
`
`1036
`
`RNA, Vol. 9, No. 9
`
`

`

`
`
`Downloaded from rnajournal.cshlp.org on December 26, 2017 - Published by Cold Spring Harbor Laboratory Press
`
`
`
`
`
`
`
`Chemical modification of siRNAs
`
`TABLE 1. RNA interference mediated by chemically modified siRNAs
`
`Row no.
`
`EGFP siRNA
`
`Sense strand
`
`Antisense strand
`
`RNAi activity
`(%)
`
`RNAi activity
`(+ or −)
`
`1
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`12
`13
`14
`15
`16
`17
`18
`19
`20
`21
`22
`23
`24
`25
`26
`27
`28
`29
`30
`31
`32
`
`Unmodified Unmodified
`DS (WT)
`2⬘-FU, FC
`SS/AS-2⬘-FU, FC
`Unmodified
`2⬘-FU, FC
`SS-2⬘-FU, FC/AS
`Unmodified
`2⬘-FU, FC
`2⬘-FU, FC
`DS-2⬘-FU, FC
`2⬘-FU, FC + (9, 10, 13) dA, dG
`SS/AS-2⬘-FU, FC + (9, 10, 13) dA, dG
`Unmodified
`SS-2⬘-FU, FC/AS-2⬘-FU, FC + (9, 10, 13) dA, dG 2⬘-FU, FC
`2⬘-FU, FC + (9, 10, 13) dA, dG
`SS/AS-2⬘-FU, FC + dA, dG
`2⬘-FU, FC + dA, dG
`Unmodified
`SS-2⬘-FU, FC/AS-2⬘-FU, FC + dA, dG
`2⬘-FU, FC
`2⬘-FU, FC + dA, dG
`SS/AS-Deoxy
`Unmodified Deoxy
`SS-Deoxy/AS
`Deoxy
`Unmodified
`DS-Deoxy
`Deoxy
`Deoxy
`SS/AS-2⬘-OMe
`2⬘-OMe
`Unmodified
`SS-2⬘-OMe/AS
`2⬘-OMe
`Unmodified
`DS-2⬘-OMe
`2⬘-OMe
`2⬘-OMe
`SS/AS-P-S
`Unmodified
`P-S
`SS-P-S/AS
`P-S
`Unmodified
`DS-P-S
`P-S
`P-S
`SS/AS-2⬘-FU, FC + P-S
`2⬘-FU, FC + P-S
`Unmodified
`SS/AS-U[5Br]
`Unmodified U[5Br]
`SS/AS-U[5I]
`Unmodified U[5I]
`SS/AS-DAP
`Unmodified DAP
`SS-2⬘-FU, FC/AS-U[5Br]
`2⬘-FU, FC
`U[5Br]
`SS-2⬘-FU, FC, FC/AS-U[5I]
`2⬘-FU, FC
`U[5I]
`SS-2⬘-FU, FC/AS-DAP
`2⬘-FU, FC
`DAP
`SS/AS-3MU
`Unmodified
`3MU
`SS/AS-(11) 3MU
`Unmodified
`(11) 3MU
`SS/AS-(1, 2) mm
`Unmodified
`(1, 2) mm
`SS/AS-(18, 19) mm
`Unmodified
`(18, 19) mm
`SS/AS-2⬘-FU, FC + (1-13) dA, dG
`2⬘-FU, FC + (1-13) dA, dG
`Unmodified
`SS-2⬘-FU, FC/AS-2⬘-FU, FC + (1-13) dA, dG
`2⬘-FU, FC
`2⬘-FU, FC + (1-13) dA, dG
`SS/AS-2⬘-FU, FC + (9-19) dA, dG
`2⬘-FU, FC + (9-19) dA, dG
`Unmodified
`SS-2⬘-FU, FC/AS-2⬘-FU, FC + (9-19) dA, dG
`2⬘-FU, FC
`2⬘-FU, FC + (9-19) dA, dG
`
`93 ± 0.70
`83 ± 3.48
`92 ± 0.98
`83 ± 0.01
`85 ± 2.10
`64 ± 2.89
`42 ± 1.66
`44 ± 0.60
`0 ± 5.97
`38 ± 2.95
`0 ± 0.01
`16 ± 4.41
`25 ± 1.75
`0 ± 0.01
`42 ± 6.03
`62 ± 0.07
`47 ± 0.03
`22 ± 0.03
`70 ± 1.88
`59 ± 11.2
`51 ± 0.57
`31 ± 1.88
`42 ± 5.02
`35 ± 7.69
`0 ± 6.65
`0 ± 1.71
`35 ± 5.69
`77 ± 2.00
`43 ± 0.09
`45 ± 2.23
`91 ± 0.36
`64 ± 0.42
`
`++++
`++++
`++++
`++++
`++++
`+++
`++
`++
`−
`+
`−
`−
`+
`−
`++
`+++
`++
`+
`+++
`+++
`++
`+
`++
`+
`−
`−
`+
`+++
`++
`++
`++++
`+++
`
`Summary of the specific chemical modifications analyzed, the particular siRNA strand(s) modified, and the effect of the chemically modified
`siRNA on RNAi activity in HeLa cells. RNAi activity was quantified by the dual fluorescence assay and is presented as the inhibition efficiency
`of target gene (EGFP) expression when cells were treated with 50 nM modified siRNAs. For comparison, the RNAi activity of unmodified, or
`wild-type, duplex siRNA (DS) normalized to 93% was designated (++++). Modified siRNAs assigned (++++) showed >80% RNAi activity, (+++)
`showed 60%–80%, (++) showed 40%–60%, and (+) showed 20%–40%. Modified siRNAs showing <20% RNAi activity were considered
`nonfunctional (−) in the RNAi pathway. Each experiment measuring RNAi activity of siRNAs was completed in duplicate and repeated twice.
`
`which deoxynucleotides could substitute for ribonucleo-
`tides because replacing the entire siRNA sense strand with
`deoxynucleotides decreased EGFP gene silencing to ∼38%
`inhibition, and replacing either the antisense strand or both
`strands entirely with deoxynucleotides completely abolished
`EGFP RNAi (Fig. 2B; Table 1, rows 9–11). Nonetheless,
`these results collectively showed that nucleotides with either
`2⬘-F or 2⬘-H groups can selectively replace ribonucleotides
`within the siRNA sequence to effectively induce RNAi.
`An interesting result was seen by modifying the 2⬘-OH to
`a bulky methyl group to create 2⬘-OMe nucleotides that
`were incorporated into sense, antisense, or both strands of
`EGFP siRNAs (Fig. 1B). This modification was hypoth-
`esized to improve RNAi efficacy because 2⬘-OMe groups are
`thought to increase RNA stability by inducing an altered
`RNA conformation that is more resistant to nucleases
`(Cummins et al. 1995). This modification is also thought to
`
`increase RNA affinity for RNA targets and improve hybrid-
`ization kinetics (Majlessi et al. 1998). Despite these poten-
`tial benefits, 2⬘-OMe nucleotides incorporated into either
`the sense or antisense strand greatly diminished EGFP gene
`silencing to ∼25% or ∼16%, respectively, whereas double-
`stranded 2⬘-OMe-modified siRNAs completely abolished
`RNAi (Table 1, rows 12–14). These results indicated that
`the methyl group, as a bulky group, may severely limit the
`interactions between siRNAs, target mRNAs, and the RNAi
`machinery required for successfully mediating RNAi. It is
`worth noting that because the bulkiness of the methyl group
`would likely be the cause of decreased RNAi activity rather
`than the actual lack of the 2⬘-OH specifically, these studies
`still supported the conclusion that the 2⬘-OH was not re-
`quired for RNAi.
`In a final analysis of modifications that may potentially
`increase siRNA stability without disrupting RNAi potency,
`
`www.rnajournal.org
`
`1037
`
`

`

`
`
`rnajournal.cshlp.org Cold Spring Harbor Laboratory Press on December 26, 2017 - Published by
`
`
`
`
`
`Downloaded from
`
`Chiu and Rana
`
`A
`
`S' GCAGCACGACUUCUUCAA TdT
`c CGUCGUGCUGAAGAAGUUC s·
`
`s· GCAGCACGACUUCUUCAAG TdT
`dTd CGUCGUGCUGAAGAAGUUC s·
`
`WTDS siRNA
`
`SS/AS-2'FU, FC
`
`2' structure
`(cid:127) -OH
`(cid:127) -ti
`(cid:127) -F
`
`s· GCAGCACGACUUCUUCAAGHdT
`dTdTCGUCGUG GAAGAAGUUC s·
`
`SS/AS -Deoxy siRNA
`
`s· GCAGCACGACUUCUUCAAG JTdT
`Sense strand (SS)
`slRNA Antlsense strand (AS) dT TCGUCGUGCUG GAAGUUC 5'
`
`m7GpppG~ AAGCAGCACGACUUCUUCAAG--.....__,,, Poly A
`
`SS/AS -2'FU, FC+ (9, 10, 13) dA, dG
`
`EGFP mRNA
`
`Cleavage on mRNA
`
`B
`
`1.40 . - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - ,
`
`1.20
`
`~
`~ 1.00
`0:::
`a:
`~ ffi 0.80
`
`"C
`Cl)
`N
`: : 0,60
`CV
`
`e 0
`
`Z 0.40
`
`~-"
`
`1.
`
`❖
`❖
`
`2
`
`3
`
`N
`
`4
`.,,
`
`5
`6
`~ SI
`
`7
`
`8
`
`N
`
`0.20
`
`0.00
`
`.,.
`8
`:IE
`
`EGFP
`siRNA
`(nM)
`
`10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30
`9
`It) ~ !<l
`SI
`It) ~ !<l SI
`It) ~ !<l SI
`
`0
`0
`~ !<l
`
`N
`
`0
`0
`~ !<l
`
`N
`
`0
`0
`~ !<l
`
`OS
`
`SS/AS-Oeoxy
`
`SS/AS-2'FU, FC
`
`SS/AS -2'FU, FC +
`(9, 10, 13) dA, dG
`
`FIGURE 2. siRNA 2⬘-OH is not required to guide mRNA cleavage. (A) Sequence and structure of siRNA duplexes with modification at the
`2⬘-position of the sugar unit. Nucleotides with 2⬘-hydroxyl groups (-OH) are black. Nucleotides with 2⬘-deoxy groups (-H) are cyan. Nucleotides
`with 2⬘-fluoro groups (-F) are red. The cleavage site on the target mRNA is also shown (red arrow). (B) Ratios of normalized GFP to RFP
`fluorescence intensity in lysates from modified siRNA-treated HeLa cells. The fluorescence intensity ratio of target (GFP) to control (RFP)
`fluorophores was determined in the presence of EGFP siRNA duplexes with modifications at the 2⬘-position of the sugar unit. Normalized ratios
`at <1.0 indicate specific RNA interference effects. For comparison, results from unmodified duplex siRNA-treated cells are included.
`
`a thioate linkage (P–S) was integrated into the backbone of
`the EGFP siRNA strand(s). P–S linkages were previously
`used in antisense methodology for increasing resistance to
`ribonucleases (for review, see Stein 1996) and, therefore,
`were postulated to enhance the stability of siRNAs. Incor-
`porating the P–S linkages into the double-stranded siRNA
`sense strand led to moderate levels of RNAi activity (62%
`inhibition), whereas P–S linkages in either the antisense or
`both strands of the siRNAs led to just less than ∼50% RNAi-
`
`induced inhibition (Table 1, rows 15–17). These results im-
`plied that the P–S modifications did not prohibit RNAi-
`mediated degradation and only moderately affected the ef-
`ficiency of RNAi. Interestingly, incorporating 2⬘-FU, 2⬘-FC
`modifications into the antisense strand in addition to the
`added P–S linkages showed lower levels of EGFP gene si-
`lencing (Table 1, row 18), indicating that there was a syn-
`ergistic effect that decreased but did not inhibit RNAi-me-
`diated degradation when both the 2⬘-F groups and the
`
`1038
`
`RNA, Vol. 9, No. 9
`
`

`

`Downloaded from
`
`
`
`rnajournal.cshlp.org Cold Spring Harbor Laboratory Press on December 26, 2017 - Published by
`
`
`
`
`
`Chemical modification of siRNAs
`
`P–S linkages were incorporated into
`siRNAs.
`
`A
`
`(a)
`
`- - AS OS
`
`-- AS-2'FU, FC
`-- SSIAS-2'FU, FC
`
`DS-2'FU, FC
`
`10
`
`20
`
`"'
`"'
`Tlme(mln)
`
`50
`
`60
`
`- - AS
`- - OS
`_.,_ AS-P-S
`_.,_ SS/AS-P-S
`_.,_ 05-P-S
`
`100.0%
`
`.,_ ..
`
`80.0%
`
`< z
`a,:
`·;;;
`0
`~ 40.0%
`.5
`
`20.0%
`
`100.0%
`
`80.0"fo
`
`< z 60.0%
`a,:
`·;;
`0
`~ 40.0%
`.5
`
`20.0%
`
`(b)
`
`Stability of modified siRNAs and the
`persistence of their RNAi activity in
`vitro and in vivo
`
`As the above experiments showed that
`siRNAs modified with stabilizing 2⬘-FU,
`2⬘-FC groups could effectively mediate
`RNAi to levels comparable to wild type,
`it was necessary to show that
`these
`modifications did in fact enhance siRNA
`stability. To measure the stability of
`siRNA in cell extracts, unmodified or
`2⬘-FU, 2⬘-FC-modified EGFP antisense
`5⬘-labeled
`strand
`siRNAs
`with
`[␥-32P]ATP were annealed with sense
`strand siRNAs to form duplex siRNAs,
`which were then incubated in HeLa cell
`extracts. At various time points, siRNAs
`were extracted, analyzed on a 20% poly-
`acrylamide gel containing 7 M urea, and
`visualized by phosphorimager analysis.
`Smaller siRNA degradation products
`were visualized in this analysis (data not
`shown), indicating that the lost of intact
`siRNA observed during these experi-
`ments was not caused by dephosphory-
`lation of siRNAs. The top panel (a) of
`Figure 3A shows the stability of the vari-
`ous 2⬘-FU, 2⬘-FC-modified siRNAs as
`compared with wild-type siRNAs over
`time. Wild-type
`double-stranded
`siRNAs showed a steady loss of intact
`siRNAs over the course of the experi-
`ment, with only ∼7% of the original
`concentration of intact siRNAs remain-
`ing after 1 h inextract (Fig. 3A[a], dark
`blue line). Intact modified or unmodi-
`fied single-stranded antisense siRNAs
`were quickly lost over the time course
`and were virtually undetectable by 30
`min in extract (Fig. 3A[a], black and red
`lines). This result showed that single-
`stranded modified siRNA was as suscep-
`tible to degradation as wild-type siRNA,
`indicating that single-stranded siRNAs,
`modified or unmodified, are inherently
`less stable than duplex siRNA. Double-
`stranded siRNAs with 2⬘-FU, 2⬘-FC
`modifications in either the antisense
`strand or both strands remained pre-
`dominantly intact over the course of the
`experiment with ∼68% or ∼81%, respec-
`
`10
`
`20
`
`"'
`30
`Time(min)
`
`B
`
`1 . 20
`
`a. 1.00
`LL.
`
`f 0 .80
`
`LL.
`C,
`w
`'Cl 0 .60
`G.I
`N
`ii 0 .40
`E
`0 z 0 . 20
`
`EGFP siRNA
`aMOCK
`EIWT OS
`D DS-2'FU, FC
`
`o.oo
`
`6
`
`1 8
`
`30
`
`42
`
`54
`66
`Time {h)
`
`78
`
`90
`
`120
`
`144
`
`FIGURE 3. Extending the half-life of siRNA duplexes prolongs the persistence of RNA inter-
`ference in vivo. (A) Comparing the stability of unmodified siRNAs with siRNAs containing
`2⬘-fluoro-uridine and 2⬘-fluoro-cytidine (2⬘-FU, 2⬘-FC) modifications (a) and thioate linkage
`(P–S) modifications (b). Unmodified or modified EGFP antisense strand siRNAs (AS) were
`5⬘-labeled with [␥-32P]ATP by T4 polynucleotide kinases. Duplex siRNAs were formed by
`annealing equal molar ratios of sense-strand (SS) siRNAs with the 5⬘-32P-labeled antisense
`strand. To analyze siRNA stability in HeLa cell extract, 50 pmole of siRNA was incubated with
`500 µg of HeLa cell extract in 50 µL of reaction mixture containing 20 mM HEPES (pH 7.9),
`100 mM KCl, 10 mM NaCl, 2 mM MgCl2, and 10% glycerol. At various time points, siRNAs
`were extracted and analyzed on 20% polyacrylamide gels containing 7 M urea followed by
`phosphorimage analysis (Fugi). (B) Kinetics of RNAi effects of duplex siRNA with 2⬘-fluoro-
`uridine and 2⬘-fluoro-cytidine modification in HeLa cells over a 144-h time course. The
`fluorescence intensity ratio of target (GFP) to control (RFP) protein was determined in the
`presence of unmodified dsRNA (blue bars) and duplex siRNA with 2⬘-fluoro-uridine and
`-cytidine modifications (DS-2⬘-FU, 2⬘-FC, cyan bar) and normalized to the ratio observed in
`the presence of mock-treated cells (red bars). Normalized ratios at <1.0 indicated specific RNA
`interference.
`
`www.rnajournal.org
`
`1039
`
`

`

`
`
`Downloaded from rnajournal.cshlp.org on December 26, 2017 - Published by Cold Spring Harbor Laboratory Press
`
`
`
`
`
`
`
`Chiu and Rana
`
`tively, of the original siRNA population remaining intact
`throughout the duration of the experiment (Fig. 3A[a],
`green and light blue lines). These results indicated that the
`2⬘-FU, 2⬘-FC modifications did, indeed, increase the stabil-
`ity of the siRNAs upon exposure to extract and that having
`these modifications in both strands provided the siRNAs
`with the most stability.
`In a similar experiment, the stability of P–S-modified
`EGFP siRNAs was evaluated. Unmodified, double-stranded
`antisense siRNAs showed about the same rate of siRNA loss
`as described in the above experiment (Fig. 3A[b], dark blue
`lines). However, P–S-modified single-stranded antisense
`siRNAs demonstrated a markedly increased rate of stability
`over the course of the experiment, showing ∼63% of the
`original siRNAs remaining intact after 1 h inextract as
`compared with 0% intact for single-stranded unmodified
`antisense siRNAs (Fig. 3A[b], black and red lines). The
`stability of double-stranded siRNAs with P–S modifications
`in both strands was comparable to the stability seen with the
`modified single-stranded antisense strand, with ∼63% of the
`original siRNA population remaining intact after 1 h (Fig.
`3A[b], light blue lines). Double-stranded siRNAs with P–S
`modifications in only the antisense strand showed weaker
`but still significant stability with ∼42% of the original
`siRNA population remaining intact through 1 h in extract
`(Fig. 3A[b], green lines). These results showed that the P–S
`modifications increased the stability of the siRNAs and,
`most notably, increased the stability of both single- and
`double-stranded siRNAs.
`These in vitro results indicated that siRNA stability is
`prolonged by these different modifications; however, it is
`important to note that these experiments address the gen-
`eral stability of siRNA in the context of endonucleases
`present in whole-cell extracts. Therefore, these experiments
`cannot distinguish whether the endonucleases affecting
`siRNAs in the in vitro assay would necessarily affect the
`stability of
`these various siRNAs in vivo. To address
`whether increased stability seen with modified siRNAs pro-
`longed the duration of RNAi in vivo, RNAi, induced by
`unmodified and 2⬘-FU, 2⬘-FC-modified double-stranded
`EGFP siRNAs, was assayed in the dual fluorescence reporter
`assay over a period of 144 h. To visualize RNAi effects over
`an even longer period of time, HeLa cells were transfected
`with modified or unmodified siRNA and, 36 h later, trans-
`fected with dual fluorescence reporter plasmids; RNAi ac-
`tivity persisted but was tapering by 168 h (data not shown).
`Also, growth of cells containing modified siRNAs was com-
`parable to cells containing wild-type siRNA, indicating that
`modified siRNAs were not affecting cell division (data not
`shown). Although 2⬘-FU, 2⬘-FC-modified EGFP siRNAs
`were slower to show RNAi effects by 6–18 h, maximal RNAi
`effects occurred by 42 h posttransfection for both modified
`and unmodified siRNAs. The maximal activity for both
`siRNAs was also in the same range, with both showing
`∼85%–90% inhibition of GFP expression. However, the
`
`1040
`
`RNA, Vol. 9, No. 9
`
`RNAi effects observed over the period of 66–120 h revealed
`that the effect of modified siRNAs was much more persis-
`tent than that of unmodified siRNAs. By 120 h posttrans-
`fection, the effect of modified siRNAs still remained at
`∼80% inhibition of GFP expression but the effect of un-
`modified siRNAs had dropped to less than ∼40% inhibition.
`Similarly, prolonged RNAi activity was observed with
`2⬘-FU, 2⬘-FC-modified siRNAs targeting endogenous hu-
`man Cyclin T1 mRNA when compared with wild-type siR-
`NAs targeting Cyclin T1 (see Discussion; Y.L. Chiu and
`T.M. Rana, unpubl.). Altogether, these results strongly in-
`dicated that there was a direct link between the duration of
`the RNAi effects and siRNA stability in human cells. Fur-
`thermore, these results showed conclusively that siRNAs
`stabilized by chemical modifications, like the 2⬘-FU, 2⬘-FC
`modifications, can be used to effectively induce and signifi-
`cantly prolong RNAi-mediated gene silencing in vivo.
`
`Modified siRNAs that stabilize A–U base-pair
`interactions can induce RNAi
`
`In addition to incorporating modifications that affected the
`stability of siRNAs, nucleotides chemically modified to
`strengthen the base-pair interactions between two comple-
`mentary bases were analyzed. In theory, increasing the sta-
`bility of base-pair interactions may increase the targeting
`efficiency of siRNAs to target mRNA sequences. Increasing
`targeting efficiency may then induce more robust RNAi
`effects with siRNAs that are weaker at binding to their target
`sequence or have mismatched sequences, and thus, are not
`showing a high degree of RNAi. This type of approach may
`also be used to significantly inhibit expression of one allele
`over another when both alleles are present in the same cell.
`To bolster base-pairing interactions, 5-bromo-uridine
`(U[5Br]), 5-iodo-uridine (U[5I]), or 2,6-diaminopurine
`(DAP; Fig. 1B), which are modified nucleotides known to
`increase the association constant between A–U base pairs
`(Saenger 1984), were incorporated into siRNAs and tested
`in the dual
`fluorescence report assay. Double-stranded
`siRNAs having U[5Br], U[5I], or DAP modifications incor-
`porated into the antisense strand were capable of inducing
`RNAi activity at levels of ∼70% for U[5Br], ∼59% fo

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket