Download as pdf or txt
Download as pdf or txt
You are on page 1of 120

Virus Research 102 (2004) 1–2

Preface

In the last 2 years, we have witnessed the implementation including transposon mobilization, silencing of transgenes,
of a new technology that promises to revolutionize the way regulation of some of the early stages of development, as
post-genomic research in the mammalian cells is conducted, well as in the regulation of expression at the genome level.
and the field of virology will not lag behind in this revolution. In plants RNAi represents a defense mechanism against vi-
The new technology is based on a phenomenon known as ral infections, and it might also have the same role in inver-
RNA interference (RNAi), RNA silencing, or postranscrip- tebrate organisms. It is unclear whether RNAi serves as an
tional gene silencing, which is an evolutionarily conserved antiviral defense mechanism in mammals or not. However,
mechanism found to operate in fungi, plants, and animals. the observation that this system is functional in vertebrates
RNAi is a process that responds to double-stranded RNA prompted several groups to explore the interaction between
(dsRNA) by silencing the expression of the gene homolo- virus infection and the RNA silencing machinery in mam-
gous to the dsRNA sequence. malian cells.
RNAi is triggered in plants and invertebrates by long The application of this technology in the field of animal
dsRNA molecules which are processed by a dsRNA-specific virology is just the beginning, but it has already proven
RNAse, named Dicer, into small fragments of 20–25 base to constitute an insightful method for the characterization
pairs known as short interfering RNAs (siRNAs). These of viral gene function. RNAi will be especially helpful
siRNAs selectively associate with a multiprotein complex to study those viruses for which a reverse genetics sys-
(RISC, for RNA-induced silencing complex), which un- tem has not been implemented. It is foreseeable that the
winds the siRNA duplex to produce a single-stranded RNA, studies employing RNAi will soon extend to the analysis
which further guides RISC to the target mRNA. This mRNA of cellular genes that interact with the viral components
is then cleaved by a ribonuclease component of RISC, silenc- of virus replication cycles, and, more in the mid term,
ing the expression of the cognate gene. SiRNAs can also be to the analysis of viral gene function in complete animal
generated endogenously from precursor hairpin RNA struc- models.
tures 60–90 nucleotides in length, encoded in the genome. There are also high hopes for applications of RNAi in
Those are processed by Dicer to yield single-stranded RNAs the treatment of viral diseases, as therapeutic or prophylac-
that bind to RISC and arrest the translation of the target tic tools. This may be particularly relevant in the case of
mRNA without promoting its degradation. The small regu- viruses that establish chronic infections (e.g., human im-
latory endogenous RNAs which act in this way and of which munodeficiency virus, hepatitis B and C viruses) or latent in-
hundreds have been described in animals and plants, are fections (e.g., herpes viruses). The main challenge for these
known as microRNAs (miRNAs). Their sequences are not applications is the development of long-lasting, efficient and
absolutely complementary to the target mRNAs, as in the tissue-specific methods to deliver the siRNAs to whole or-
case of the siRNAs, but contain mismatches. ganisms.
The RNAi pathway was more difficult to be demonstrated The opening chapter reviews the role of siRNAs and
in mammalian cells in which the dsRNA fragments longer miRNAs in RNAi. The emphasis of this Special Issue is
than 30 bp activate components of the non-specific interferon on reviews which describes the effectiveness of RNAi to
response, that mediates a generalized inhibition of gene ex- silence gene expression in animal viruses. These include
pression through the non-specific shutdown of protein syn- viruses with different genome replication strategies, such
thesis and mRNA degradation. The recent observation that as positive, negative and double-stranded RNA viruses as
synthetic 21-nt siRNA duplexes effectively inhibit mam- well as retroviruses (Chapters 2–8). Additional chapters
malian endogenous gene expression in a sequence-specific review RNAi applied to insect and plant virus replication
manner without activating the non-specific interferon re- (Chapters 9–11), and two chapters deal with suppres-
sponse, offered a great opportunity to study the func- sors of RNA silencing (Chapter 12) and application of
tion of vertebrate genes, and the genes of vertebrate RNAi as antivirals in plants and animals (Chapter 13).
viruses. The last chapter (Chapter 14) outlines the strategies to
RNAi has been proposed to play a central role in the reg- generate an RNA expression library to analyse a whole
ulation of many cellular processes at the translation level genome.

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.008
2 Preface / Virus Research 102 (2004) 1–2

The topic of gene silencing is relatively new, and the Susana López∗
reader will find certain repetition in the citation of some Carlos F. Arias
references and (to a lesser extent) in the text. We decided Dept. de Genetica del Desarrollo y Fisiologia Molecular
not to edit this out. We considered that any harsh editing Instituto de Biotecnologia, Universidad Nacional
would diminish the excitement for the reader to participate Autonoma de Mexico, Avenida Universidad 2001
in the explosion of high quality knowledge increase based on Col. Chamilpa, Cuernavaca, Morelos 62210, Mexico
relatively few but excellent papers in basic science. Finally, ∗ Corresponding author. Tel.: +52-777-3291615

we thank all the contributors for taking the time to share fax: +52-777-3172388
their experiences in this emerging and exciting field. E-mail address: susana@ibt.unam.mx (S. López)
Virus Research 102 (2004) 3–9

Derivation and function of small interfering RNAs and microRNAs


Bryan R. Cullen∗
Department of Molecular Genetics and Microbiology, Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA

Abstract

Small interfering RNA (siRNA) duplexes are generally produced by Dicer cleavage of double-stranded RNAs of frequently exogenous
origin and can induce the cleavage and degradation of mRNAs bearing an identical sequence. In contrast, microRNAs (miRNAs) are encoded
within the eukaryotic genome as short RNA hairpin structures. While these pre-miRNAs are also processed by Dicer, mature miRNAs appear
to function primarily by inhibiting the translation of mRNAs bearing multiple, partially mismatched target sites. Nevertheless, recent data
argue that the posttranscriptional regulatory machinery utilized by siRNAs and miRNAs is largely or entirely identical. In this review, I will
discuss recent progress in unraveling the RNA processing pathway utilized for the biosynthesis of mature miRNAs and argue that this pathway
offers at least three distinct entry points for the functional expression of artificial siRNAs in vertebrate cells. While each of these entry points
offers distinct advantages and disadvantages, they all have the potential to induce the effective knock-down of specific genes either in cell
culture or in experimental animals.
© 2004 Elsevier B.V. All rights reserved.

Keywords: miRNA; RNA interference; RNA processing; siRNA

1. Introduction 2000). This cleavage is mediated by a processive cytoplas-


mic RNase III family enzyme called Dicer (Bernstein et al.,
RNA interference (RNAi) was first defined in C. ele- 2001a; Grishok et al., 2001; Hutvágner et al., 2001; Knight
gans as a mechanism that induced the posttranscriptional and Bass, 2001). One strand of this siRNA duplex is then
silencing of genes in response to the introduction of long selectively incorporated into a large protein complex termed
double-stranded RNAs (dsRNAs) of identical sequence (Fire the RNA induced silencing complex or RISC (Hammond
et al., 1998). It was soon realized that RNAi was closely et al., 2000; Nykänen et al., 2001). This RNA strand then
related to previously described, but poorly understood, gene acts as a guide RNA to target RISC to homologous mR-
inactivation pathways in plants and certain fungi (reviewed NAs (Martinez et al., 2002; Schwarz et al., 2003). Once
by Bernstein et al., 2001b). While long dsRNAs also proved RISC is bound, a currently unidentified ribonuclease com-
highly effective at inducing RNAi in Drosophila (Hammond ponent cleaves the target mRNA opposite the center of the
et al., 2000), initial efforts to demonstrate RNAi in verte- bound guide siRNA (Fig. 1). RISC is then released, to seek
brate cells were largely unsuccessful. This failure resulted additional mRNA targets, while the two fragments of the
from the fact that long dsRNAs also activate the vertebrate mRNA are degraded by cellular exonucleases (Hutvágner
interferon response, a complex defense system, lacking in and Zamore, 2002).
non-vertebrate species, that leads to a global posttranscrip- A critical discovery, made initially in Drosophila and
tional inhibition of gene expression (Sen, 2001). subsequently confirmed in vertebrate cells, is that synthetic
Efforts to understand how RNAi works in plants and siRNA duplexes can also program RISC and induce the in-
invertebrates demonstrated that long dsRNAs are initially activation of specific target genes (Elbashir et al., 2001a,b).
cleaved into characteristic ∼21 nt dsRNAs, bearing 2 nt 3 This result was made possible in vertebrate cells by the
overhangs, termed small interfering RNA (siRNA) duplexes fact that the interferon response is activated by dsRNAs of
(Fig. 1) (Hamilton and Baulcombe, 1999; Hammond et al., >30 bp (Manche et al., 1992), while siRNA duplexes bear
a double-stranded region of only ∼19 bp. This observa-
tion demonstrated that the RNAi pathway was indeed fully
∗ Tel.: +1-919-684-3369; fax: +1-919-681-8979. active in mammalian cells and raised the possibility, now
E-mail address: culle002@mc.duke.edu (B.R. Cullen). confirmed, that RNAi could be used to perform reverse

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.009
4 B.R. Cullen / Virus Research 102 (2004) 3–9

Fig. 1. MicroRNA and siRNA processing and function. A proposed processing pathway for endogenously encoded miRNAs is shown at the center of
the figure. This proceeds from transcription through nuclear processing, nuclear export, cytoplasmic processing by Dicer and finally to incorporation into
RISC. This results in the initial primary miRNA transcript (pri-miRNA) being processed to a pre-miRNA and finally to a miRNA duplex intermediate,
one strand of which then serves as a substrate for RISC incorporation. The assembled RISC can then target mRNAs bearing a perfectly complementary
target site for degradation or can inhibit the translation of an mRNA that contains multiple, partly mismatched target sites. Entry points for designed
siRNAs are shown at the left of the figure. These include synthetic siRNA duplexes as well as short hairpin RNAs, transcribed from expression plasmids,
that closely mimic pre-miRNAs. Finally, artificial miRNA genes can also be used to express novel siRNAs. Long dsRNAs, which can generate siRNAs
in invertebrate and plant cells, activate the interferon response in vertebrate cells and are therefore not useful in the latter system.

genetic analyses in human cells (reviewed by Dykxhoorn et al., 2001; Lau et al., 2001; Lee and Ambros, 2001). These
et al., 2003). RNAs, which are now termed microRNAs (miRNAs), are
predicted to form part of one strand of an imperfect precur-
sor RNA hairpin structure of between 60 and 90 nucleotides
2. The discovery of microRNAs in length. This contrasts with siRNAs, which are invariably
derived from long dsRNAs. In addition, while miRNAs are
As noted above, long dsRNAs are processed by Dicer to by definition of endogenous origin, siRNAs can be endoge-
give ∼21 bp siRNA duplexes, one strand of which is then nous or exogenous.
incorporated into RISC. Efforts in the Drosophila and C. el- While the discovery of this large family of miRNAs was
egans systems to clone these short non-coding RNAs based unexpected, two miRNAs had in fact been known for several
on their size led to the surprising discovery that not only years in C. elegans. These miRNAs, termed let-7 and lin-4,
these organisms, but also vertebrates, express over 200 ge- emerged from mutational screens designed to identify genes
nomically encoded ∼21 nucleotide RNAs (Lagos-Quintana involved in regulating larval development in C. elegans
B.R. Cullen / Virus Research 102 (2004) 3–9 5

(Lee et al., 1993; Reinhart et al., 2000). Both let-7 and lin-4 RNA polymerase II, and it is therefore likely that these
are 21 nt in length and both are initially transcribed as one pri-miRNAs are capped and polyadenylated. It remains pos-
arm of an ∼70 nt stem-loop RNA precursor. Both let-7 and sible that other pri-miRNAs might be transcribed by other
lin-4 are expressed in a developmentally regulated manner cellular polymerases.
and both inhibit the expression of specific, developmentally The first step in miRNA processing is the excision
important mRNAs by forming duplexes with multiple con- of the ∼70 nt miRNA hairpin intermediate, termed the
served sequence elements present in their 3 untranslated pre-miRNA, from the much longer pri-miRNA (Fig. 1) (Lee
region (3 UTR). However, unlike siRNAs, this interaction et al., 2002; Zeng and Cullen, 2003). This cleavage, which
specifically blocks the translation of these target mRNAs, occurs largely or exclusively in the nucleus, is mediated by
rather than inducing their degradation (Fig. 1) (Olsen and an enzyme termed RNase III or Drosha, a member of the
Ambros, 1999). Interestingly, known target sequences for family of large RNases that also includes Dicer (Lee et al.,
let-7 and lin-4 bear central mismatches so that the bound 2003b). Analysis of the pre-miRNA cleavage product that
let-7 and lin-4 miRNAs would be predicted to form bulges results from processing of the miR-30 pri-miRNA shows
(Lee et al., 1993; Reinhart et al., 2000). Recent evidence that this cleavage both defines the 3 end of the miR-30
indicates that it is these mismatches that prevent target miRNA and leaves a 2 nt 3 overhang (Lee et al., 2002;
mRNA cleavage by these miRNAs, and not some intrinsic Zeng and Cullen, 2003). Importantly, this cleavage does
difference in miRNA versus siRNA function (see below). not occur at the base of the stem of the predicted 80 nt
Although many different miRNAs have been reported in miR-30 precursor RNA hairpin but rather ∼7 nt above the
a range of species, very few have as yet been assigned a base, resulting in a 63 nt pre-miRNA intermediate. The se-
function. Exceptions include not only let-7 and lin-4 in C. quences that determine the sites of cleavage of the miR-30
elegans but also two miRNAs termed bantam and miR-14 in pri-miRNA by RNase III remain to be defined. However,
Drosophila, miR-171 in Arabidopsis and miR-23 in human it is clear that the helical nature of the basal region of the
cells (Llave et al., 2002; Brennecke et al., 2003; Kawasaki miR-30 stem-loop precursor plays a critical role and that
and Taira, 2003; Xu et al., 2003). As in the case of let-7 insertions or deletions in the precursor stem can result in a
and lin-4, these miRNAs are expressed in a developmen- shift in the cleavage sites (Zeng and Cullen, 2003).
tally regulated manner and inhibit the expression of target The next step in the formation of the mature miR-30
mRNAs bearing complementary sequences. Consistent with miRNA is export of the pre-miRNA to the cytoplasm. Re-
the hypothesis that other miRNAs may also play a role in cent evidence (Yi et al., 2003) indicates that the factor re-
development, many vertebrate and non-vertebrate miRNAs sponsible for nuclear export of pre-miRNAs is Exportin 5
have been found to be expressed in a tissue specific and/or (Exp5), a member of the karyopherin family of nucleocy-
developmentally regulated manner (Lagos-Quintana et al., toplasmic transport factors (Fig. 1) (reviewed by Cullen,
2001, 2002; Lau et al., 2001; Lee and Ambros, 2001). More- 2003). Exp5 is known to mediate the nuclear export of a
over, mutational disruption of genes encoding proteins that range of other small non-coding RNAs, including the aden-
play a global role in miRNA biogenesis, such as Dicer, has ovirus VA1 RNA and the human Y1 RNA (Gwizdek et al.,
profound deleterious affects on the development of mutant 2003). Like other karyopherin family members involved in
organisms (Grishok et al., 2001; Park et al., 2002). nuclear export, Exp5 is dependent on the GTP-bound form of
the Ran co-factor for specific binding to its export substrate
in the cell nucleus and is predicted to release its RNA cargo
3. Expression, processing and function of microRNAs in the cytoplasm after hydrolysis of Ran·GTP to Ran·GDP
by the cytoplasmic Ran GTPase activating protein. Although
The emerging importance of miRNAs in regulating the pre-miRNAs are recognized as specific substrates for Exp5
appropriate development and differentiation of multicellular mediated nuclear export (Yi et al., 2003), the characteristics
organisms, and the similarity of miRNAs to siRNAs, has of the pre-miRNA that mediate this recognition remain to
prompted efforts to more fully understand miRNA deriva- be determined.
tion and function. Although these studies remain incom- Dicer is a processive, ATP-dependent RNase that binds
plete, the pathway delineated in Fig. 1 is likely to represent with high affinity to the ends of dsRNAs bearing 2 nt 3
a fairly accurate overview of this process. overhangs, i.e. the product that it normally generates during
miRNAs are found either as individual miRNA genes or in processing of long dsRNAs (Zhang et al., 2002). Dicer then
tandemly arrayed clusters that may suggest a functional re- cleaves both RNA strands ∼21 nt from the bound end. The
lationship analogous to a bacterial operon (Lagos-Quintana structure of the pre-miRNA, which consists of an RNA hair-
et al., 2001; Lau et al., 2001). In any event, the miRNA pin ending with a 2 nt 3 overhang (Zeng and Cullen, 2003),
genes that have been studied in detail thus far, such as is closely analogous to intermediates generated during pro-
human miR-30, appear to be transcribed as part of a sev- cessing of long dsRNAs except that one end is closed by
eral hundred nucleotide long primary transcript termed a a loop. Importantly, the fact that Dicer cleaves at a prede-
pri-miRNA (Fig. 1) (Lee et al., 2002; Zeng and Cullen, termined distance from the end of the pre-miRNA hairpin
2003). In at least some cases, transcription is mediated by means that the structure of the pre-miRNA predetermines
6 B.R. Cullen / Virus Research 102 (2004) 3–9

the sequence of the miRNA duplex intermediate that is gen- tive, particularly at lower levels of miRNA/siRNA expres-
erated by Dicer processing. This emphasizes the importance sion, than is inhibition via mRNA cleavage. However, the
of the initial, nuclear RNA processing event in determining requirement for binding of multiple RISC complexes for ef-
the final product of this pathway (Fig. 1). fective mRNA translation inhibition does offer the potential
Although it is now clearly established that Dicer process- for coordinate regulation of mRNA expression by multiple,
ing of pre-miRNAs gives rise to a duplex RNA intermediate, distinct miRNAs.
this is generally short-lived and therefore hard to detect. Although it initially seemed possible that miRNAs and
However, while the majority of miRNA precursors give rise siRNAs might be functionally distinct, with the former in-
to only a single mature miRNA, a small number, including ducing translational inhibition and the latter mRNA cleav-
miR-30, do give rise to mature miRNAs derived from both age, this is no longer believed to be true. Endogenously
strands (Zeng et al., 2002). This allows the structure of the encoded or overexpressed human miRNAs have now been
miRNA duplex intermediate to be clearly deduced. In fact, shown to induce the cleavage of artificial mRNA substrates
this miRNA duplex is identical in structure to an siRNA bearing perfectly homologous target sites both in vitro and in
duplex, with the caveat that miRNA precursors frequently vivo (Hutvágner and Zamore, 2002; Zeng et al., 2003). Sim-
contain one or two mismatches in the stem, while siRNAs ilarly, artificial siRNAs have been shown to inhibit gene ex-
are generally perfectly double-stranded (Fig. 1). pression from mRNAs bearing imperfect targets without af-
The short half-life of the miRNA duplex intermediate fecting mRNA expression levels (Doench et al., 2003; Zeng
is apparently due largely to the fact that assembly of one et al., 2003). Together, these data demonstrate that siRNAs
strand of the duplex into RISC is rapid and efficient. It is and miRNAs are able to program RISC in a functionally in-
believed that a helicase component of RISC binds to one distinguishable manner (Fig. 1). On the other hand, it does
end of the duplex and unravels the double-strand, a process remain possible that mRNA cleavage and mRNA transla-
that requires ATP (Nykänen et al., 2001). The strand whose tion inhibition are mediated by distinct forms of RISC, per-
5 end is at the bound end of the duplex is then incorpo- haps containing different members of the Argonaut protein
rated into RISC. This process is non-random, in that RISC family. However, in that case these alternate forms must be
binding strongly favors the end of the miRNA duplex in- equivalently programmable by both siRNAs and miRNAs.
termediate that is less tightly helical (Schwarz et al., 2003).
Therefore, it is commonly observed that the 5 end of the
miRNA strand incorporated into RISC is poorly base paired 4. Strategies for expression of designed siRNAs
in the predicted duplex intermediate, while the 5 end of
the excluded strand forms a strong basepair, such as an G:C The miRNA processing pathway delineated in Fig. 1 is
(Lagos-Quintana et al., 2001; Schwarz et al., 2003). conserved in most, possibly all, higher eukaryotic cells and
While the composition of the RISC complex remains to be can be used to promote the entry of exogenously encoded
fully established, it is clear that RISC contains one or more or synthetic siRNAs into this highly effective posttranscrip-
members of the Argonaut family of proteins (Hammond tional pathway of gene inactivation. In many invertebrates,
et al., 2001; Carmell et al., 2002; Mourelatos et al., 2002). long dsRNAs are an effective means of inducing gene
Certain Argonaut proteins have been found to be essential specific RNAi. However, as noted above, long dsRNAs
for miRNA function in C. elegans and for siRNA func- activate the interferon response in vertebrate cells and are
tion in cultured human cells (Tabara et al., 1999; Doi et al., therefore not useful in these organisms. To overcome this
2003). However, their actual role, and whether different hu- problem, it is possible to program RISC by transfection of
man Argonaut proteins have different roles, remains to be cells with synthetic siRNA duplexes that closely mimic the
determined. miRNA duplex intermediate (Elbashir et al., 2001a). This
As noted above, the C. elegans let-7 and lin-4 miRNAs approach can result in the specific and effective destruction
inhibit the expression of target mRNAs after interacting with of target mRNAs (Fig. 1). However, synthetic siRNAs can
multiple imperfect target sites in the 3 UTR. Importantly, be quite expensive and RNA transfection is not efficient in
this does not result in a marked drop in the cytoplasmic all cell types of interest, particularly primary cells. More-
level of the target mRNA (Olsen and Ambros, 1999). Inhi- over, because only a finite amount of the siRNA duplex is
bition at the translational level has therefore been inferred, introduced into cells, inhibition is lost over a period of a
even though the polysome profile of the target mRNAs is few days due to dilution in the growing cell culture and/or
largely unaffected. While the mechanism underlying this in- degradation of the transfected siRNAs.
hibition remains unclear, it appears to be cooperative in that To overcome this problem, several groups have devel-
the binding of multiple RISC complexes is required (Doench oped plasmids that can express short hairpin RNAs that
et al., 2003). Unlike mRNA cleavage by RISC, which al- are structurally analogous to pre-miRNA hairpin interme-
lows turnover of RISC (Hutvágner and Zamore, 2002), this diates, using promoters dependent on RNA polymerase
translational inhibition also appears to require stoichiomet- III (Brummelkamp et al., 2002; Paddison et al., 2002; Sui
ric levels of RISC and hence of the relevant miRNA (Zeng et al., 2002). In a typical expression plasmid of this type,
et al., 2003). Translational inhibition is therefore less effec- the U6 or H1 promoter drives the expression of an RNA
B.R. Cullen / Virus Research 102 (2004) 3–9 7

that is predicted to fold into a short RNA hairpin, with the tissue-specific or developmentally-regulated promoters for
sequence of one side of the stem being complementary to expression of particular siRNAs in vivo. In contrast, the
the mRNA target sequence of interest. Transcription termi- level of expression of siRNAs transcribed from vectors that
nation is induced by a run of five thymidine residues, which depend on RNA polymerase III is not easily controlled, al-
causes termination of transcription by RNA polymerase III though this approach does permit the constitutive expression
after the second thymidine residue. This allows the short of high siRNA levels.
hairpin to acquire a 2 nt 3 overhang, exactly like authentic
pre-miRNAs, and recent evidence indicates that these short
hairpin RNAs are exported from the nucleus by Exp5, just 5. Conclusions
like an authentic pre-miRNA (Fig. 1) (Yi et al., 2003).
Subsequent work has refined the parameters for design Although RNAi plays a critical role in antiviral defense in
of short hairpin RNAs for maximal likelihood of successful plants, and is important in inhibiting transposon activation
RNAi. Thus, it is now clear that stems of from 25 to 29 bp in invertebrates (Plasterk, 2002), it remains unclear whether
in length give rise to better RNA processing and siRNA ex- RNAi serves similar functional roles in vertebrate species.
pression than do shorter stems (stems >29 bp in length can What is clear is that miRNAs are expressed in all higher
induce the interferon response). Because Dicer cleaves ∼21 eukaryotes and, at minimum, play a critical role in the post-
nt from the base of the short hairpin RNA, after binding to transcriptional regulation of genes during differentiation and
the end of this RNA structure, it is the more basal sequence development. The pathway that mediates miRNA process-
of the stem that gives rise to the siRNA duplex intermedi- ing and function is becoming increasingly well understood
ate (Lee et al., 2003a). To promote incorporation of the an- and it is now apparent that this pathway can support several
tisense strand into RISC, it is important to ensure that the different strategies for the introduction of functional, exoge-
projected 5 end of the antisense strand is poorly basepaired nously produced siRNAs (Fig. 1). While it remains uncertain
(Schwarz et al., 2003). Even in the absence of these modifi- whether RNAi plays any role in controlling virus infections
cations, short hairpin RNA expression plasmids have proven in animals, there is no question that the artificial induction
to be quite effective in inducing RNAi. However, these plas- of RNAi can be used to target viral transcripts, or host
mids again have practical problems in terms of the ability genes that produce co-factors critical for virus replication,
to effectively transfect cells of interest and in terms of the and thereby effectively block virus replication in culture.
transience of the inhibitory effect observed. Whether RNAi will lead to effective antiviral treatments in
To overcome these problems, several groups have devel- the future remains to be seen. What is clear, however, is that
oped viral vectors, based on murine leukemia virus or hu- RNAi is an extremely powerful tool to identify and char-
man immunodeficiency virus that incorporate short hairpin acterize cellular factors that are required for effective virus
RNA expression cassettes (Barton and Medzhitov, 2002; Lee replication. In doing so, RNAi has the potential to identify
et al., 2003a; Qin et al., 2003; Stewart et al., 2003). These targets for chemotherapeutic intervention that would be
vectors can be used to efficiently infect target cells in culture very difficult to define using more conventional approaches.
or in vivo and have been shown to give rise to long term ex-
pression of the encoded siRNA and, hence, to the long term
suppression of the target mRNA of interest. Most recently, References
lentivirus-based short hairpin RNA expression vectors have
been used to effectively and stably suppress the expression Barton, G.M., Medzhitov, R., 2002. Retroviral delivery of small interfering
of genes in transgenic mice (Rubinson et al., 2003; Tiscornia RNA into primary cells. Proc. Natl. Acad. Sci. U.S.A. 99, 14943–
et al., 2003), thus hopefully heralding a new era of simple 14945.
reverse genetic experiments in experimental animals. Bernstein, E., Caudy, A.A., Hammond, S.M., Hannon, G.J., 2001a. Role
for bidentate ribonuclease in the initiation step of RNA interference.
A final approach to the expression of siRNAs uses as its
Nature 409, 363–366.
entry point the first step in the expression pathway for en- Bernstein, E., Denli, A.M., Hannon, G.J., 2001b. The rest is silence. RNA
dogenous miRNAs (Fig. 1). Because pri-miRNAs contain 7, 1509–1521.
all the sequences required for miRNA processing in cis, it Brennecke, J., Hipfner, D.R., Stark, A., Russell, R.B., Cohen, S.M.,
should in principle be possible to switch the part of the 2003. Bantam encodes a developmentally regulated microRNA that
controls cell proliferation and regulates the proapoptotic gene hid in
miRNA gene that encodes the mature miRNA, while leav-
Drosophila. Cell 113, 25–36.
ing the processing signals intact, and hence use the entire Brummelkamp, T.R., Bernards, R., Agami, R., 2002. A system for stable
miRNA processing machinery to produce an siRNA, or ar- expression of short interfering RNAs in mammalian cells. Science
tificial miRNA, of interest. In fact, using the miR-30 gene 296, 550–553.
as a starting point, this has indeed been achieved using sev- Carmell, M.A., Xuan, Z., Zhang, M.Q., Hannon, G.J., 2002. The argonaute
family: tentacles that reach into RNAi, developmental control, stem
eral different siRNA sequences (Zeng et al., 2002; Zeng and
cell maintenance, and tumorigenesis. Genes Dev. 16, 2733–2742.
Cullen, 2003). A major advantage of this approach to siRNA Cullen, B.R., 2003. Nuclear RNA export. J. Cell Sci. 116, 587–597.
expression is that it relies on transcription via RNA poly- Doench, J.G., Petersen, C.P., Sharp, P.A., 2003. siRNAs can function as
merase II and thus has the potential to permit the use of miRNAs. Genes Dev. 17, 438–442.
8 B.R. Cullen / Virus Research 102 (2004) 3–9

Doi, N., Zenno, S., Ueda, R., Ohki-Hamazaki, H., Ui-Tei, K., Saigo, K., Llave, C., Xie, Z., Kasschau, K.D., Carrington, J.C., 2002. Cleavage
2003. Short-interfering-RNA-mediated gene silencing in mammalian of scarecrow-like mRNA targets directed by a class of Arabidopsis
cells requires Dicer and eIF2C translation initiation factors. Curr. Biol. miRNA. Science 297, 2053–2056.
13, 41–46. Manche, L., Green, S.R., Schmedt, C., Mathews, M.B., 1992. Interactions
Dykxhoorn, D.M., Novina, C.D., Sharp, P.A., 2003. Killing the messenger: between double-stranded RNA regulators and the protein kinase DAI.
short RNAs that silence gene expression. Nat. Rev. 4, 457–467. Mol. Cell. Biol. 12, 5238–5248.
Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl, Martinez, J., Patkaniowska, A., Urlaub, H., Lührmann, R., Tuschl, T.,
T., 2001a. Duplexes of 21-nucleotide RNAs mediate RNA interference 2002. Single-stranded antisense siRNAs guide target RNA cleavage
in cultured mammalian cells. Nature 411, 494–498. in RNAi. Cell 110, 563–574.
Elbashir, S.M., Lendeckel, W., Tuschl, T., 2001b. RNA interference is Mourelatos, Z., Dostie, J., Paushkin, S., Sharma, A., Charroux, B., Abel,
mediated by 21- and 22-nucleotide RNAs. Genes Dev. 15, 188–200. L., Rappsilber, J., Mann, M., Dreyfuss, G., 2002. miRNPs: a novel
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello, class of ribonucleoproteins containing numerous microRNAs. Genes
C.C., 1998. Potent and specific genetic interference by double-stranded Dev. 16, 720–728.
RNA in Caenorhabditis elegans. Nature 391, 806–811. Nykänen, A., Haley, B., Zamore, P.D., 2001. ATP requirements and small
Grishok, A., Pasquinelli, A.E., Conte, D., Li, N., Parrish, S., Ha, I., Baillie, interfering RNA structure in the RNA interference pathway. Cell 107,
D.L., Fire, A., Ruvkun, G., Mello, C.C., 2001. Genes and mechanisms 309–321.
related to RNA interference regulate expression of the small temporal Olsen, P.H., Ambros, V., 1999. The lin-4 regulatory RNA controls de-
RNAs that control C. elegans developmental timing. Cell 106, 23–34. velopmental timing in Caenorhabditis elegans by blocking LIN-14
Gwizdek, C., Ossarch-Nazari, B., Brownawell, A.M., Doglio, A., Bertrand, protein synthesis after the initiation of translation. Dev. Biol. 216,
E., Macara, I.G., Dargemont, C., 2003. Exportin-5 mediates nuclear 671–680.
export of minihelix-containing RNAs. J. Biol. Chem. 278, 5505– Paddison, P.J., Caudy, A.A., Bernstein, E., Hannon, G.J., Conklin, D.S.,
5508. 2002. Short hairpin RNAs (shRNAs) induce sequence-specific silenc-
Hamilton, A.J., Baulcombe, D.C., 1999. A species of small antisense RNA ing in mammalian cells. Genes Dev. 16, 948–958.
in posttranscriptional gene silencing in plants. Science 286, 950–952. Park, W., Li, J., Song, R., Messing, J., Chen, X., 2002. Carpel factory,
Hammond, S.M., Bernstein, E., Beach, D., Hannon, G.J., 2000. An a Dicer homolog, and HEN1, a novel protein, act in microRNA
RNA-directed nuclease mediates post-transcriptional gene silencing in metabolism in Arabidopsis thaliana. Curr. Biol. 12, 1484–1495.
Drosophila cells. Nature 404, 293–295. Plasterk, R.H.A., 2002. RNA silencing: the genome’s immune system.
Hammond, S.M., Boettcher, S., Caudy, A.A., Kobayashi, R., Hannon, G.J., Science 296, 1263–1265.
2001. Argonaute 2, a link between genetic and biochemical analyses Qin, X.-F., An, D.S., Chen, I.S.Y., Baltimore, D., 2003. Inhibiting HIV-1
of RNAi. Science 293, 1146–1150. infection in human T cells by lentiviral-mediated delivery of small
Hutvágner, G., McLachlan, J., Pasquinelli, A.E., Bálint, É., Tuschl, T., interfering RNA against CCR5. Proc. Natl. Acad. Sci. U.S.A. 100,
Zamore, P.D., 2001. A cellular function for the RNA-interference 183–188.
enzyme Dicer in the maturation of the let-7 small temporal RNA. Reinhart, B.J., Slack, F.J., Basson, M., Pasquinelli, A.E., Bettinger, J.C.,
Science 293, 834–838. Rougvie, A.E., Horvitz, H.R., Ruvkun, G., 2000. The 21-nucleotide
Hutvágner, G., Zamore, P.D., 2002. A microRNA in a multiple-turnover let-7 RNA regulates developmental timing in Caenorhabditis elegans.
RNAi enzyme complex. Science 297, 2056–2060. Nature 403, 901–906.
Kawasaki, H., Taira, K., 2003. Hes1 is a target of microRNA-23 during Rubinson, D.A., Dillon, C.P., Kwiatkowski, A.V., Sievers, C., Yang, L.,
retinoic-acid-induced neuronal differentiation of NT2 cells. Nature Kopinja, J., Zhang, M., McManus, M.T., Gertler, F.B., Scott, M.L.,
423, 838–842. Van Parijs, L., 2003. A lentivirus-based system to functionally silence
Knight, S.W., Bass, B.L., 2001. A role for the RNase III enzyme DCR-1 genes in primary mammalian cells, stem cells and transgenic mice by
in RNA interference and germ line development in Caenorhabditis RNA interference. Nat. Gen. 33, 401–406.
elegans. Science 293, 2269–2271. Schwarz, D.S., Hutvágner, G., Du, T., Xu, Z., Aronin, N., Zamore, P.D.,
Lagos-Quintana, M., Rauhut, R., Lendeckel, W., Tuschl, T., 2001. Iden- 2003. Asymmetry in the assembly of the RNAi enzyme complex. Cell
tification of novel genes coding for small expressed RNAs. Science 115, 199–208.
294, 853–858. Sen, G.C., 2001. Viruses and interferons. Annu. Rev. Microbiol. 55, 255–
Lagos-Quintana, M., Rauhut, R., Yalcin, A., Meyer, J., Lendeckel, W., 281.
Tuschl, T., 2002. Identification of tissue-specific microRNAs from Stewart, S.A., Dykxhoorn, D.M., Palliser, D., Mizuno, H., Yu, E.Y., An,
mouse. Curr. Biol. 12, 735–739. D.S., Sabatini, D.M., Chen, I.S.Y., Hahn, W.C., Sharp, P.A., Weinberg,
Lau, N.C., Lim, L.P., Weinstein, E.G., Bartel, D.P., 2001. An abundant R.A., Novina, C.D., 2003. Lentivirus-delivered stable gene silencing
class of tiny RNAs with probable regulatory roles in Caenorhabditis by RNAi in primary cells. RNA 9, 493–501.
elegans. Science 294, 858–862. Sui, G., Soohoo, C., Affar, E.B., Gay, F., Shi, Y., Forrester, W.C., Shi,
Lee, M.-T., Coburn, G.A., McClure, M.O., Cullen, B.R., 2003a. Inhi- Y., 2002. A DNA vector-based RNAi technology to suppress gene
bition of HIV-1 replication in primary macrophages using Tat or expression in mammalian cells. Proc. Natl. Acad. Sci. U.S.A. 99,
CCR5-specific small interfering RNAs expressed from a lentiviral 5515–5520.
vector. J. Virol. 77, 11964–11972. Tabara, H., Sarkissian, M., Kelly, W.G., Fleenor, J., Grishok, A., Timmons,
Lee, R.C., Ambros, V., 2001. An extensive class of small RNAs in L., Fire, A., Mello, C.C., 1999. The rde-1 gene, RNA interference,
Caenorhabditis elegans. Science 294, 862–864. and transposon silencing in C. elegans. Cell 99, 123–132.
Lee, R.C., Feinbaum, R.L., Ambros, V., 1993. The C. elegans hete- Tiscornia, G., Singer, O., Ikawa, M., Verma, I.M., 2003. A general method
rochronic gene lin-4 encodes small RNAs with antisense complemen- for gene knockdown in mice by using lentiviral vectors expressing
tarity to lin-14. Cell 75, 843–854. small interfering RNA. Proc. Natl. Acad. Sci. U.S.A. 100, 1844–1848.
Lee, Y., Ahn, C., Han, J., Choi, H., Provost, P., Kim, S., Kim, V.N., Xu, P., Vernooy, S.Y., Guo, M., Hay, B.A., 2003. The Drosophila mi-
2003b. The nuclear RNase III drosha initiates microRNA processing. croRNA Mir-14 suppresses cell death and is required for normal fat
Nature 425, 415–419. metabolism. Curr. Biol. 13, 790–795.
Lee, Y., Jeon, K., Lee, J.T., Kim, S., Kim, V.N., 2002. MicroRNA matu- Yi, R., Qin, Y., Macara, I.G., Cullen, B.R., 2003. Exportin-5 Mediates
ration: stepwise processing and subcellular localization. EMBO J. 21, the nuclear export of pre-microRNAs and short hairpin RNAs. Genes
4663–4670. Dev. 17, 3011–3016.
B.R. Cullen / Virus Research 102 (2004) 3–9 9

Zeng, Y., Cullen, B.R., 2003. Sequence requirements for micro Zeng, Y., Yi, R., Cullen, B.R., 2003. MicroRNAs and small interfering
RNA processing and function in human cells. RNA 9, 112– RNAs can inhibit mRNA expression by similar mechanisms. Proc.
123. Natl. Acad. Sci. U.S.A. 100, 5779–5784.
Zeng, Y., Wagner, E.J., Cullen, B.R., 2002. Both natural and designed Zhang, H., Kolb, F.A., Brondani, V., Filipowicz, W., 2002. Human Dicer
micro RNAs can inhibit the expression of cognate mRNAs when preferentially cleaves dsRNAs at their termini without a requirement
expressed in human cells. Mol. Cell 9, 1327–1333. for ATP. EMBO J. 21, 5875–5885.
Virus Research 102 (2004) 11–17

RNA silencing in viral infections: insights from poliovirus


Maria-Carla Saleh1 , Ronald P. Van Rij1 , Raul Andino∗
Department of Microbiology and Immunology, University of California, 513 Parnassus Avenue, Box 0414, San Francisco, CA 94143-2280, USA

Abstract

RNA interference or RNA silencing is a dsRNA guided mechanism that mediates sequence specific degradation of RNA. The recent
demonstration that RNA interference can be used to inhibit virus replication has initiated an exciting field of research: first, as a potential
novel antiviral therapeutic approach and, second, as a tool for dissecting virus–host interactions. Here we review and discuss the current data
and perspectives on the use of RNA interference in the study of poliovirus as a model for positive strand RNA viruses.
© 2004 Elsevier B.V. All rights reserved.

Keywords: RNA interference; RNA silencing; siRNA; Poliovirus; Picornaviridae

1. Introduction Because of the extreme specificity and efficiency of the


RNAi machinery, this mechanism has the ability to clear
Clearance of viral pathogens from a variety of hosts is of- plant and mammalian cells from viral infection (Gitlin and
ten the result of the destruction of infected cells. However, Andino, 2003; Randall et al., 2003; Waterhouse et al., 2001).
mechanisms that enable cell survival following infection
have been described (Guidotti and Chisari, 2001). Studies
in plants suggest that one of such mechanisms may be 2. Multiple functions of RNAi
based on RNA-induced gene silencing, now referred to as
RNA silencing or RNA interference (RNAi). This ancient The RNAi process is very efficient: a few dsRNA
pathway is conserved among various species from different molecules can trigger inactivation of a continuously tran-
kingdoms (fungi, animals and plants) and it seems to control scribed target mRNA for prolonged periods of time (Ruvkun,
gene expression at transcriptional and post-transcriptional 2001). The RNAi-induced inactivation persists through cell
levels (Bernstein et al., 2001b; Hammond et al., 2001b). division and in some organisms can spread to untreated cells
The post-transcriptional activity of the RNAi machinery and tissues; when the RNAi spreads into germ line cells it
to degrade cytoplasmic RNA in a sequence specific manner can even be inherited by subsequent generations (Hammond
is key to its antiviral function. Biochemical studies indicate et al., 2001b). Although amplification and spreading of
that RNAi proceeds via a two-step mechanism. In the first RNA silencing have been demonstrated in plants, nema-
step, long dsRNAs are produced locally or taken up by todes and in vitro in Drosophila extracts (Fire et al., 1998;
the cells and cleaved by the RNase III-like nuclease Dicer Lipardi et al., 2001; Lucas et al., 2001; Roignant et al.,
(Bernstein et al., 2001a), which generates 21–23 nucleotide 2003) they have not been observed in mammalian systems
duplex RNAs, called small interfering RNAs (siRNA). In the (Chi et al., 2003; Stein et al., 2003).
second step, siRNAs are incorporated into a multicompo- A role for RNAi pathways in the regulation of endoge-
nent nuclease complex, the RNA-induced silencing complex nous genes was suggested through the analysis of plants
(RISC) (Hammond et al., 2001a). The antisense strand of the and animals containing dysfunctional RNAi components.
duplex serves as a guide that directs RISC to recognize and For example, mutations in a component of RISC cause
cleave cognate mRNAs (Martinez et al., 2002a) (Fig. 1b). pleiotropic developmental abnormalities in Arabidopsis
(Bohmert et al., 1998). A mutation in the Arabidopsis Dicer
∗ Corresponding author. Tel.: +1-415-502-6358;
orthologue causes defects in leaf development and overpro-
fax: +1-415-476-0939.
liferation of floral meristems (Jacobsen et al., 1999). Mu-
E-mail address: andino@itsa.ucsf.edu (R. Andino). tations in Argonaute family members in Drosophila impact
1 Author contributed equally to this review. normal development with drastic effects in neuronal devel-

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.010
12 M.-C. Saleh et al. / Virus Research 102 (2004) 11–17

(a) (b)
Substrate
for Dicer? dsRNA
Entry P1 P2 P3 AAAA

Dicer

Translation 5' 3'


siRNA

Target after siRNA Target RNA


Negative treatment?
strand 5' 3' (+)
synthesis RISC
3Dpol 5' (-)

(+) Degradation
(+) (+) of target RNA
Positive
strand 3' 5' (-)
synthesis

Packaging

Fig. 1. (a) Overview of poliovirus life cycle. After receptor mediated entry and uncoating of the virus, the viral RNA is translated in a cap-independent
fashion by the host cell machinery. The viral RNA is used as a template for negative strand synthesis by the virus encoded 3D polymerase. The negative
strand is subsequently used as a template for the production of positive viral RNA strands, which are packaged into new virions. (b) Overview of RNAi
pathway. The central component is dsRNA which is converted by Dicer into siRNA, small dsRNA of ∼21 nucleotides length with a two nucleotide
overhang at the 3 end. One strand is incorporated in the RISC complex and guides recognition and cleavage of the target RNA. Open questions with
respect to RNA silencing of positive strand RNA viruses include whether dsRNA intermediates produced during virus replication are a target for the
RISC complex, whether viruses encode proteins that inhibit the RNAi pathway and which of the RNA strands are best targeted after introduction of
synthetic siRNAs.

opment (Kataoka et al., 2001), and mutations in another with the RNA silencing machinery in Drosophila cells (Li
RISC component, piwi, result in defects in both germline et al., 2002). Furthermore, many C. elegans mutants defi-
stem-cell proliferation and maintenance (Cox et al., 1998). cient in RNAi show transposon mobilization with obvious
The emerging view is that RNA silencing is part of a so- implications for genome stability (Ketting et al., 1999). A
phisticated network of interconnected pathways for cellular key question that remains unanswered is whether RNAi has
defense (pathogen resistance and stabilization of mobile ge- a role as a natural antiviral defense in mammals. Five criti-
netic elements), RNA surveillance (chromatin remodeling, cal issues have recently been proposed to establish whether
genome organization and stability) and development. In ad- RNAi plays a role in natural defense against viruses (Gitlin
dition, RNAi may become a powerful tool to manipulate and Andino, 2003):
gene expression experimentally. Here we review and discuss
the current data and perspectives on the use of RNAi in the (1) Generation of siRNA during the course of a natural in-
study of positive strand RNA viruses. Indeed RNAi opens fection.
up exciting possibilities for virus research: first, as a poten- (2) Up-regulation of RNAi machinery components during
tial novel antiviral therapeutic approach and, second, as a viral infection.
tool for dissecting virus–host interactions. (3) Viral mechanisms to suppress or escape RNAi.
(4) Increased susceptibility to viral infection after mutation
or deletion of RNAi components.
(5) Local or systemic spread of an RNAi-based antiviral
3. Antiviral function of RNAi response.

There is a good deal of genetic support for the importance Even though synthetic siRNA transfected into mammalian
of RNAi in genome defense. In plants, RNAi is clearly cells can inhibit positive strand virus replication (Gitlin and
involved in the response to viruses: Arabidopsis strains Andino, 2003; Kapadia et al., 2003; Randall et al., 2003;
defective in post transcriptional gene silencing are more Seo et al., 2003; Wilson et al., 2003), it is not clear whether
susceptible to virus infections (Mourrain et al., 2000), and a RNA silencing can be elicited naturally during these viral in-
substantial number of plant viruses encode proteins that fections. Although the replication intermediates may contain
counter silencing (Brigneti et al., 1998; Voinnet, 2001; long stretches of dsRNA, especially during negative strand
Voinnet et al., 1999). RNA silencing also appears to con- synthesis, it is not know whether they can be targeted for
tribute to antiviral defense in invertebrates as it was shown processing by Dicer. Localization of dsRNA intermediates in
by studies of flock house virus (FHV) and its interaction membranous and/or proteinous replication complexes may
M.-C. Saleh et al. / Virus Research 102 (2004) 11–17 13

shield these intermediates from the RNAi machinery. An- der control. More recently, thanks to a global effort to erad-
swers to these and other related issues have not been obtained icate poliovirus, the disease has been eliminated from most
yet, but an exciting field of research has been opened up. of the world, and only seven countries worldwide remain
polio-endemic (http://www.polioeradication.org). However,
as we face the final stages of the Poliovirus Global Erad-
4. Poliovirus as a model system to study RNAi ication Initiative it is important to develop effective anti-
poliovirus therapies.
Until the 1950s, poliovirus (PV), the etiologic agent of Poliovirus has served as an excellent model to study
poliomyelitis, crippled thousands of children every year. the molecular and cellular events that lead to viral replica-
Poliovirus is able to invade the nervous system, and can tion. As a consequence, much is known about its biology,
cause total paralysis in a matter of hours. The virus enters life cycle and interactions with the host. Poliovirus is a
the body through the mouth and multiplies in the ton- positive-stranded RNA virus, member of the family Picor-
sils and the Peyer’s patches of the small intestine before naviridae. The genome comprises a long open reading frame
spreading to other sites by a viraemia. Initial symptoms encoding a single polyprotein of more than 2000 amino
are fever, fatigue, headache, vomiting, and stiffness in the acids, which is processed into functional proteins by vi-
neck and pain in the limbs. One in 200 infections leads rally encoded proteases. The poliovirus polyprotein can be
to irreversible paralysis (usually in the legs). Among those divided into three major parts (P1–P3). The P1 region con-
paralyzed, 5–10% die when their breathing muscles become tains the four structural proteins (VP1–VP4) that constitute
immobilized (Minor, 1997). the capsid. Proteins derived from the P2 and the P3 region
After the introduction of effective vaccines in the late are involved in viral replication. After receptor-mediated
1950s (inactivated polio vaccine (IPV)) and early 1960s entry, the viral genome is directly translated by the cellular
(oral polio vaccine (OPV)), poliomyelitis was brought un- translation machinery. The same viral RNA is then used as

Fig. 2. Poliovirus replication can be suppressed by siRNA. (a) Pretreatment of HeLa cells with siRNA directed against the capsid region (siC) is able
to prevent plaque formation. Suppression of replication is not observed after treatment of cells with single stranded RNA (ssC(+) and ssC(−)), an
unrelated siRNA (directed against luciferase (siL)), or dsDNA of identical sequence of siC (C-DNA). (b) Western blot analysis with anti 3Dpol antibody
of poliovirus infected cells. siP indicates siRNA directed against the poliovirus polymerase region. (c) Northern blot analysis of poliovirus infected cells
at 6 and 8 h after infection. Reprinted from Gitlin et al. (2002) with permission.
14 M.-C. Saleh et al. / Virus Research 102 (2004) 11–17

template for negative strand RNA synthesis that results in studies, in which injection of siRNA or plasmids encoding
the formation of a double-stranded RNA replicative form small hairpin RNAs (shRNAs) directed against luciferase,
(RF). The negative strand RNA can in turn serve as a GFP, or a secreted form of human placental alkaline phos-
template for synthesis of new positive strand RNA. Repli- phatase (SEAP) could inhibit expression of these genes from
cation occurs in replication complexes tightly associated plasmids after high pressure tail injection (hydrodynamic
with smooth membrane vesicles, which accumulate in the transfection) (Lewis et al., 2002; McCaffrey et al., 2002).
cytoplasm during viral infection (Fig. 1a) (Rueckert, 1996). Silencing of endogenous genes was shown by downregu-
Can RNAi suppress replication of this highly lytic virus? lation of GFP in a transgenic mouse (Lewis et al., 2002).
In a recent study, Gitlin et al. (2002) used siRNA directed The first application of RNAi technology to prevent disease
against the poliovirus genome to show that RNAi can was illustrated by protection of mice against antibody- or
effectively prevent viral replication in mammalian cells concanavalin A-induced hepatitis by using siRNA directed
(Fig. 2). Pretreatment of human and murine cells with against fas protein (Song et al., 2003). Recently, shRNAs
double-stranded siRNA to the poliovirus genome (capsid against HBV mRNAs were shown to be able to suppress
and 3D polymerase) prevented plaque formation and re- virus replication from co-injected HBV expression plasmids
sulted in a 100-fold reduction of the progeny titer in one-step in hepatocytes (McCaffrey et al., 2003), providing proof of
growth curves. In these experiments, siRNAs promote principle for the use of RNAi-based antiviral strategies in
clearance of the virus from most infected cells. The RNAi animal models.
antiviral effect, for which the mechanistic detail remains to Despite these promising results, significant hurdles exist
be elucidated, is sequence-specific and not attributable to in the successful application of RNAi-based antiviral strate-
either classical antisense mechanism or interferon response gies in a clinical setting. Several of these issues may be ad-
effectors PKR and RNaseL. dressed using poliovirus as a model virus.
This study yielded insights into the accessibility of the First, as shown in Gitlin et al. (2002), a limited number
viral genome to RNAi, and the ability of virus to escape of mismatches between siRNA and target RNA may already
this defense mechanism. Several siRNAs targeting regions abolish the suppressive action of the siRNA (Elbashir et al.,
in capsid and polymerase that are well conserved among 2001; Martinez et al., 2002b). The error-prone nature of the
enteroviruses were effective in inhibiting viral replication. virally encoded RNA-dependent RNA polymerase will gen-
In contrast, siRNA that targeted the 5 non-coding region erate a spectrum of mutants in each replication cycle, and
(5 UTR), also highly conserved among all picornaviruses, it can be expected that escape mutants will rapidly arise
failed to reduce virus replication (L. Gitlin and R. Andino, under incomplete suppression by siRNA. Indeed, point mu-
unpublished observations). This result implies that some re- tations arose in the center of the target region of the po-
gions of the 5 UTR are less accessible to the RNAi machin- liovirus genome, when infections were performed at high
ery. Indeed, secondary and tertiary structure of the RNA, as MOI (Gitlin et al., 2002). Although single mismatches have
well as proteins binding to RNA elements of the 5 UTR, been shown to render a siRNA ineffective (Elbashir et al.,
could potentially shield the viral target RNA from the RNAi 2001), they may be tolerated by the RNAi machinery to
machinery. Since siRNAs directed against the hepatitis C some extent (Jacque et al., 2002). Simultaneously targeting
virus IRES have been recently shown to effectively inhibit of multiple regions or of regions that constitute indispens-
replication of HCV replicons (Yokota et al., 2003), there able RNA structures for virus replication and thus do not
may be differences in the accessibility of different regula- allow viral escape, might be approaches to circumvent vi-
tory regions of different viral genomes. Notably, viruses can ral escape, that can readily be explored using the poliovirus
eventually become resistant to synthetic short RNAs such model system. Alternatively, targeting essential cellular fac-
as those described by Gitlin et al. (2002). Escape mutants tors, such as CC-chemokine receptor 5 (CCR5) and CD4 in
emerged from cells treated with single siRNAs after po- the case of HIV-1 (Novina et al., 2002; Qin et al., 2003) or
liovirus infection at a high multiplicity of infection. These co-administration with other antiviral drugs may limit the
escape mutants presented subtle nucleotide changes in the possibility for viral escape.
siRNA target sequences. It appears that a single mismatch A second aspect requiring further investigation is that
located approximately in the center of the siRNA nearly not all siRNAs are equally effective in the suppression of a
abolishes silencing of poliovirus (Gitlin and Andino, unpub- target RNA (Harborth et al., 2001). The sequence require-
lished observations). ments for effective RNAi-based suppression have not been
well defined and efficient siRNAs need to be developed by
trial and error. It is likely that optimal siRNA sequences
5. Therapeutic potential of RNAi as an antiviral agent are determined by the interaction of the siRNAs with both
multicomponent nuclease RISC and with the target RNA
The susceptibility of viruses to RNAi in tissue culture sequence. Other open questions that apply specifically
suggests that RNAi-based antiviral strategies might be fea- to viral genomes as targets for RNAi, are whether there
sible also in entire mammals. Evidence that RNAi can be is a differential susceptibility to RNAi between coding
induced in vivo has been obtained in a number of mouse and non-coding regions, and whether protein bound RNA
M.-C. Saleh et al. / Virus Research 102 (2004) 11–17 15

structures are less accessible to the RNAi machinery. If be a powerful approach. Traditional knockout strategies are
conserved secondary structures in the untranslated regions labor intensive and may result in cellular toxicity, as can be
of the virus are indeed susceptible, these might be the re- expected for proteins that play important roles in basic bio-
gions of choice to develop siRNAs as mutations in these chemical processes in the host cell. For example, the poly
structures would lead to loss of function. A binding protein has been implicated in positive-stranded
Another aspect that is not yet properly defined is whether RNA virus replication, such as poliovirus (Herold and
it is possible to block viral replication once initiated. Effec- Andino, 2001; Svitkin et al., 2001). However, the protein
tive inhibition of poliovirus was observed when cells were is also involved in translation initiation and is essential
transfected with siRNA before infection (Gitlin et al., 2002). for cell growth in Saccharomyces cerevisiae (Sachs et al.,
Although in theory siRNA could target newly synthesized 1987). Transient, inducible and non-complete knock-down
RNA strands of either positive and negative orientation, by RNAi may not interfere with cell survival, but still
replication of poliovirus on membranous vesicles and cou- provide a window of opportunity to study the role of cel-
pling of RNA synthesis with virion assembly might shield lular factors in virus replication in the cell. Furthermore,
replicative intermediates from degradation by the RNAi ma- complementation by an expression plasmid that encodes
chinery. the protein of interest in a mutated form could provide
Virally encoded suppressors of the RNAi pathway have additional information about sites of interaction.
been described in a variety of plant viruses (Brigneti et al., For poliovirus, host factors involved in cap-independent
1998; Voinnet et al., 1999) and in FHV, which naturally in- translation initiation and negative strand RNA synthesis have
fects vertebrate and invertebrate hosts (Li et al., 2002). Thus begun to be defined through in vitro studies. Factors that have
far, similar suppressors have not been reported in mam- been implied in these processes include canonical translation
malian viruses. The presence of putative suppressors in spe- initiation factors, poly A binding protein, poly(rC) binding
cific mammalian virus families would limit the potential of proteins 1 and 2, La antigen, and poly-pyrimidine tract bind-
RNAi-based therapeutic strategies. ing proteins (reviewed in Andino et al., 1999; Belsham and
Finally, recent RNAi studies in animals rely heavily on Sonenberg, 2000; Semler and Wimmer, 2002). Additional
the efficient uptake and expression of siRNA in hepatocytes interactions with host factors include the interaction with the
(Liu et al., 1999; Song et al., 2003; Zhang et al., 1999). poliovirus receptor (CD155) for viral entry (Ren et al., 1990)
Other methods to deliver shRNA by adenovirus-mediated and possible interactions with proteins of the COPII system
delivery or lentiviral vectors have been successful in vivo leading to alterations in the secretory pathway to establish
as well (Tiscornia et al., 2003; Xia et al., 2002). However, membranous vesicles which function as sites for virus repli-
as viruses differ greatly in tissue and cell tropism, specific cation (Rust et al., 2001). It is likely that in the near future
targeting of siRNA expression systems to specific tissues approaches that employ downregulation of these factors us-
will be crucial for the success of RNAi-based therapeutic ing RNAi will yield insights in their specific function during
approaches. These methods may therefore encounter similar viral replication.
problems and limitations as gene therapy (Thomas et al.,
2003).
7. Conclusion

6. RNA silencing as a tool to study virus replication Much information about the mechanism and the use of
and pathogenesis RNAi as a tool to manipulate gene expression has become
available in recent years. A better understanding of this
RNA interference has been developed into a powerful novel defense mechanism may result in effective therapeutic
technique to manipulate gene expression experimentally, approaches for intractable viral diseases that are currently
which may benefit the study of virus replication. For afflicting humans. However, although poliovirus (and other
positive-stranded, nonsegmented viruses, such as poliovirus, viruses) can be targeted by artificially introduced siRNA
any siRNAs directed against its genome would result in in vitro, still much research needs to be done to establish
down regulation of the entire polyprotein. In contrast, for the role of RNAi in natural virus infection, and several
negative-stranded and segmented RNA viruses, such as critical issues need to be addressed, before RNAi-based
paramyxoviruses and orthomyxoviruses, it is possible to technology might be used for the treatment of viral
downregulate specific viral genes using RNAi. This pro- diseases.
vides the basis for a novel approach in which individual
viral proteins can be downregulated with RNAi to be re-
placed with modified proteins provided in trans (e.g., from Acknowledgements
plasmid vectors).
Viruses depend on host factors at every stage of their This work was financially supported by an EMBO long
replication cycle. To establish the role of putative host fac- term fellowship to RvR and by NIH grant AI40085 to R.A.
tors in virus replication, RNAi-based downregulation may We would like to thank L. Gitlin for helpful discussions.
16 M.-C. Saleh et al. / Virus Research 102 (2004) 11–17

References Li, H.W., Li, W.X., Ding, S.W., 2002. Induction and suppression of RNA
silencing by an animal virus. Science 296, 1319–1321.
Lipardi, C., Wei, Q., Paterson, B.M., 2001. RNAi as random degradative
Andino, R., Boddeker, N., Silvera, D., Gamarnik, A.V., 1999. Intracellular
PCR: siRNA primers convert mRNA into dsRNAs that are degraded
determinants of picornavirus replication. Trends Microbiol. 7, 76–82.
to generate new siRNAs. Cell 107, 297–307.
Belsham, G.J., Sonenberg, N., 2000. Picornavirus RNA translation: roles
Liu, F., Song, Y., Liu, D., 1999. Hydrodynamics-based transfection in
for cellular proteins. Trends Microbiol. 8, 330–335.
animals by systemic administration of plasmid DNA. Gene Ther. 6,
Bernstein, E., Caudy, A.A., Hammond, S.M., Hannon, G.J., 2001a. Role
1258–1266.
for a bidentate ribonuclease in the initiation step of RNA interference.
Lucas, W.J., Yoo, B.C., Kragler, F., 2001. RNA as a long-distance infor-
Nature 409, 363–366.
mation macromolecule in plants. Nat. Rev. Mol. Cell Biol. 2, 849–857.
Bernstein, E., Denli, A.M., Hannon, G.J., 2001b. The rest is silence. RNA
Martinez, J., Patkaniowska, A., Urlaub, H., Luhrmann, R., Tuschl, T.,
7, 1509–1521.
2002a. Single-stranded sntisense siRNAs guide target RNA cleavage
Bohmert, K., Camus, I., Bellini, C., Bouchez, D., Caboche, M., Benning,
in RNAi. Cell 110, 563–574.
C., 1998. AGO1 defines a novel locus of Arabidopsis controlling leaf
Martinez, L.A., Naguibneva, I., Lehrmann, H., Vervisch, A., Tchenio,
development. EMBO J. 17, 170–180.
T., Lozano, G., Harel-Bellan, A., 2002b. Synthetic small inhibiting
Brigneti, G., Voinnet, O., Li, W.X., Ji, L.H., Ding, S.W., Baulcombe, D.C., RNAs: efficient tools to inactivate oncogenic mutations and restore
1998. Viral pathogenicity determinants are suppressors of transgene p53 pathways. Proc. Natl. Acad. Sci. U.S.A. 99, 14849–14854.
silencing in Nicotiana benthamiana. EMBO J. 17, 6739–6746.
McCaffrey, A.P., Meuse, L., Pham, T.T., Conklin, D.S., Hannon, G.J.,
Chi, J.T., Chang, H.Y., Wang, N.N., Chang, D.S., Dunphy, N., Brown, Kay, M.A., 2002. RNA interference in adult mice. Nature 418, 38–39.
P.O., 2003. Genomewide view of gene silencing by small interfering McCaffrey, A.P., Nakai, H., Pandey, K., Huang, Z., Salazar, F.H., Xu, H.,
RNAs. Proc. Natl. Acad. Sci. U.S.A. 100, 6343–6346. Wieland, S.F., Marion, P.L., Kay, M.A., 2003. Inhibition of hepatitis
Cox, D.N., Chao, A., Baker, J., Chang, L., Qiao, D., Lin, H., 1998. A B virus replication in mice by RNA interference. Nat. Biotechnol. 21,
novel class of evolutionarily conserved genes defined by piwi are 639–644.
essential for stem cell self-renewal. Genes Dev. 12, 3715–3727. Minor, P.D., 1997. Poliovirus. In: Nathanson, N., Ahmed, R. (Eds.), Viral
Elbashir, S.M., Martinez, J., Patkaniowska, A., Lendeckel, W., Tuschl, T., Pathogenesis. Lippincott-Raven, Philadelphia, pp. 555–574.
2001. Functional anatomy of siRNAs for mediating efficient RNAi in Mourrain, P., Beclin, C., Elmayan, T., Feuerbach, F., Godon, C., Morel,
Drosophila melanogaster embryo lysate. EMBO J. 20, 6877–6888. J.B., Jouette, D., Lacombe, A.M., Nikic, S., Picault, N., Remoue, K.,
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello, Sanial, M., Vo, T.A., Vaucheret, H., 2000. Arabidopsis SGS2 and
C.C., 1998. Potent and specific genetic interference by double-stranded SGS3 genes are required for posttranscriptional gene silencing and
RNA in Caenorhabditis elegans. Nature 391, 806–811. natural virus resistance. Cell 101, 533–542.
Gitlin, L., Andino, R., 2003. Nucleic acid-based immune system: the Novina, C.D., Murray, M.F., Dykxhoorn, D.M., Beresford, P.J., Riess, J.,
antiviral potential of mammalian RNA silencing. J. Virol. 77, 7159– Lee, S.K., Collman, R.G., Lieberman, J., Shankar, P., Sharp, P.A.,
7165. 2002. siRNA-directed inhibition of HIV-1 infection. Nat. Med. 8,
Gitlin, L., Karelsky, S., Andino, R., 2002. Short interfering RNA confers 681–686.
intracellular antiviral immunity in human cells. Nature 418, 430–434. Qin, X.F., An, D.S., Chen, I.S., Baltimore, D., 2003. Inhibiting HIV-1 in-
Guidotti, L.G., Chisari, F.V., 2001. Noncytolytic control of viral infections fection in human T cells by lentiviral-mediated delivery of small interf-
by the innate and adaptive immune response. Annu. Rev. Immunol. ering RNA against CCR5. Proc. Natl. Acad. Sci. U.S.A. 100, 183–188.
19, 65–91. Randall, G., Grakoui, A., Rice, C.M., 2003. Clearance of replicating
Hammond, S.M., Boettcher, S., Caudy, A.A., Kobayashi, R., Hannon, G.J., hepatitis C virus replicon RNAs in cell culture by small interfering
2001a. Argonaute2, a link between genetic and biochemical analyses RNAs. Proc. Natl. Acad. Sci. U.S.A. 100, 235–240.
of RNAi. Science 293, 1146–1150. Ren, R.B., Costantini, F., Gorgacz, E.J., Lee, J.J., Racaniello, V.R., 1990.
Hammond, S.M., Caudy, A.A., Hannon, G.J., 2001b. Post-transcriptional Transgenic mice expressing a human poliovirus receptor: a new model
gene silencing by double-stranded RNA. Nat. Rev. Genet. 2, 110–119. for poliomyelitis. Cell 63, 353–362.
Harborth, J., Elbashir, S.M., Bechert, K., Tuschl, T., Weber, K., 2001. Roignant, J.Y., Carre, C., Mugat, B., Szymczak, D., Lepesant, J.A., An-
Identification of essential genes in cultured mammalian cells using toniewski, C., 2003. Absence of transitive and systemic pathways al-
small interfering RNAs. J. Cell Sci. 114, 4557–4565. lows cell-specific and isoform-specific RNAi in Drosophila. RNA 9,
Herold, J., Andino, R., 2001. Poliovirus RNA replication requires genome 299–308.
circularization through a protein–protein bridge. Mol. Cell 7, 581–591. Rueckert, R.R., 1996. Picornaviridae and their replication. In: Fields,
Jacobsen, S.E., Running, M.P., Meyerowitz, E.M., 1999. Disruption of an B.N., Knipe, D.M., Howley, P.M., et al. (Eds.), Fields Virology, 3rd
RNA helicase/RNAse III gene in Arabidopsis causes unregulated cell ed. Lippincott-Raven Publishers, Philadelphia, pp. 609–652.
division in floral meristems. Development 126, 5231–5243. Rust, R.C., Landmann, L., Gosert, R., Tang, B.L., Hong, W., Hauri, H.P.,
Jacque, J.M., Triques, K., Stevenson, M., 2002. Modulation of HIV-1 Egger, D., Bienz, K., 2001. Cellular COPII proteins are involved in
replication by RNA interference. Nature 418, 435–438. production of the vesicles that form the poliovirus replication complex.
Kapadia, S.B., Brideau-Andersen, A., Chisari, F.V., 2003. Interference of J. Virol. 75, 9808–9818.
hepatitis C virus RNA replication by short interfering RNAs. Proc. Ruvkun, G., 2001. Molecular biology. Glimpses of a tiny RNA world.
Natl. Acad. Sci. U.S.A. 100, 2014–2018. Science 294, 797–799.
Kataoka, Y., Takeichi, M., Uemura, T., 2001. Developmental roles and Sachs, A.B., Davis, R.W., Kornberg, R.D., 1987. A single domain of yeast
molecular characterization of a Drosophila homologue of Arabidopsis poly(A)-binding protein is necessary and sufficient for RNA binding
Argonaute1, the founder of a novel gene superfamily. Genes Cells 6, and cell viability. Mol. Cell Biol. 7, 3268–3276.
313–325. Semler, B.L., Wimmer, E., 2002. Molecular Biology of Picornaviruses.
Ketting, R.F., Haverkamp, T.H., van Luenen, H.G., Plasterk, R.H., 1999. American Society for Microbiology Press, Washington.
Mut-7 of C. elegans, required for transposon silencing and RNA Seo, M.Y., Abrignani, S., Houghton, M., Han, J.H., 2003. Small interfering
interference, is a homolog of Werner syndrome helicase and RNaseD. RNA-mediated inhibition of hepatitis C virus replication in the human
Cell 99, 133–141. hepatoma cell line Huh-7. J. Virol. 77, 810–812.
Lewis, D.L., Hagstrom, J.E., Loomis, A.G., Wolff, J.A., Herweijer, H., Song, E., Lee, S.K., Wang, J., Ince, N., Ouyang, N., Min, J., Chen,
2002. Efficient delivery of siRNA for inhibition of gene expression J., Shankar, P., Lieberman, J., 2003. RNA interference targeting Fas
in postnatal mice. Nat. Genet. 32, 107–108. protects mice from fulminant hepatitis. Nat. Med. 9, 347–351.
M.-C. Saleh et al. / Virus Research 102 (2004) 11–17 17

Stein, P., Svoboda, P., Anger, M., Schultz, R.M., 2003. RNAi: mammalian Waterhouse, P.M., Wang, M.B., Lough, T., 2001. Gene silencing as an
oocytes do it without RNA-dependent RNA polymerase. RNA 9, 187– adaptive defence against viruses. Nature 411, 834–842.
192. Wilson, J.A., Jayasena, S., Khvorova, A., Sabatinos, S., Rodrigue-Gervais,
Svitkin, Y.V., Imataka, H., Khaleghpour, K., Kahvejian, A., Liebig, H.D., I.G., Arya, S., Sarangi, F., Harris-Brandts, M., Beaulieu, S., Richard-
Sonenberg, N., 2001. Poly(A)-binding protein interaction with elF4G son, C.D., 2003. RNA interference blocks gene expression and RNA
stimulates picornavirus IRES-dependent translation. RNA 7, 1743– synthesis from hepatitis C replicons propagated in human liver cells.
1752. Proc. Natl. Acad. Sci. U.S.A. 100, 2783–2788.
Thomas, C.E., Ehrhardt, A., Kay, M.A., 2003. Progress and problems with Xia, H., Mao, Q., Paulson, H.L., Davidson, B.L., 2002. siRNA-mediated
the use of viral vectors for gene therapy. Nat. Rev. Genet. 4, 346–358. gene silencing in vitro and in vivo. Nat. Biotechnol. 10, 1006–1010.
Tiscornia, G., Singer, O., Ikawa, M., Verma, I.M., 2003. A general method Yokota, T., Sakamoto, N., Enomoto, N., Tanabe, Y., Miyagishi, M.,
for gene knockdown in mice by using lentiviral vectors expressing Maekawa, S., Yi, L., Kurosaki, M., Taira, K., Watanabe, M., Mizu-
small interfering RNA. Proc. Natl. Acad. Sci. U.S.A. 100, 1844–1848. sawa, H., 2003. Inhibition of intracellular hepatitis C virus replication
Voinnet, O., 2001. RNA silencing as a plant immune system against by synthetic and vector-derived small interfering RNAs. EMBO Rep.
viruses. Trends Genet. 17, 449–459. 4, 602–608.
Voinnet, O., Pinto, Y.M., Baulcombe, D.C., 1999. Suppression of gene Zhang, G., Budker, V., Wolff, J.A., 1999. High levels of foreign gene
silencing: a general strategy used by diverse DNA and RNA viruses expression in hepatocytes after tail vein injections of naked plasmid
of plants. Proc. Natl. Acad. Sci. U.S.A. 96, 14147–14152. DNA. Hum. Gene. Ther. 10, 1735–1737.
Virus Research 102 (2004) 19–25

Interfering with hepatitis C virus RNA replication


Glenn Randall, Charles M. Rice∗
Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University,
1230 York Avenue, Box 64, New York, NY 10021, USA

Abstract

The emergence of RNA interference (RNAi) as a powerful tool for silencing gene expression has spurred considerable interest in its
experimental and therapeutic potential. RNAi is a cellular process of gene silencing in which small duplexes of RNA specifically target a
homologous sequence for cleavage by cellular ribonucleases. The introduction of 21–23 nucleotide RNA duplexes, termed small interfering
RNAs (siRNAs), into mammalian cells can specifically degrade homologous mRNAs. RNAi efficiently silences the expression of both cellular
and viral RNAs. A number of groups have demonstrated that siRNAs interfere with hepatitis C virus (HCV) gene expression and replication.
Additionally, cellular genes are efficiently silenced in the presence of replicating HCV. These studies lay the foundation for using RNAi as an
experimental tool for studying HCV replication and defining host genes that are significant for viral replication. The potential for RNAi as an
antiviral therapy remains less clear, as it will face many of the challenges that have hindered nucleic acid therapies in the past.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Hepatitis C virus; RNA interference; Small interfering RNAs; Antiviral agents

1. RNA interference 2002). siRNAs are unwound, and single-stranded siRNA


then acts as a guide to substrate selection, leading to the
Mechanisms of gene silencing by the targeting of RNAs cleavage of a homologous target RNA molecule (Hammond
for degradation are conserved in plants, fungi, and animals. et al., 2000).
While post-transcriptional gene silencing (PTGS) has been The demonstration of RNAi in mammalian cells was
described in plants for over a decade, the demonstration in complicated by the existence of the interferon (IFN)
2001 that the closely related process of RNAi has activity signaling pathway. dsRNAs longer than 30 nucleotides
in mammalian cells has greatly expanded the size of its efficiently induce components of the IFN response, in-
scientific audience (Elbashir et al., 2001). It has since been cluding the dsRNA-activated protein kinase R (PKR),
concluded that the molecular machinery of gene silencing 2 ,5 -oligoadenylate synthetase, and RNase L (Manche
appears to be ancient and highly conserved (reviewed in et al., 1992; Minks et al., 1979). These factors promote a
Bosher and Labouesse, 2000; Hannon, 2002). The mecha- global inhibition of gene expression by inhibiting transla-
nism and functions of RNAi are described elsewhere in this tion via the phosphorylation of translation initiation factor
issue, so they will be only briefly summarized in this review. eIF2␣ by PKR and degradation of RNAs by RNase L.
It has been shown in Drosophila that larger double-stranded The induction of IFN was avoided by introducing chem-
RNAs (dsRNAs) are processed into short ∼22-mer siRNAs ically synthesized 21-mer siRNAs into mammalian cells.
by an RNase III-like enzyme, dicer (Bernstein et al., 2001). A siRNA homologous to the cellular mRNA encoding the
These siRNAs associate with the RNA-induced silencing nuclear filament protein lamin A/C specifically silenced the
complex (RISC) (Nykanen et al., 2001). RISC is a multi- target gene expression in mammalian cells (Elbashir et al.,
protein complex composed of members of the argonaute 2001). Silencing was effective, but transient, as the effect
family. In humans, there are seven identified argonaute diminished after 5 days. The transient nature of silencing is
family members: hAgo1/eIF2C1, hAgo2/eIF2C2, hAgo3, likely due to the dilution of siRNAs by cell division. These
hAgo4, Hili, Hiwi, and Hiwi2 (reviewed in Carmell et al., results demonstrated the activity of a gene silencing path-
way that is induced by dsRNA, similar to IFN responses.
∗ Corresponding author. Tel.: +1-212-327-7046; Unlike IFN, however, RNAi initiates a sequence specific
fax: +1-212-327-7048. silencing that is dependent on entirely different molecular
E-mail address: ricec@rockefeller.edu (C.M. Rice). machinery.

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.011
20 G. Randall, C.M. Rice / Virus Research 102 (2004) 19–25

RNAi has been proposed to play a central role in the reg- 2. Hepatitis C virus
ulation of many cellular processes. It has been demonstrated
to silence the expression of transgenes and transposons in HCV is a major public health problem, with 170 mil-
plants. It has been implicated in development and stem cell lion chronically infected people throughout the world
fate (reviewed in Carmell et al., 2002). RNAi also plays a (Anonymous, 1997). Chronically infected individuals are
role in chromosome maintenance. In Saccharomyces pombe, a reservoir for new infections as well as being at risk for
the disruption of RNAi resulted in a loss of silencing het- progression to cirrhosis and hepatocellular carcinoma. Con-
erochromatic repeats at chromosomal centromeres (Volpe sequently, HCV infection is the leading indicator of liver
et al., 2002). The related PTGS is a well-documented an- transplant (Fishman et al., 1996). Antiviral therapy con-
tiviral system in plants (Al-Kaff et al., 1998; Dougherty sisting of peginterferon-alfa 2a-ribavirin shows sustained
et al., 1994; Ruiz et al., 1998), and RNAi has been shown to response in only about half of the treated patients that are in-
have antiviral function in insect cells (Li et al., 2002). These fected with HCV genotypes 1 and 4 (Heathcote et al., 2000;
processes silence the expression of transposons, repetitive Zeuzem et al., 2000). These factors have led to a push to de-
elements, and viruses (reviewed in Voinnet et al., 1999). velop new antiviral therapies. Additionally, the emergence
Viral inhibitors of RNAi have been isolated from plant of drug resistance in RNA virus infections emphasizes the
viruses (Brigneti et al., 1998; Li and Ding, 2001; Voinnet need for multiple drug targets to limit viral escape.
et al., 2000) and also the animal virus, flock house virus HCV belongs to the family Flaviviridae, which includes
(Li et al., 2002; Lindenbach and Rice, 2002). It is currently the flaviviruses, pestiviruses, hepaciviruses, and the GB
unclear whether RNAi is an innate antiviral defense in viruses (reviewed in Lindenbach and Rice, 2001). The
mammals. 9.6-kb HCV RNA genome of positive (+) polarity consists

Fig. 1. Schematic representation of the HCV genome and siRNAs reported to silence HCV. (A) Shown is the HCV genome; with the 5 NTR, followed
by a large open reading frame that is translated and processed into at least 10 proteins (C, E1, E2, p7, NS2, NS3, NS4A, NS4B, NS5A, and NS5B) and
the 3 NTR. The numbers and arrows refer to the siRNA sites. (B) siRNAs that have been reported to successfully silence HCV. siRNAs are described
both by their sequence location (nucleotide) and the names given to them in the respective publications. The nucleotide number given is the position
of the 5 nucleotide of the siRNA within the HCV-Con1 sequence. The reported siRNA sequences, the gene that is targeted by the siRNA, and the
reference for each siRNA are given. Abbreviations: C, core; E, envelope glycoprotein; NS, nonstructural protein; NTR, nontranslated region.
G. Randall, C.M. Rice / Virus Research 102 (2004) 19–25 21

of a 5 nontranslated region (NTR) containing an internal quickly began to study the relevance of RNAi to their virus
ribosome entry site (IRES), a single open reading frame of interest. The leap for hepatitis C virologists is relatively
of approximately 3000 codons, and a 3 NTR containing small. As an RNA virus, the genome may be susceptible to
a highly conserved 98-nucleotide sequence (Fig. 1A). The cleavage. Since HCV replication generates dsRNAs, HCV
IRES directs the translation of a polypeptide, which is sub- may naturally activate RNAi, which would in turn serve as
sequently processed by host and viral proteases into at least an innate host defense against virus infection. Finally, a great
10 individual proteins. The structural proteins C (core), need exists for the development of effective HCV antiviral
E1, and E2 (envelope glycoproteins) are contained at the therapies. The specific and effective nature of silencing by
N-terminus of the polyprotein, and are followed by p7 RNAi are attractive therapeutic characteristics.
and the nonstructural proteins (NS) 2, 3, 4A, 4B, 5A, and A number of groups have used the HCV replicon system
5B. NS 3–5B comprise a functional replication complex to evaluate the antiviral potential of RNAi (Kapadia et al.,
that promotes transcription of a genomic (−) strand inter- 2003; Randall et al., 2003; Seo et al., 2003; Wilson et al.,
mediate. This serves as a template for production of (+) 2003; Yokota et al., 2003). These studies have focused pri-
strands that are either translated or packaged into virions as marily on two questions: (i) Does HCV interfere with RNAi?
genomic RNAs. (ii) Is HCV sensitive to RNAi? Regarding the first question,
The structural proteins C, E1, and E2 compose the cap- it appears that HCV does not inhibit RNAi following the in-
sid and constituents of the viral envelope. E1 and E2 are troduction of siRNAs into cells containing replicating HCV.
thought to mediate HCV binding to the cell and viral en- Both cellular (lamin A/C) and viral (HCV) RNAs were effec-
try. Enzymatic properties of HCV proteins are well defined; tively silenced in the presence of replicating HCV (Randall
NS5B is the RNA-dependent RNA polymerase, NS3 has et al., 2003). The efficiencies of silencing lamin A/C expres-
helicase and serine protease activity, and NS2-NS3 form a sion in Huh-7.5 cells were similar either in the presence or
distinct autoprotease. NS4A is a cofactor for NS3 proteol- absence of replicating HCV RNAs. Since the introduction
ysis. The functions of p7, NS2 following autoproteolysis, of siRNAs into cells bypasses the initial processing steps of
NS4B, and NS5A are less clear. A number of studies have larger dsRNAs into siRNAs by Dicer, it is possible that HCV
attempted to define functions of viral proteins, particularly may interfere with an earlier step in the RNAi pathway. It
NS5A and the core protein, by ectopic protein expression or should be re-emphasized that so far no viruses that infect
the identification of protein interaction partners (reviewed mammals have been reported to encode inhibitors of RNAi.
in Tellinghuisen and Rice, 2002). The significance of these A general consensus was reached that HCV RNAs, like
putative functions and interactions for HCV replication was those of many other viruses, are indeed sensitive to RNAi.
not tested by genetic analysis, owing to the lack of an effi- siRNAs were designed to target cleavage of HCV RNAs
cient cell culture system. at various regions of the HCV genome (Fig. 1). The in-
Although HCV is notoriously difficult to grow in vitro, troduction of siRNAs into cells containing HCV replicons
systems have been developed that sustain efficient repli- effectively silenced HCV expression in a number of differ-
cation of HCV in cell culture. These are replicon-based ent assays. HCV antigen expression declined after treatment
strategies in which the HCV IRES drives expression of the with siRNAs, as assayed by either the level of replicon
neomycin phosphotransferase gene, while a heterologous reporter expression or by the levels of HCV protein expres-
IRES from encephalomyocarditis virus (EMCV) promotes sion in all studies. siRNAs triggered an exponential decline
translation of the HCV polypeptide (Blight et al., 2000; in HCV RNA levels, with a maximal reduction achieved
Lohmann et al., 1999). HCV replicon replication promotes and maintained after 4 days of a single siRNA treatment
continued expression of the neomycin phosphotransferase (Randall et al., 2003). Strand-specific probing for HCV
gene that confers G418 resistance to cells. This allows the RNAs revealed that siRNAs reduced the levels of both (+)
establishment of cell populations that contain replicating and (−) strand RNA (Wilson et al., 2003).
HCV. G418-resistant foci are also a means of quantifying As shown in Fig. 1B, the siRNAs effectively targeted re-
the number of cells that contain stably replicating HCV. The gions in the 5 NTR, Core, NS3, NS4B, and NS5B. While
replicon system is useful for dissection of HCV replication the relative efficiencies of individual siRNAs vary, it appears
and protein functions; however, the production of infectious that all regions of HCV RNA are accessible to the RNAi
virus in vitro has yet to be documented. In addition to molec- machinery. It should be noted that not all siRNAs were ef-
ular studies of HCV replication, replicons provide an excel- fective. Success rates varied between studies, but it appears
lent system to evaluate HCV antiviral agents in cell culture that approximately half of the siRNAs were effective in at
(reviewed in Randall and Rice, 2001). least partially silencing HCV.
The efficiencies of silencing HCV varied depending on
the experimental conditions of each study, probably indica-
3. HCV is sensitive to RNAi tive of the efficiency of siRNA introduction into cells. Under
optimal conditions, it was shown that siRNAs could clear
While the initial demonstrations of RNAi in mammalian replicating HCV RNAs from >98% of cells in a series of two
cells showed silencing of cellular transcripts, virologists experiments (Randall et al., 2003). In the first experiment,
22 G. Randall, C.M. Rice / Virus Research 102 (2004) 19–25

HCV NS5A expression was only detectable by immunoflu- derived siRNAs (Wilson et al., 2003). The majority of cells
orescence in 1% of cells, as compared with cells treated within this population were resistant to the establishment
with an irrelevant siRNA. The second experiment examined of HCV replication. Recently, vectors that express shRNAs,
the formation of G418-resistant colonies following treatment but not chemically synthesized siRNAs, have been shown
by siRNAs. As described earlier, G418-resistant colonies to induce interferon in a subset of constructs (Bridge et al.,
form dependent upon HCV replication and the expression of 2003). This result is likely to be due to either the increased
the neomycin phosphotransferase gene from HCV replicon length of dsRNA (siRNA stem plus an extended loop) or
RNAs. Cells containing HCV replicons were treated with aberrant transcription initiation or termination resulting in
siRNAs and grown in the absence of selection for 1 week. long dsRNAs. The studies on HCV silencing by siRNA pro-
G418 selection was then reapplied and G418 resistant foci ducing vectors will need to be re-examined for the induction
were counted 10 days later. Cells treated with HCV siR- of interferon. Assuming that these vectors silence HCV in
NAs formed G418 resistant foci in less than 2% of cells, as an interferon-independent manner, these results establish the
compared with cells treated with irrelevant siRNAs. HCV concept of creating HCV resistant cells. Additionally, the
siRNAs did not affect cell viability, since treated cells grew stable expressing cell populations should provide a model
in the absence of selection. It was unclear whether the re- system to study viral escape from RNAi.
maining G418 resistant cells received sufficient levels of the
siRNAs; or alternatively, if these cells contained viral RNAs
which escaped RNAi either due to mutation of viral se- 4. Prospects and questions for HCV and RNAi
quences or mutations in the RNAi machinery. Nonetheless,
the ability of siRNAs to produce noncytolytic clearance of Numerous questions follow these initial studies that
HCV is attractive from a therapeutic viewpoint. demonstrated the sensitivity of HCV to RNAi. Some of
A hallmark of RNAi, as compared with IFN, is the these include the following: (i) Is RNAi an innate antiviral
sequence-specific nature of RNAi gene silencing. siRNAs pathway in mammals? (ii) What is the mechanism of HCV
that differed from their HCV target sequence by two or more clearance? (iii) How do we exploit RNAi to define the sig-
bases did not efficiently silence HCV. In one study, two nificance of host interactions for the HCV life cycle? (iv)
HCV genotype 1b replicons that differed from each other by What is the therapeutic potential of RNAi?
three bases within the siRNA target sequence were silenced
only by the exact homologous siRNA (Randall et al., 2003). 4.1. Is RNAi an innate antiviral pathway in mammals?
The introduction of siRNAs did not induce the expression
of IFN-induced genes MxA and PKR, and additionally, To date, it has not been demonstrated that RNAi is an
siRNAs were more effective at reducing HCV RNA levels innate antiviral defense in mammals; or conversely, that
than high doses (100 units/ml) of IFN-␣ (Kapadia et al., viruses express functions that inhibit mammalian RNAi. Ad-
2003). It was not tested whether the addition of IFN-␣ and ditionally, a large number of viruses have been reported to
HCV siRNAs showed an additive effect in reducing HCV be sensitive to RNAi (reviewed in Gitlin and Andino, 2003).
RNA levels. Taken together, these experiments showed that Since mammals have an immune system and interferon path-
HCV silencing by siRNAs was independent of, and perhaps way that perform antiviral roles, it is possible that RNAi
more effective than, the effects of IFN-␣. has evolved to perform other gene regulatory functions in
The previously described experiments tested the ability of mammals. To determine whether RNAi is an innate antiviral
siRNAs to clear replicating HCV RNAs from cells. It was pathway, researchers need to address whether (i) HCV repli-
also demonstrated that the introduction of HCV siRNAs into cation induces the expression or activity of components of
Huh-7 cells prior to HCV replicon RNAs precluded the es- the RNAi pathway, (ii) replicating HCV is naturally targeted
tablishment of HCV replication (Wilson et al., 2003). This by the RNAi machinery in the absence of experimentally
effect was transient, with efficient inhibition of replication introduced siRNAs or shRNAs, (iii) inhibition of RNAi en-
lasting for 5 days. In order to improve upon the transient hances HCV replication, and (iv) HCV encodes inhibitors
nature of silencing following siRNAs, a number of strate- of the RNAi pathway.
gies now exist for stable RNAi. These are based on vectors
containing a pol III promoter driving expression of either (i) 4.2. What is the mechanism of anti-HCV action for siRNAs?
dual single stranded RNAs that could form a ds siRNA or
(ii) small hairpin RNAs (shRNAs) which are then processed Replicating HCV RNAs are at least partially double
in vivo to yield a functional siRNA (Brummelkamp et al., stranded and should in theory be susceptible to RNAi; how-
2002; Paddison et al., 2002). It was examined whether HCV ever, the RNAs may be resistant to the RNAi machinery due
RNAs could be silenced using vector-based stable silencing. to either protection from ribonucleoproteins or a privileged
HCV antigen expression was partially silenced following subcellular localization. The targeting of mRNAs, but not
the transient introduction of the vectors into cells containing genomic viral RNAs has been proposed as the mechanism
replicating HCV (Yokota et al., 2003). In another study, sta- of siRNA clearance for both influenza virus and respiratory
ble cell populations were established that expressed vector syncytial virus (Bitko and Barik, 2001; Ge et al., 2003). It is
G. Randall, C.M. Rice / Virus Research 102 (2004) 19–25 23

unclear whether the anti-HCV activity of siRNAs functions systems to early stages of the HCV life cycle since HCV
by a similar mechanism. This is difficult to test in the case glycoprotein binding and viral entry into cells can be stud-
of HCV, since the (+) strand can represent both genome ied. Earlier studies tested the binding efficiencies of solu-
and message. ble, truncated versions of HCV glycoproteins either to cells,
The majority of siRNAs that efficiently silence HCV were or cellular proteins that were proposed to represent puta-
designed to target the (+) strand, or both (+) and (−) strand. tive HCV receptors. This led to the identification of several
In one study, a reduction of both (+) and (−) strand RNAs putative receptors, including CD81, low density lipid recep-
was observed following silencing (Wilson et al., 2003). This tor, scavenger receptor B1, DC-SIGN and L-SIGN (Agnello
suggest that either (i) the HCV replicase and minus strand et al., 1999; Gardner et al., 2003; Lozach et al., 2003;
RNAs in particular are sensitive to RNAi, or (ii) the HCV Pileri et al., 1998; Pohlmann et al., 2003; Scarselli et al.,
replication complex is relatively unstable and that targeting 2002). The significance of these putative receptors for HCV
of the (+) strands indirectly results in a decrease in (−) glycoprotein-dependent entry can be tested by silencing the
strands. One siRNA (286, Fig. 1B) has been described that receptor candidates with siRNAs and testing the ability of
should target only (−) strands (Seo et al., 2003; Yokota et al., these cells to be infected with HIV-HCV pseudotypes. Sim-
2003). When compared with other siRNAs in the study, it ilar approaches have confirmed the significance of CD4 and
was partially effective at silencing HCV. A 30% reduction CCR5 for HIV entry (Martinez et al., 2002; Novina et al.,
in HCV replicon reporter activity was observed as compared 2002). RNAi will continue to be a powerful tool to probe
with a 90% reduction for the optimal siRNA in this exper- HCV–host interactions as systems to study other aspects of
iment (Yokota et al., 2003). Although cleavage of the (−) the viral life cycle, such as viral assembly and egress, are
strand was not demonstrated, this result suggests that the developed.
replicase complex may be at least partially accessible to siR-
NAs. It remains to be determined whether polysomal RNAs 4.4. What is the therapeutic potential of RNAi?
and RNAs in the replication complex, as either templates or
nascent products, are similarly accessible to RNAi-mediated The initial studies demonstrating that HCV is suscep-
cleavage. tible to RNAi provide proof of principle for RNAi-based
anti-HCV therapy. However, many obstacles remain along
4.3. How do we exploit RNAi to define the significance of the journey from “bench to bedside.” These include issues
host interactions for the HCV life cycle? regarding efficacy, delivery, and resistance/viral escape. Effi-
cacy has traditionally been a stumbling block for nucleic acid
HCV has an inordinately large number of reported therapy strategies, whether they be naked nucleic acids or vi-
viral–host interactions that have no known significance in ral vectors designed to express the therapeutic nucleic acids.
the viral life cycle. Since HCV does not interfere with the RNAs are generally unstable in cells and patient sera. At-
silencing of host genes, RNAi has significant potential as a tempts are underway to both stabilize and target chemically
tool to investigate the significance of these interactions for synthesized RNAs in vivo without foregoing the efficacy of
the HCV life cycle. The applicability of RNAi is currently these RNAs for silencing gene expression. One advantage to
limited by the systems with which to characterize the HCV HCV as a disease model is that the liver is relatively profi-
life cycle. The previously described replicon system holds cient in the uptake of nucleic acids. Indeed, the liver has been
great promise in characterizing the significance of host the organ of choice for demonstrating the potential of RNAi
genes for the translation and replication of HCV RNAs. therapy in vivo. Liver uptake of siRNAs in mice is efficient,
This approach would encompass host genes that were iden- resulting in the silencing of the targeted mRNA sequence,
tified to interact with HCV RNAs and components of the whether it is an HCV transgene or cellular genes (McCaffrey
viral replicase complex (NS3, 4A, 4B, 5A, and 5B). Ad- et al., 2002). Silencing of Fas or caspase 8 by siRNAs in vivo
ditionally, cellular signaling pathways that intersect with prevents the symptoms of hepatitis in mouse model systems
HCV can be similarly examined. For example, the mech- (Song et al., 2003; Zender et al., 2003). Recently, RNAi was
anism of action for IFN-␣, -␤, or -␥ in the clearance of shown to inhibit hepatitis B virus replication in mouse liver
HCV RNAs is currently unknown, both in vitro and in vivo. (McCaffrey et al., 2003). The anti-HCV potential of siRNAs
IFN-induced genes can be silenced prior to the addition of in a system that produces infectious virus has yet to be doc-
IFN to identify IFN-induced genes that perform effector umented. Mice that contain chimeric mouse/human livers
functions able to promote the clearance of HCV. support viral infection and replication and would provide a
While systems that promote HCV assembly and egress model to test the efficacy of siRNAs against HCV in vivo,
are still elusive, improved models to study HCV entry have in a fully infectious system (Mercer et al., 2001).
been described recently. Retroviral pseudotypes, in which The specific silencing by RNAi without induction of non-
HIV genomes are enveloped in HCV glycoproteins, infect a specific IFN responses is attractive from a therapeutic stand-
subset of liver cell lines in a HCV glycoprotein-dependent point. However, the specificity of silencing highlights the
manner (Bartosch et al., 2003; Hsu et al., 2003). This model possibility of viral escape mutants. A limited number of
represents an advance compared with previous surrogate highly conserved RNA sequences that should be constrained
24 G. Randall, C.M. Rice / Virus Research 102 (2004) 19–25

for viral escape exist in the HCV 5 and 3 NTRs. Mul- density lipoprotein receptor. Proc. Natl. Acad. Sci. U.S.A. 96, 12766–
12771.
tiple siRNAs targeting evolutionarily restricted HCV RNA
Al-Kaff, N.S., Covey, S.N., Kreike, M.M., Page, A.M., Pinder, R., Dale,
sequences may limit the emergence of escape mutants. Sta- P.J., 1998. Transcriptional and posttranscriptional plant gene silencing
ble RNAi silencing vectors have been used to generate cell in response to a pathogen. Science 279, 2113–2115.
lines that constitutively express high levels of HCV siRNAs Anonymous, 1997. World Health Organization—Hepatitis C: global preva-
(Wilson et al., 2003). These cell lines could be used to ad- lence. Wkly. Epidemiol. Rec. 72, 341–344.
dress the question of viral escape in cell culture since the Bartosch, B., Dubuisson, J., Cosset, F.L., 2003. Infectious hepatitis C
virus pseudo-particles containing functional E1-E2 envelope protein
caveats associated with the transfection efficiency of chem- complexes. J. Exp. Med. 197, 633–642.
ically synthesized siRNAs into cells are avoided. Variations Bernstein, E., Caudy, A.A., Hammond, S.M., Hannon, G.J., 2001. Role
of these stable RNAi silencing vectors may also function in for a bidentate ribonuclease in the initiation step of RNA interference.
gene therapy wherein numerous modes exist for expressing Nature 409, 363–366.
therapeutic RNAs, such as antisense RNAs or ribozymes. Bitko, V., Barik, S., 2001. Phenotypic silencing of cytoplasmic genes
using sequence-specific double-stranded short interfering RNA and its
A goal would be the prevention of productive HCV infec- application in the reverse genetics of wild type negative-strand RNA
tion by siRNA expression in liver cells. Successful treat- viruses. BMC Microbiol. 1, 34.
ment may not require a high efficiency of siRNA delivery Blight, K.J., Kolykhalov, A.A., Rice, C.M., 2000. Efficient initiation of
since the liver has regenerative potential. Infected liver cells HCV RNA replication in cell culture. Science 290, 1972–1974.
that are killed by T cell-mediated cytolytic clearance would Bosher, J.M., Labouesse, M., 2000. RNA interference: genetic wand and
genetic watchdog. Nat. Cell. Biol. 2, E31–E36.
gradually be replaced by HCV resistant cells. In theory, this Bridge, A.J., Pebernard, S., Ducraux, A., Nicoulaz, A.L., Iggo, R., 2003.
would result in a HCV resistant liver repopulated by cells Induction of an interferon response by RNAi vectors in mammalian
expressing HCV siRNAs. cells. Nat. Genet. 34, 263–264.
Brigneti, G., Voinnet, O., Li, W.X., Ji, L.H., Ding, S.W., Baulcombe, D.C.,
1998. Viral pathogenicity determinants are suppressors of transgene
silencing in Nicotiana benthamiana. EMBO J. 17, 6739–6746.
5. Conclusions Brummelkamp, T.R., Bernards, R., Agami, R., 2002. A system for stable
expression of short interfering RNAs in mammalian cells. Science
Recent studies have solidified the use of siRNAs to inhibit 296, 550–553.
the expression of target RNAs, whether they are cellular or Carmell, M.A., Xuan, Z., Zhang, M.Q., Hannon, G.J., 2002. The Arg-
viral in origin. RNAi as an “experimental tool” will undoubt- onaute family: tentacles that reach into RNAi, developmental control,
stem cell maintenance, and tumorigenesis. Genes Dev. 16, 2733–2742.
edly be exploited to define HCV replication and pathogene- Dougherty, W.G., Lindbo, J.A., Smith, H.A., Parks, T.D., Swaney, S.,
sis. These experimental approaches can be applied to under- Proebsting, W.M., 1994. RNA-mediated virus resistance in transgenic
standing HCV RNA replication and the significance of host plants: exploitation of a cellular pathway possibly involved in RNA
genes at various stages of the viral life cycle. A consensus degradation. Mol. Plant Microb. Interact. 7, 544–552.
has been achieved that HCV is sensitive to the introduction Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl,
T., 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference
of siRNAs. Although RNAi can be activated to clear HCV in cultured mammalian cells. Nature 411, 494–498.
RNAs, it remains to be shown whether RNAi is a natural host Fishman, J.A., Rubin, R.H., Koziel, M.J., Periera, B.J., 1996. Hepatitis C
defense against HCV. Similarly, it will be interesting to see virus and organ transplantation. Transplantation 62, 147–154.
whether HCV gene products interact with the RNAi machin- Gardner, J.P., Durso, R.J., Arrigale, R.R., Donovan, G.P., Maddon, P.J.,
ery. Finally, a major reason for studying HCV is the clini- Dragic, T., Olson, W.C., 2003. L-SIGN (CD 209L) is a liver-specific
capture receptor for hepatitis C virus. Proc. Natl. Acad. Sci. U.S.A.
cal need for improved therapeutics. Despite many obstacles, 100, 4498–4503.
RNAi provides a new approach that, in addition to traditional Ge, Q., McManus, M.T., Nguyen, T., Shen, C.H., Sharp, P.A., Eisen,
anti-viral therapeutics, may effectively treat HCV infection. H.N., Chen, J., 2003. RNA interference of influenza virus production
by directly targeting mRNA for degradation and indirectly inhibiting
all viral RNA transcription. Proc. Natl. Acad. Sci. U.S.A. 100, 2718–
2723.
Acknowledgements Gitlin, L., Andino, R., 2003. Nucleic acid-based immune system: the
antiviral potential of mammalian RNA silencing. J. Virol. 77, 7159–
G.R. and C.M.R. are supported in part by grants from 7165.
the Public Health Service (CA57973 and AI40034) and the Hammond, S.M., Bernstein, E., Beach, D., Hannon, G.J., 2000. An
Greenberg Medical Research Institute. G.R. is supported by RNA-directed nuclease mediates post-transcriptional gene silencing in
Drosophila cells. Nature 404, 293–296.
postdoctoral fellowship PF-02-016-01-MBC from the Amer- Hannon, G.J., 2002. RNA interference. Nature 418, 244–251.
ican Cancer Society. We thank Catherine Murray for critical Heathcote, E.J., Shiffman, M.L., Cooksley, W.G., Dusheiko, G.M., Lee,
reading of the manuscript. S.S., Balart, L., Reindollar, R., Reddy, R.K., Wright, T.L., Lin, A.,
Hoffman, J., De Pamphilis, J., 2000. Peginterferon alfa-2a in patients
with chronic hepatitis C and cirrhosis. N. Engl. J. Med. 343, 1673–
1680.
References Hsu, M., Zhang, J., Flint, M., Logvinoff, C., Cheng-Mayer, C., Rice,
C.M., McKeating, J.A., 2003. Hepatitis C virus glycoproteins mediate
Agnello, V., Abel, G., Elfahal, M., Knight, G.B., Zhang, Q.X., 1999. pH-dependent cell entry of pseudotyped retroviral particles. Proc. Natl.
Hepatitis C virus and other Flaviviridae viruses enter cells via low Acad. Sci. U.S.A. 100, 7271–7276.
G. Randall, C.M. Rice / Virus Research 102 (2004) 19–25 25

Kapadia, S.B., Brideau-Andersen, A., Chisari, F.V., 2003. Interference of Pohlmann, S., Zhang, J., Baribaud, F., Chen, Z., Leslie, G.J., Lin, G.,
hepatitis C virus RNA replication by short interfering RNAs. Proc. Granelli-Piperno, A., Doms, R.W., Rice, C.M., McKeating, J.A.,
Natl. Acad. Sci. U.S.A. 100, 2014–2018. 2003. Hepatitis C virus glycoproteins interact with DC-SIGN and
Li, W.X., Ding, S.W., 2001. Viral suppressors of RNA silencing. Curr. DC-SIGNR. J. Virol. 77, 4070–4080.
Opin. Biotechnol. 12, 150–154. Randall, G., Rice, C.M., 2001. Hepatitis C virus cell culture replication
Li, H., Li, W.X., Ding, S.W., 2002. Induction and suppression of RNA systems: their potential use for the development of antiviral therapies.
silencing by an animal virus. Science 296, 1319–1321. Curr. Opin. Infect. Dis. 14, 743–747.
Lindenbach, B.D., Rice, C.M., 2001. Flaviviridae: the viruses and their Randall, G., Grakoui, A., Rice, C.M., 2003. Clearance of replicating
replication. In: Knipe, D.M., Howley, P.M. (Eds.), Fields Virology, hepatitis C virus replicon RNAs in cell culture by small interfering
4th ed. Lippincott-Williams and Wilkins, Philadelphia, pp. 991–1041. RNAs. Proc. Natl. Acad. Sci. U.S.A. 100, 235–240.
Lindenbach, B.D., Rice, C.M., 2002. RNAi targeting an animal virus: Ruiz, M.T., Voinnet, O., Baulcombe, D.C., 1998. Initiation and mainte-
news from the front. Mol. Cell. 9, 925–927. nance of virus-induced gene silencing. Plant Cell 10, 937–946.
Lohmann, V., Korner, F., Koch, J.O., Herian, U., Theilmann, L., Barten- Scarselli, E., Ansuini, H., Cerino, R., Roccasecca, R.M., Acali, S., Filo-
schlager, R., 1999. Replication of subgenomic hepatitis C virus RNAs camo, G., Traboni, C., Nicosia, A., Cortese, R., Vitelli, A., 2002. The
in a hepatoma cell line. Science 285, 110–113. human scavenger receptor class B type I is a novel candidate receptor
Lozach, P.Y., Lortat-Jacob, H., de Lacroix de Lavalette, A., Staropoli, I., for the hepatitis C virus. EMBO J. 21, 5017–5025.
Foung, S., Amara, A., Houles, C., Fieschi, F., Schwartz, O., Vire- Seo, M.Y., Abrignani, S., Houghton, M., Han, J.H., 2003. Small interfering
lizier, J.L., Arenzana-Seisdedos, F., Altmeyer, R., 2003. DC-SIGN RNA-mediated inhibition of hepatitis C virus replication in the human
and L-SIGN are high affinity binding receptors for hepatitis C virus hepatoma cell line Huh-7. J. Virol. 77, 810–812.
glycoprotein E2. J. Biol. Chem. 278, 20358–20366. Song, E., Lee, S.K., Wang, J., Ince, N., Ouyang, N., Min, J., Chen,
Manche, L., Green, S.R., Schmedt, C., Mathews, M.B., 1992. Interactions J., Shankar, P., Lieberman, J., 2003. RNA interference targeting Fas
between double-stranded RNA regulators and the protein kinase DAI. protects mice from fulminant hepatitis. Nat. Med. 9, 347–351.
Mol. Cell. Biol. 12, 5238–5248. Tellinghuisen, T.L., Rice, C.M., 2002. Interaction between hepatitis C
Martinez, M.A., Gutierrez, A., Armand-Ugon, M., Blanco, J., Parera, M., virus proteins and host cell factors. Curr. Opin. Microbiol. 5, 419–427.
Gomez, J., Clotet, B., Este, J.A., 2002. Suppression of chemokine Voinnet, O., Pinto, Y.M., Baulcombe, D.C., 1999. Suppression of gene
receptor expression by RNA interference allows for inhibition of silencing: a general strategy used by diverse DNA and RNA viruses
HIV-1 replication. AIDS 16, 2385–2390. of plants. Proc. Natl. Acad. Sci. U.S.A. 96, 14147–14152.
McCaffrey, A.P., Meuse, L., Pham, T.T., Conklin, D.S., Hannon, G.J., Voinnet, O., Lederer, C., Baulcombe, D.C., 2000. A viral movement
Kay, M.A., 2002. RNA interference in adult mice. Nature 418, 38–39. protein prevents spread of the gene silencing signal in Nicotiana
McCaffrey, A.P., Nakai, H., Pandey, K., Huang, Z., Salazar, F.H., Xu, H., benthamiana. Cell 103, 157–167.
Wieland, S.F., Marion, P.L., Kay, M.A., 2003. Inhibition of hepatitis Volpe, T.A., Kidner, C., Hall, I.M., Teng, G., Grewal, S.I., Martienssen,
B virus in mice by RNA interference. Nat. Biotechnol. 21, 639–644. R.A., 2002. Regulation of heterochromatic silencing and histone H3
Mercer, D.F., Schiller, D.E., Elliott, J.F., Douglas, D.N., Hao, C., Rinfret, lysine-9 methylation by RNAi. Science 297, 1833–1837.
A., Addison, W.R., Fischer, K.P., Churchill, T.A., Lakey, J.R., Tyrrell, Wilson, J.A., Jayasena, S., Khvorova, A., Sabatinos, S., Rodrigue-Gervais,
D.L., Kneteman, N.M., 2001. Hepatitis C virus replication in mice I.G., Arya, S., Sarangi, F., Harris-Brandts, M., Beaulieu, S., Richard-
with chimeric human livers. Nat. Med. 7, 927–933. son, C.D., 2003. RNA interference blocks gene expression and RNA
Minks, M.A., West, D.K., Benvin, S., Baglioni, C., 1979. Structural re- synthesis from hepatitis C replicons propagated in human liver cells.
quirements of double-stranded RNA for the activation of 2 ,5 -oligo(A) Proc. Natl. Acad. Sci. U.S.A. 100, 2783–2788.
polymerase and protein kinase of interferon-treated HeLa cells. J. Yokota, T., Sakamoto, N., Enomoto, N., Tanabe, Y., Miyagishi, M.,
Biol. Chem. 254, 10180–10183. Maekawa, S., Yi, L., Kurosaki, M., Taira, K., Watanabe, M., Mizu-
Novina, C.D., Murray, M.F., Dykxhoorn, D.M., Beresford, P.J., Riess, J., sawa, H., 2003. Inhibition of intracellular hepatitis C virus replication
Lee, S.K., Collman, R.G., Lieberman, J., Shankar, P., Sharp, P.A., by synthetic and vector-derived small interfering RNAs. EMBO Rep.
2002. siRNA-directed inhibition of HIV-1 infection. Nat. Med. 3, 3. 4, 602–608.
Nykanen, A., Haley, B., Zamore, P.D., 2001. ATP requirements and small Zender, L., Hutker, S., Liedtke, C., Tillmann, H.L., Zender, S., Mundt, B.,
interfering RNA structure in the RNA interference pathway. Cell 107, Waltemathe, M., Gosling, T., Flemming, P., Malek, N.P., Trautwein,
309–321. C., Manns, M.P., Kuhnel, F., Kubicka, S., 2003. Caspase 8 small
Paddison, P.J., Caudy, A.A., Bernstein, E., Hannon, G.J., Conklin, D.S., interfering RNA prevents acute liver failure in mice. Proc. Natl. Acad.
2002. Short hairpin RNAs (shRNAs) induce sequence-specific silenc- Sci. U.S.A. 100, 7797–7802.
ing in mammalian cells. Genes Dev. 16, 948–958. Zeuzem, S., Feinman, S.V., Rasenack, J., Heathcote, E.J., Lai, M.Y., Gane,
Pileri, P., Uematsu, Y., Campagnoli, S., Galli, G., Falugi, F., Petracca, E., O’Grady, J., Reichen, J., Diago, M., Lin, A., Hoffman, J., Brunda,
R., Weiner, A.J., Houghton, M., Rosa, D., Grandi, G., Abrignani, S., M.J., 2000. Peginterferon alfa-2a in patients with chronic hepatitis C.
1998. Binding of hepatitis C virus to CD81. Science 282, 938–941. N. Engl. J. Med. 343, 1666–1672.
Virus Research 102 (2004) 27–35

Control of nonsegmented negative-strand RNA


virus replication by siRNA
Sailen Barik∗
Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama,
307 University Blvd., Mobile, AL 36688, USA
I dedicate this article to my Ph.D. advisor and mentor, Professor N.C. Mandal, on the eve of his retirement from Bose Institute,
Calcutta; his love of science continues to be a guiding light

Abstract

Our laboratory provided the first proof-of-concept that double-stranded short interfering RNA (ds-siRNA) can act as potent and specific
antiviral agents. Designed against specific mRNAs of nonsegmented negative-stranded RNA (NNR) viruses, siRNAs abrogated expression
of the corresponding viral proteins, and generated the predicted viral phenotypes. Knockdown was demonstrated across different genera:
respiratory syncytial virus (RSV), a pneumovirus; vesicular stomatitis virus (VSV), a rhabdovirus; and human parainfluenza virus (HPIV),
a paramyxovirus. The targeted genes could have a wide range of functions, thus documenting the versatility of the technique. Interestingly,
antisense single-stranded siRNA (ss-siRNA) was also effective, albeit at a higher concentration. NNR viral genomic and antigenomic RNA,
which are encapsidated by nucleocapsid protein and serve as templates for viral RNA-dependent RNA polymerase, were resistant to siRNA.
Together, siRNAs offer complementary advantages over traditional mutational analyses that are difficult to perform in NNR viruses, and are
also an important new tool to dissect host–virus interactive pathways.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Respiratory syncytial virus; Vesicular stomatitis virus; Parainfluenza virus; RNA interference; siRNA; Host–virus interaction

1. Introduction The specificity and efficacy of siRNA-mediated PTGS


against intracellular cytoplasmic parasites was originally
RNA interference (RNAi), mediated by short interfering documented using respiratory syncytial virus (RSV) as a
RNA molecules (siRNA), is now recognized as a major test model (Bitko and Barik, 2001b). Subsequently, a flurry
mechanism of post-transcriptional gene silencing (PTGS) of exciting studies reproduced similar success in a variety of
in all metazoan eukaryotes (Hannon, 2002; Pickford and important animal and plant viruses (See other articles of this
Cogoni, 2003). The application of the siRNA technology volume). The NNR virus family, in particular, encompasses
in mammalian cell culture received a major impetus fol- a large roster of members that infect practically every terres-
lowing the discovery that specific double-stranded siRNA trial species and cause diseases of enormous consequence
molecules in the 21-nucleotide range effectively degrade the in human and animal health (Banerjee et al., 1991; Palese
cognate mRNAs, and thus abrogate the expression of the et al., 1996). Two major families of the NNR viruses are:
corresponding proteins (Elbashir et al., 2001). These siR- Paramyxoviridae and Rhabodoviridae. Clinically important
NAs are apparently too short to activate the cellular “inter- paramyxoviruses include human and bovine respiratory
feron response” that is known to cause a general inhibition syncytial viruses (RSV) and parainfluenza viruses (PIV),
of the cellular translation machinery (Elbashir et al., 2001; measles virus, mumps virus, canine distemper virus, and the
Bitko and Barik, 2001b). encephalitis-causing Nipah virus. The rhaboviruses include
vesicular stomatitis (VSV) and rabies viruses. This review
summarizes progress in the use of siRNA in the phenotypic
knockdown of representative NNR viral genes, technical
∗ Tel.: +1-251-460-6860; fax: +1-251-460-6127. considerations, and the extension of the technology to
E-mail address: sbarik@jaguar1.usouthal.edu (S. Barik). studies of host–virus interaction.

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.012
28 S. Barik / Virus Research 102 (2004) 27–35

2. Nonsegmented negative-strand RNA (NNR) viruses size in different NNR viruses, and is intimately wrapped
with the nucleocapsid protein N. The resultant nucleoprotein
2.1. An overview of the NNR viral life cycle: transcription complex, termed N-RNA template, associates itself with the
versus replication RdRP subunits, primarily L and P, and is finally packaged
inside the structural shell of the virion, mainly made up of
Historically, the early siRNA targets were all cellular mR- viral glycoproteins. The nucleocapsid inside the virion is,
NAs, transcribed in the nucleus by DNA-dependent RNA therefore, transcription-ready, and thus, primary viral tran-
polymerase (Cogoni and Macino, 2000). In contrast, the scription begins shortly after infection. A notable physical
vast majority of viral RNA genomes are transcribed ex- property of the N-RNA template is its extreme resistance to
clusively in the cytoplasm, catalyzed by virally encoded RNases, suggesting that the tightly bound N protein offers
RNA-dependent RNA polymerases (RdRP) (Barik et al., protection. Only this N-RNA complex serves as the biolog-
1990; O’Reilly and Kao, 1998; Ahlquist, 2002). Fig. 1 sum- ical template for the viral RdRP, which would not recognize
marizes the generalized scheme of transcription and replica- pure deproteinized genomic RNA.
tion of the NNR viruses; a more comprehensive treatise can The RdRP functions in two operationally distinct modes:
be found in earlier reviews (Banerjee et al., 1991; Banerjee transcription and replication (Banerjee and Barik, 1992). In
and Barik, 1992). A scholarly minireview by Ahlquist (2002) the transcription mode, it copies the negative-sense genome
has recently discussed these steps with special reference to to produce mRNAs corresponding to each individual gene
siRNA. The minimal RdRP of NNR viruses consists of the by a stop–restart mechanism (Barr et al., 2002). The mR-
large viral protein L and the accessory phosphoprotein P NAs have the features of standard eukaryotic mRNAs, i.e.,
(Banerjee and Barik, 1992; Barik et al., 1995). Depending they are 5 -capped and 3 -polyadenylated (Bisaillon and
on the particular virus, optimal transcription may require ad- Lemay, 1997; Barik, 1993). The transcription process ex-
ditional viral proteins, cytoskeletal proteins (such as tubulin hibits “polarity,” such that the genes most proximal to the
in VSV and actin in RSV) (Moyer et al., 1986; Burke et al., promoter (3 end of the negative-strand genome) are tran-
1998), and possibly additional cellular proteins (such as pro- scribed most abundantly (Barik, 1992). This is also reflected
filin in RSV) (Burke et al., 2000; Bitko et al., 2003). The in the relative abundance of the corresponding proteins, and
viral genome is a single, linear, negative-sense (anti-mRNA fits the function of the different proteins while allowing
sense) RNA molecule, ranging roughly from 10 to 15 kb in transcriptional economy. For example, the N gene, usually

Fig. 1. NNR viral life cycle (see text for details). A hypothetical NNR virus genome is shown for the purposes of illustration only. In specific viruses
some of these genes may be absent and others present. For example, VSV does not contain F, and RSV contains additional genes such as SH, HPIV-3
contains HN, etc. Note that in contrast to the mRNA, the full-length genomic and antigenomic RNA (− and + sense, respectively) are neither capped
nor polyadenylated, and are wrapped with N protein and hence, are not accessible to siRNA.
S. Barik / Virus Research 102 (2004) 27–35 29

being the closest to the promoter, produces the most mRNA


and protein, which befits the stoichiometric need of N pro-
tein to wrap the large full-length genomic and antigenomic
(see below) RNA. The L gene, coding for the large subunit
of the RdRP, is usually the farthest (i.e., at the 5 end of the
negative-strand genome), and produces the least amounts
of mRNA. As the RdRP is only needed in catalytic quan-
tities, small amounts of the L mRNA suffice the functional
need.
The NNR viral mRNAs are translated by the host’s trans-
lation machinery. Since the virion particles do not pack-
age any mRNA (or any positive-sense RNA, for that mat-
ter), viral transcription is an obligatory pre-requisite to viral
protein synthesis. Transcription and translation eventually
generate more of the RdRP, boosting transcription further
(Fig. 1). Once sufficient N protein is synthesized, it binds to
the newly transcribed nascent RNA, and this somehow pro-
vokes the leading RdRP to switch from the transcription to
the replication mode, such that the RdRP now ignores the
intergenic stop signals, producing full-length complemen-
tary positive strand (mRNA-sense) RNA, called antigenome.
Like the genome, the antigenome is also encapsidated with
the N protein. In contrast to the genome, it is not tran- Fig. 2. Inhibition of RSV gene expression by siRNA (see Bitko and
Barik, 2001b for details). A549 cells were transfected with indicated
scribed, but simply serves as a replication template to pro-
concentrations of anti-P siRNA, and infected with RSV 16 h thereafter.
duce more of the full-length negative-strand genome. At the Lane ‘U’ indicates control, uninfected cells. Top: Immunoblot (Western)
end of the intracellular life cycle of the virus, the encapsi- of total cell extracts was performed with either rabbit anti-P or mono-
dated negative-strand genomes, along with other structural clonal anti-actin antibody (Boehringer-Mannheim), as indicated. Bottom:
proteins are packaged into virion particles. Viral protein synthesis in siRNA-treated cells was measured by standard
immunoprecipitation procedures. Cells were metabolically labeled with
All siRNAs used against NNR viruses in published pa- 35 S-(methionine plus cysteine) at 18 h post-infection, followed by im-
pers so far have been double-stranded and synthetic, and de- munoprecipitation with anti-RSV antibody, and analysis of the labeled
signed according to the recommendations of Elbashir et al. proteins by SDS–PAGE and autoradiography. ‘La’ represents treatment
(2001); this issue and its exceptions are further discussed with 100 nM anti-lamin A/C siRNA. The different viral protein bands are
in detail in Section 4.1. In most experiments, the cells were so indicated.
pre-transfected with the antiviral siRNA 10–24 h before in-
fection. 5 days, which was the longest time point in the experiment.
In fact, siRNA-treated infected cells were virtually indistin-
2.2. Silencing of viral RdRP guishable from uninfected ones (Bitko and Barik, 2001b).
On a clinical note, one can envisage two classes of an-
Based on the foregoing, silencing of the minimal essen- tivirals that would inhibit infection at even earlier steps, and
tial RdRP subunits, namely L and P, should lead to a nearly both have pros and cons.
total loss of all RNA synthesis, and this is what we have
observed in all three test viruses (RSV, VSV, and HPIV-3). (i) Inhibitors of virus adsorption: Recently, a novel aro-
RSV is shown as an example in Fig. 2, in which the ablation matic compound, dubbed RFI-641, has been shown to
of P protein by siRNA led to a drastic inhibition of overall inhibit RSV infection (Huntley et al., 2002) by blocking
viral gene expression (Bitko and Barik, 2001b). It is logi- the RSV glycoprotein, F, essential for fusion of the viral
cal to assume that primary transcription, which is catalyzed envelope with the host cell membrane (see Section 2.3).
by preformed viral RdRP, would be unaffected by siRNAs Complete exclusion of the virus, however, may affect
targeting the RdRP. However, in routine infections at low natural immunity, which is still the best form of pro-
m.o.i., primary transcription produces only small amounts tection against many NNR viruses, especially RSV and
of viral mRNA, and it is the secondary transcription that HPIV-3 (Crowe and Williams, 2003).
contributes to the bulk of viral mRNA and protein synthesis. (ii) Direct inhibitors of RdRP activity: The unique sequence
Thus, abolition of new RdRP synthesis by siRNA results in a features of NNR viral L and P proteins (Barik et al.,
strong and sustained antiviral effect at an early intracellular 1990; Delarue et al., 1990; Banerjee and Barik, 1992)
step in the NNR viral life cycle while still allowing a small and the absence of similar proteins in mammalian cells
amount of viral transcription and translation to occur. In our make them ideal candidates for structure-based drug de-
experience ex vivo, the antiviral effect persisted for at least sign. However, technical difficulties such as problems of
30 S. Barik / Virus Research 102 (2004) 27–35

recombinant expression and lack of three-dimensional


structures have hindered progress in this area. Thus, the
siRNA is a potential alternative for clinical intervention
of NNR virus infection.

As elaborated in the viral life cycle (Section 2.1; Fig. 1),


NNR virus growth generates both the negative-strand
genome and the positive-strand antigenome in the infected
cell. However, it has been shown that they are resistant to
siRNA (Bitko and Barik, 2001b). The most likely expla-
nation is that their obligatory encapsidation by N protein
protects them from annealing to the siRNA or to each other
(Fig. 1), just as it protects them from RNase action in vitro.
In any case, it is clear that the NNR viral mRNAs, and not
the genomes, are viable targets for RNA interference.

2.3. Silencing of viral accessory genes

There are a number of NNR viral proteins that are not


involved in viral RNA or protein synthesis, but take part
in important pre- or post-transcriptional aspects of the vi-
ral life cycle. For example, paramyxoviruses initiate infec-
tion by attaching to cell surface receptors and fusing vi-
Fig. 3. Inhibition of HPIV-3 syncytium formation by siRNA. A549 cell
ral and cell membranes. Viral attachment proteins, such
monolayers were transfected with 50 nM of the following double-stranded
as hemagglutinin-neuraminidase (HN) or glycoprotein (G) siRNA against the F or HN sequence of HPIV-3 (GenBank accession
bind receptors, whereas fusion (F) proteins direct membrane numbers X05303 and Z26523, respectively): F: CCAUGAAAUGGA-
fusion (Dutch et al., 2000). The fusion protein, F, is gen- GAGCUGU; HN: CUCAGACUUGGUACCUGAC. Only the N19 portion
erally essential for cell fusion (formation of syncytia), al- (Elbashir et al., 2001) of the sense strand of the siRNA is shown. Trans-
fection and subsequent virus infection was carried out as in Fig. 2, and
though the relative importance of the attachment proteins
phase-contrast pictures were taken as described (Bitko and Barik, 2001b;
in fusion is under active study. Recombinant expression Bitko et al., 2003). Top: Note the large fused mass of cells (syncytium) at
has sometimes produced variable results. For instance, in the center of the panel labeled “HPIV-3,” and prevention of such fusion
transient transfection experiments co-expression of all three by ds-siRNA (100 nM) targeting F and HN. Bottom: Immunoblot shows
RSV glycoproteins—F, G, and SH—was required for opti- loss of HN upon siRNA-treatment.
mal fusion (Heminway et al., 1994a,b), whereas recombi-
nant RSV lacking SH formed fused cells as efficiently as raminidase activity, but is obviously capable of forming syn-
the wild type (Bukreyev et al., 1997). It is possible that the cytia. Thus, we retested the role of the HN protein in syn-
exact outcome is dictated by the relative amounts of the re- cytium formation in wild type HPIV-3-infected cells using
combinant proteins and/or the expression system used. The siRNA. As shown in Fig. 3, abrogation of either F or HN by
siRNA approach, in contrast, obviates recombinant expres- the respective siRNA resulted in unfused cells, confirming
sion, and allows phenotypic analysis in a virus-infected cell the need for both proteins for syncytium formation.
by removing one protein while leaving all others intact. This Although the vast majority of NNR viral mRNAs pro-
has been illustrated by individually abrogating the F or the duce a single polypeptide, there are a few examples of mul-
SH protein in RSV-infected cells by siRNA. Knockdown of tiple initiations. In a recent study, VSV matrix gene (M)
F resulted in complete loss of syncytium (Bitko and Barik, mRNA was shown to produce three proteins M1, M2, and
2001b), without affecting G or SH synthesis (unpublished M3 (Jayakar and Whitt, 2002). M1 was the longest polypep-
results). Reciprocally, abrogation of SH by siRNA had no tide initiating at the most 5 -proximal ATG; M2 and M3
effect on F or G, and also had no appreciable effect on syn- were shorter, and produced by initiation at internal ATG’s in
cytium (unpublished results). Together, these results validate the same reading frame. While the exact roles of these dif-
an essential role of F and the dispensability of SH in syn- ferent proteins remain unclear, the M protein(s) have been
cytium formation by RSV. shown to inhibit host gene expression (Jayakar and Whitt,
In another example, a number of studies using reverse ge- 2002; Kopecky and Lyles, 2003). Recent studies have sug-
netics and variant virus strains have shown that both the fu- gested that the M gene is also important for apoptosis of the
sion (F) protein and hemagglutinin-neuraminidase (HN) are host cell, manifested by the characteristic rounding and de-
essential for syncytium in HPIV-3 infected cells (Heminway tachment of VSV-infected cells (Jayakar and Whitt, 2002;
et al., 1994a,b; Dutch et al., 2000; Porotto et al., 2003). Cu- Kopecky and Lyles, 2003). In an attempt to determine which
riously, RSV encodes neither a hemagglutinin nor a neu- product of the M gene mRNA is involved in apoptosis and
S. Barik / Virus Research 102 (2004) 27–35 31

(a) Virus-resistant cells: It is possible to generate, by con-


stitutive or inducible expression of a transgene siRNA,
a cell line that would resist virus infection. Specialized
vectors have been constructed that drive siRNA expres-
sion from RNase III-based tRNA or snRNA (mainly
U6) gene promoter (Yu et al., 2002; Kawasaki and
Taira, 2003), and are marketed by commercial vendors.
This would establish a form of “intracellular immunity”
(Carmichael, 2002) that can be extended to produce
transgenic virus-resistant animals.
(b) Complementation experiments: As a rule, siRNAs lead
to the loss of the protein rather than its mutational al-
teration, and thus, cannot be used for structure–function
analysis. However, the phenotypic viral mutant can be
complemented by transient transfection of the corre-
sponding cDNA clone. The latter can be subjected to
site-directed mutagenesis, and the mutants that fail to
complement would define the amino acid residues im-
portant for function.

3. Cellular genes important in NNR virus–host


interaction

3.1. Profilin and RSV: a case in point

Silencing of cellular genes by siRNA can be used to ad-


Fig. 4. Inhibition of VSV cytopathic effect by ds-siRNA. Monolayers of dress the role of such genes in host–virus interaction. We
HEp-2 cells were transfected with double-stranded siRNA (80 nM) corre- have recently applied this approach to determine the role
sponding to the following sequence of the VSV matrix (M) mRNA (Indi- of profilin in the RSV life cycle (Bitko et al., 2003). Previ-
ana serotype; GenBank accession number X04452): AGGUAAGAAAU-
ous in vitro studies had shown that profilin stimulates RSV
CUAAGAAA. Subsequent infection with VSV, phase-contrast microgra-
phy, and immunoblot were done as described in Fig. 3. The antibody transcription, but is not absolutely necessary for virus repli-
was made against a C-terminal peptide common to all three M proteins: cation (Burke et al., 2000). Knockdown of profilin showed
KFSDFREKALMFGLIVEEE (Jayakar and Whitt, 2002). Top: Note that multiple effects: (a) it had a minor effect on viral transcrip-
siRNA-treated cells look as if uninfected. Bottom: Immunoblot shows spe- tion (and hence translation), supporting in vitro results; (b)
cific loss of the three M proteins, but not P, upon treatment with anti-M
it strongly inhibited virion morphogenesis such that progeny
siRNA.
virions were not produced; (c) it inhibited formation of actin
stress fibers in cells where such fibers form in response to
cell-rounding, we targeted the region of the mRNA that is RSV infection (e.g., HEp-2 and L4 cells); and (d) it abro-
unique to M1 and upstream of M2 and M3. Interestingly, gated fusion in all cells tested. These results establish an
however, the siRNA abrogated the expression of all three essential role of profilin in the post-gene expression steps of
proteins (Fig. 4). Cell detachment was concomitantly inhib- RSV growth, and in the regulation of the actin cytoskeleton
ited, demonstrating the role of the M gene in the process, and the cell membrane in the infected cell.
although we could not identify the exact protein involved.
The reason to explain the loss of all three proteins must 3.2. Future directions
await further studies, but the most likely possibilities are:
(a) loss of one part of the mRNA made the rest susceptible The ability to quickly and reversibly knock down inter-
to degradation by RNases, and (b) loss of the 5 -cap and active cellular and viral genes by siRNA will allow single
lack of internal ribosome entry inhibited expression of the and combinatorial analysis of various interacting host–virus
downstream M2 and M3 ORFs. pathways that would have been otherwise impossible. Two
examples are considered here.
2.4. Future directions (a) Genes dispensable for cellular viability but essential
for NNR virus: Although the loss of profilin was toler-
A variety of creative experiments can be designed by tar- ated in cultured cells (Bitko et al., 2003), profilin gene
geting NNR viral genes with siRNA. A few suggestions are knockout led to embryonic lethality, suggesting its es-
offered here. sential role in early development. In a seminal study
32 S. Barik / Virus Research 102 (2004) 27–35

(Harborth et al., 2001) selected nuclear and membrane


proteins were targeted by siRNA to test their essential
need in a number of cell lines in culture. Two cytoskele-
ton/membrane regulatory proteins that were found to be
nonessential are Ena/VASP and zyxin. The validity of
this approach is borne out by the fact that Ena/VASP
and zyxin knockout mice are viable (Aszódi et al., 1999;
Hoffman et al., 2003). Thus, it would be exciting to de-
termine if such (relatively) nonessential cellular genes
are important in NNR viral life cycles. If they are, it
would not only enrich our fundamental knowledge of
host–virus interaction, but may lead to antivirals that
would specifically block such interactions without gross
toxicity to the host. Fig. 5. Relative efficacy of single-stranded and double-stranded siRNAs.
(b) Specific viral triggers of cellular pathways: NNR viruses The sequence of the siRNA, designed against RSV P, has been pub-
as a class activate a variety of cellular genes and sig- lished earlier (Bitko and Barik, 2001b). The symbols represent the fol-
naling pathways. These include cellular transcription lowing: (䊊), ds-siRNA; (䊐) ss-antisense siRNA; () same ss-siRNA but
5 -phosphorylated (Martinez et al., 2002). Transfection of siRNA and in-
factors such as NF-␬B (Bitko and Barik, 1998; Helin fection with RSV were carried out as in Fig. 2. Residual P protein was
et al., 2001; Tian et al., 2002), protein kinases (Monick estimated by immunoblot, and plotted as percent of the untransfected
et al., 2001; Pazdrak et al., 2002), and apoptosis (Bitko control against the negative logarithm of siRNA concentration in molarity
and Barik, 2001a; Esolen et al., 1995; O’Donnell et al., (M). Thus, 9 on the x-axis means 1 nM siRNA, 8 means 10 nM, and so
1999; Cristina et al., 2001). The specific viral proteins forth. The dotted line marks 50% loss.
(single or multiple) that trigger these pathways can be
identified by siRNA knockdown. 4.1. Double-stranded versus single-stranded siRNA

Although the double-stranded siRNA is used with success


4. The technique in cell culture, it is the antisense RNA strand that binds to
the target mRNA and forms the silencing complex, RISC. A
In this section, we first describe the basic procedure few recent studies have indeed documented the effectiveness
that we routinely use. To design siRNA, we generally fol- of single-stranded antisense RNA (ss-siRNA) (Tijsterman
low the original recommendations (Elbashir et al., 2001). et al., 2002; Martinez et al., 2002). In our hands, ss-siRNA
Interested readers can find a detailed description and al- showed decent activity against NNR viruses, although it was
ternate siRNA designs in the Tuschl lab web page (http:// required in larger quantities to achieve a quantitatively sim-
www.rockefeller.edu/labheads/tuschl/sirna.html). Briefly, ilar effect (Fig. 5). Using RSV P mRNA as the experimen-
we look for a AA(N)19 TT sequence in the target mRNA tal target, the IC50 (amount of RNA causing 50% reduc-
that is not too close to the ATG. This RNA and its tion of P protein) of ds-siRNA and ss-siRNA were about 10
complementary strand is chemically synthesized and pur- and 75 nM, respectively. When the 5 end of the antisense
chased from commercial vendors (such as Dharmacon; ss-siRNA was phosphorylated, the IC50 slightly improved to
http://www.dharmacon.com). For transfection, various 50 nM (Fig. 5). As suggested earlier (Martinez et al., 2002),
amounts of the deprotected, desalted, double-stranded we presume that this reflects the relative stability of the RNA
siRNA (ds-siRNA) are transfected using OligofectAMINE species in the order: dsRNA > 5 -phospho-ssRNA > ss-RNA.
Reagent (Life Technologies) in OPTIMEM I (Life Tech-
nologies) (Bitko and Barik, 2001b). Care is taken to make 4.2. Mismatched siRNA and position effect: pros and cons
a homogeneous mixture of the transfection reagent and
RNA by heavy vortexing before addition to the culture. We As a rule, the siRNA is extremely stringent in its speci-
have recently switched to a newer reagent, TransIT-TKO® , ficity such that a single nucleotide mismatch may abrogate
marketed by Mirus (http://www.genetransfer.com), which its function (Elbashir et al., 2001; Bitko and Barik, 2001b;
does not require the use of serum-free media. In our expe- Chi et al., 2003; Semizarov et al., 2003). A few recent re-
rience, optimization of the DNA to TKO ratio is advised ports have challenged this stringency (Hamada et al., 2002;
as excessive TKO may cause injury to some cell lines. The Jackson et al., 2003). In an extreme example, direct silenc-
virus is generally added 6–8 h after siRNA transfection if a ing of non-targeted genes containing as few as 11 contiguous
viral mRNA is targeted and 14–18 h after transfection if a nucleotides of identity to the siRNA was observed (Jackson
cellular mRNA is targeted. et al., 2003). In our experience with NNR viruses, the toler-
In what follows, we describe some recent variations and ance to mismatch varies from one position to another on the
potential considerations in planning a siRNA experiment, same NNR viral mRNA. In other words, a single nucleotide
particularly in NNR viruses. mismatch at one site of the mRNA may prevent siRNA ac-
S. Barik / Virus Research 102 (2004) 27–35 33

tion (Bitko and Barik, 2001b), whereas at another site on mutagenesis requires a DNA template, direct mutational
the same mRNA, one mismatch may be largely tolerated analysis of selected NNR viral genes is also not possible.
(unpublished results). The reason for the sequence effect is These obstacles have been partially circumvented by the use
currently unknown, but mismatch tolerance has raised is- of cloned viral cDNA (Whelan et al., 1995; Lawson et al.,
sues of specificity. However, this is probably not a major 1995; Pekosz et al., 1999; Marriott and Easton, 1999). De-
concern with NNR viruses, since the viral genes have lit- spite its revolutionary effect on NNR viral reverse genetics,
tle or no homology with one another or with cellular genes. however, the cDNA-based strategy is not without limita-
On the contrary, expansion of the target repertoire of the tions. First, expression of recombinant viral genomic RNA
siRNA may enable it to silence multiple mutational variants and all the viral proteins in the correct ratio that reflects
of a given NNR virus that commonly occur due to the high polarity remains a grueling task. As mentioned before, the
mutation rate of the RNA genome (Domingo and Holland, 10–15 kb long RNA genome of the NNR viruses must have
1997). In any case, when designing a siRNA, a BLAST specific sequences at the 5 and 3 termini for transcription
search against the non-redundant nucleotide databases of the and replication and must be properly encapsidated by the
GenBank should be routinely done to verify specificity. nucleocapsid protein (N) in order to be recognized by the
Recent studies have also shown that the siRNA effect viral RdRP. Second, many NNR viral genomes and proteins
can be strikingly position-dependent, i.e., siRNAs directed are currently expressed from vaccinia-based cDNA clones,
against different regions of the same mRNA may exhibit generally requiring super-infection by vaccinia virus, and
a wide range of silencing efficiency (Holen et al., 2002). vaccinia itself modulates multiple cellular entities, includ-
Again, the rules governing the position effect remain un- ing MAP kinases and the actin cytoskeleton (de Magalhaes
known, but we have also observed this in silencing the NNR et al., 2001). It is, therefore, virtually impossible to study
virus. the interaction between cellular signaling pathways and
Based on these considerations, it may be wise to design NNR viruses in cells that are super-infected by vaccinia.
multiple siRNAs against a target mRNA using the prescribed Third, mutations in the recombinant DNA are permanent,
design guidelines, test all of them over a range of concen- i.e., the mutational phenotype cannot be regulated at differ-
trations, and then select the most effective one. ent times in infection. For example, if a viral gene prod-
uct has dual essential functions—one early, and one late
4.3. Abundant mRNAs and saturation of the silencing in infection—the late function will remain undiscovered,
machinery since the mutant virus will never proceed beyond the early
stage.
In principle, multiple siRNAs can be transfected or ex- In conclusion, properly designed siRNA can be a quicker
pressed in the same cell to abrogate the respective NNR viral and simpler alternative to cDNA-based reverse genetics of
proteins simultaneously. In our experience, this works, but NNR viruses. Its ability to simultaneously knock down viral
seems to have limits (unpublished result). Silencing of an and cellular gene products offers particular advantages in
abundant NNR viral mRNA (for example, that of a promoter the dissection of host–virus signaling pathways. We predict
proximal gene such as N) often leads to reduced silencing that the siRNA strategy will lead to major advances in these
of a promoter distal gene (such as L). The mechanism of areas in the immediate future.
this is being investigated. Nonetheless, quantitative analy-
sis has shown that the siRNA effect may reach a plateau
with increasing siRNA concentration (Kamath et al., 2001; Acknowledgements
Bitko and Barik, 2001b), suggesting that the RNAi-induced
silencing complexes (RISC) of the cell are saturable. Thus, The siRNA-related research in the author’s laboratory was
an abundant viral mRNA may sequester most of the cellular supported in part by NIH grants AI049682 from the National
RISC, leaving little for the less abundant ones. Conceivably, Institute of Allergy and Infectious Diseases and EY013826
such competition can also occur between cellular and vi- from the National Eye Institute. A computer program to
ral mRNAs, thus potentially limiting host–virus interaction find the AA(N)19 TT sequences was kindly written and pro-
studies. vided by Titus Barik (http://www.barikautomation.com).
Apology is offered to many whose original research had
advanced the field, but could not be cited due to space con-
5. Conclusions straints. Nevertheless, the majority of the research articles
are cross-referenced in the recent reviews cited here.
Traditionally, structure–function analyses of RNA
genomes, including those of NNR viruses, have made use
of fortuitous mutants, natural variants, or chemically mu- References
tagenized stocks (e.g., Crowe et al., 1994). Such mutations
are unpredictable and must be mapped by difficult com- Ahlquist, P., 2002. RNA-dependent RNA polymerases, viruses, and RNA
plementation analyses or sequencing. Because site-directed silencing. Science 296, 1270–1273.
34 S. Barik / Virus Research 102 (2004) 27–35

Aszódi, A., Pfeifer, A., Ahmad, M., Glauner, M., Zhou, X.H., Ny, L., mutants derived from a partially attenuated cold-passaged respiratory
Andersson, K.E., Kehrel, B., Offermanns, S., Fässler, R., 1999. The syncytial virus mutant by introduction of additional attenuating mu-
vasodilator-stimulated phosphoprotein (VASP) is involved in cGMP- tations during chemical mutagenesis. Vaccine 12, 691–699.
and cAMP-mediated inhibition of agonist-induced platelet aggregation, Crowe Jr., J.E., Williams, J.V., 2003. Immunology of viral respiratory
but is dispensable for smooth muscle function. EMBO J. 18, 37–48. tract infection in infancy. Paediatr. Respir. Rev. 4, 112–119.
Banerjee, A.K., Barik, S., 1992. Gene expression of vesicular stomatitis Delarue, M., Poch, O., Tordo, N., Moras, D., Argos, P., 1990. An attempt
virus genome RNA. Virology 188, 417–428. to unify the structure of polymerases. Protein Eng. 3, 461–467.
Banerjee, A.K., Barik, S., De, B.P., 1991. Gene expression of nonseg- de Magalhaes, J.C., Andrade, A.A., Silva, P.N., Sousa, L.P., Ropert, C.,
mented negative-strand RNA viruses. Pharmacol. Ther. 51, 47–70. Ferreira, P.C., Kroon, E.G., Gazzinelli, R.T., Bonjardim, C.A., 2001. A
Barik, S., 1992. Transcription of human respiratory syncytial virus genome mitogenic signal triggered at an early stage of vaccinia virus infection:
RNA in vitro: requirement of cellular factor(s). J. Virol. 66, 6813– implication of MEK/ERK and protein kinase A in virus multiplication.
6818. J. Biol. Chem. 276, 38353–38360.
Barik, S., 1993. The structure of the 5 terminal cap of the respiratory Domingo, E., Holland, J.J., 1997. RNA virus mutations and fitness for
syncytial virus mRNA. J. Gen. Virol. 74, 485–490. survival. Annu. Rev. Microbiol. 51, 151–178.
Barik, S., McLean, T., Dupuy, L.C., 1995. Phosphorylation of Ser232 Dutch, R.E., Jardetzky, T.S., Lamb, R.A., 2000. Virus membrane fusion
directly regulates the transcriptional activity of the P protein of human proteins: biological machines that undergo a metamorphosis. Biosci.
respiratory syncytial virus: phosphorylation of Ser237 may play an Rep. 20, 597–612.
accessory role. Virology 213, 405–412. Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl,
Barik, S., Rud, E.W., Luk, D., Banerjee, A.K., Kang, C.Y., 1990. Nu- T., 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference
cleotide sequence analysis of the L gene of vesicular stomatitis virus in cultured mammalian cells. Nature 411, 494–498.
(New Jersey serotype): identification of conserved domains in L pro- Esolen, L.M., Park, S.W., Hardwick, J.M., Griffin, D.E., 1995. Apoptosis
teins of nonsegmented negative-strand RNA viruses. Virology 175, as a cause of death in measles virus-infected cells. J. Virol. 69, 3955–
332–337. 3958.
Barr, J.N., Whelan, S.P., Wertz, G.W., 2002. Transcriptional control of Hamada, M., Ohtsuka, T., Kawaida, R., Koizumi, M., Morita, K., Fu-
the RNA-dependent RNA polymerase of vesicular stomatitis virus. rukawa, H., Imanishi, T., Miyagishi, M., Taira, K., 2002. Effects on
Biochim. Biophys. Acta 1577, 337–353. RNA interference in gene expression (RNAi) in cultured mammalian
Bisaillon, M., Lemay, G., 1997. Viral and cellular enzymes involved in cells of mismatches and the introduction of chemical modifications at
synthesis of mRNA cap structure. Virology 236, 1–7. the 3 -ends of siRNAs. Antisense Nucleic Acid Drug Dev. 12, 301–
Bitko, V., Barik, S., 1998. Persistent activation of RelA by respiratory 309.
syncytial virus involves protein kinase C, underphosphorylated Ikap- Hannon, G.J., 2002. RNA interference. Nature 418, 244–251.
paBbeta, and sequestration of protein phosphatase 2A by the viral Harborth, J., Elbashir, S.M., Bechert, K., Tuschl, T., Weber, K., 2001.
phosphoprotein. J. Virol. 72, 5610–5618. Identification of essential genes in cultured mammalian cells using
Bitko, V., Barik, S., 2001a. An endoplasmic reticulum-specific small interfering RNAs. J. Cell. Sci. 114, 4557–4565.
stress-activated caspase (caspase-12) is implicated in the apoptosis of Helin, E., Vainionpaa, R., Hyypia, T., Julkunen, I., Matikainen, S., 2001.
A549 epithelial cells by respiratory syncytial virus. J. Cell. Biochem. Measles virus activates NF-kappa B and STAT transcription factors and
80, 441–454. production of IFN-alpha/beta and IL-6 in the human lung epithelial
Bitko, V., Barik, S., 2001b. Phenotypic silencing of cytoplasmic genes cell line A549. Virology 290, 1–10.
using sequence-specific double-stranded short interfering RNA and its Heminway, B.R., Yu, Y., Galinski, M.S., 1994a. Paramyxovirus me-
application in the reverse genetics of wild type negative-strand RNA diated cell fusion requires co-expression of both the fusion and
viruses. BMC Microbiol. 1, 34. hemagglutinin-neuraminidase glycoproteins. Virus Res. 31, 1–16.
Bitko, V., Oldenburg, A., Garmon, N.E., Barik, S., 2003. Profilin is re- Heminway, B.R., Yu, Y., Tanaka, Y., Perrine, K.G., Gustafson, E., Bern-
quired for viral morphogenesis, syncytium formation, and cell-specific stein, J.M., Galinski, M.S., 1994b. Analysis of respiratory syncytial
stress fiber induction by respiratory syncytial virus. BMC Microbiol. virus F, G, and SH proteins in cell fusion. Virology 200, 801–805.
3, 9. Hoffman, L.M., Nix, D.A., Benson, B., Boot-Hanford, R., Gustafsson,
Bukreyev, A., Whitehead, S.S., Murphy, B.R., Collins, P.L., 1997. Recom- E., Jamora, C., Menzies, A.S., Goh, K.L., Jensen, C.C., Gertler, F.B.,
binant respiratory syncytial virus from which the entire SH gene has Fuchs, E., Fassler, R., Beckerle, M.C., 2003. Targeted disruption of
been deleted grows efficiently in cell culture and exhibits site-specific the murine zyxin gene. Mol. Cell. Biol. 23, 70–79.
attenuation in the respiratory tract of the mouse. J. Virol. 71, 8973– Holen, T., Amarzguioui, M., Wiiger, M.T., Babaie, E., Prydz, H., 2002.
8982. Positional effects of short interfering RNAs targeting the human co-
Burke, E., Dupuy, L., Wall, C., Barik, S., 1998. Role of cellular actin in agulation trigger Tissue Factor. Nucleic Acids Res. 30, 1757–1766.
the gene expression and morphogenesis of human respiratory syncytial Huntley, C.C., Weiss, W.J., Gazumyan, A., Buklan, A., Feld, B., Hu,
virus. Virology 252, 137–148. W., Jones, T.R., Murphy, T., Nikitenko, A.A., O’Hara, B., Prince,
Burke, E., Mahoney, N.M., Almo, S.C., Barik, S., 2000. Profilin is required G., Quartuccio, S., Raifeld, Y.E., Wyde, P., O’Connell, J.F., 2002.
for optimal actin-dependent transcription of respiratory syncytial virus RFI-641, a potent respiratory syncytial virus inhibitor. Antimicrob.
genome RNA. J. Virol. 74, 669–675. Agents Chemother. 46, 841–847.
Carmichael, G.G., 2002. Medicine: silencing viruses with RNA. Nature Jackson, A.L., Bartz, S.R., Schelter, J., Kobayashi, S.V., Burchard, J., Mao,
418, 379–380. M., Li, B., Cavet, G., Linsley, P.S., 2003. Expression profiling reveals
Chi, J.T., Chang, H.Y., Wang, N.N., Chang, D.S., Dunphy, N., Brown, off-target gene regulation by RNAi. Nat. Biotechnol. 21, 635–637.
P.O., 2003. Genomewide view of gene silencing by small interfering Jayakar, H.R., Whitt, M.A., 2002. Identification of two additional trans-
RNAs. Proc. Natl. Acad. Sci. U.S.A. 100, 6343–6346. lation products from the matrix (M) gene that contribute to vesicular
Cogoni, C., Macino, G., 2000. Post-transcriptional gene silencing across stomatitis virus cytopathology. J. Virol. 76, 8011–8018.
kingdoms. Curr. Opin. Genet. Dev. 10, 638–643. Kamath, R.S., Martinez-Campos, M., Zipperlen, P., Fraser, A.G., Ahringer,
Cristina, J., Yunus, A.S., Rockemann, D.D., Samal, S.K., 2001. Bovine J., 2001. Effectiveness of specific RNA-mediated interference through
respiratory syncytial virus can induce apoptosis in MDBK cultured ingested double-stranded RNA in Caenorhabditis elegans. Genome
cells. Vet. Microbiol. 83, 317–320. Biol. 2, RESEARCH0002.
Crowe Jr., J.E., Bui, P.T., London, W.T., Davis, A.R., Hung, P.P., Chanock, Kawasaki, H., Taira, K., 2003. Short hairpin type of dsRNAs that are
R.M., Murphy, B.R., 1994. Satisfactorily attenuated and protective controlled by tRNA(Val) promoter significantly induce RNAi-mediated
S. Barik / Virus Research 102 (2004) 27–35 35

gene silencing in the cytoplasm of human cells. Nucleic Acids Res. Pazdrak, K., Olszewska-Pazdrak, B., Liu, T., Takizawa, R., Brasier, A.R.,
31, 700–707. Garofalo, R.P., Casola, A., 2002. MAPK activation is involved in post-
Kopecky, S.A., Lyles, D.S., 2003. The cell-rounding activity of the vesic- transcriptional regulation of RSV-induced RANTES gene expression.
ular stomatitis virus matrix protein is due to the induction of cell Am. J. Physiol. Lung Cell Mol. Physiol. 283, L364–L372.
death. J. Virol. 77, 5524–5528. Pekosz, A., He, B., Lamb, R.A., 1999. Reverse genetics of negative-strand
Lawson, N., Stillman, E., Whitt, M., Rose, J., 1995. Recombinant vesicular RNA viruses: closing the circle. Proc. Natl. Acad. Sci. U.S.A. 96,
stomatitis viruses from DNA. Proc. Natl. Acad. Sci. U.S.A. 92, 4477– 8804–8806.
4481. Pickford, A.S., Cogoni, C., 2003. RNA-mediated gene silencing. Cell.
Marriott, A.C., Easton, A.J., 1999. Reverse genetics of the Paramyxoviri- Mol. Life Sci. 60, 871–882.
dae. Adv. Virus Res. 53, 321–340. Porotto, M., Murrell, M., Greengard, O., Moscona, A., 2003. Trig-
Martinez, J., Patkaniowska, A., Urlaub, H., Luhrmann, R., Tuschl, T., gering of human parainfluenza virus 3 fusion protein (F) by the
2002. Single-stranded antisense siRNAs guide target RNA cleavage hemagglutinin-neuraminidase (HN) protein: an HN mutation dimin-
in RNAi. Cell 110, 563–574. ishes the rate of F activation and fusion. J. Virol. 77, 3647–3654.
Monick, M., Staber, J., Thomas, K., Hunninghake, G., 2001. Respiratory Semizarov, D., Frost, L., Sarthy, A., Kroeger, P., Halbert, D.N., Fesik,
syncytial virus infection results in activation of multiple protein kinase S.W., 2003. Specificity of short interfering RNA determined through
C isoforms leading to activation of mitogen-activated protein kinase. gene expression signatures. Proc. Natl. Acad. Sci. U.S.A. 100, 6347–
J. Immunol. 166, 2681–2687. 6352.
Moyer, S.A., Baker, S.C., Lessard, J.L., 1986. Tubulin: a factor necessary Tian, B., Zhang, Y., Luxon, B.A., Garofalo, R.P., Casola, A., Sinha,
for the synthesis of both Sendai virus and vesicular stomatitis virus M., Brasier, A.R., 2002. Identification of NF-kappaB-dependent gene
RNAs. Proc. Natl. Acad. Sci. U.S.A. 83, 5405–5409. networks in respiratory syncytial virus-infected cells. J. Virol. 76,
O’Donnell, D.R., Milligan, L., Stark, J.M., 1999. Induction of CD95 6800–6814.
(Fas) and apoptosis in respiratory epithelial cell cultures following Tijsterman, M., Ketting, R.F., Okihara, K.L., Sijen, T., Plasterk, R.H.,
respiratory syncytial virus infection. Virology 257, 198–207. 2002. RNA helicase MUT-14-dependent gene silencing triggered in
O’Reilly, E.K., Kao, C.C., 1998. Analysis of RNA-dependent RNA poly- C. elegans by short antisense RNAs. Science 295, 694–697.
merase structure and function as guided by known polymerase struc- Whelan, S.P., Ball, L.A., Barr, J.N., Wertz, G.T., 1995. Efficient recovery
tures and computer predictions of secondary structure. Virology 252, of infectious vesicular stomatitis virus entirely from cDNA clones.
287–303. Proc. Natl. Acad. Sci. U.S.A. 92, 8388–8392.
Palese, P., Zheng, H., Engelhardt, O.G., Pleschka, S., Garcia-Sastre, A., Yu, J.Y., DeRuiter, S.L., Turner, D.L., 2002. RNA interference by ex-
1996. Negative-strand RNA viruses: genetic engineering and applica- pression of short-interfering RNAs and hairpin RNAs in mammalian
tions. Proc. Natl. Acad. Sci. U.S.A. 93, 11354–11358. cells. Proc. Natl. Acad. Sci. U.S.A. 99, 6047–6052.
Virus Research 102 (2004) 37–42

Use of siRNAs to prevent and treat influenza virus infection


Qing Ge, Herman N. Eisen, Jianzhu Chen∗
Department of Biology, Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA

Abstract

Influenza virus causes one of the most prevalent infections in humans. In a typical year, 10–20% of the population in the United States
are infected by influenza virus, resulting in up to 40,000 deaths. Current vaccines can prevent illness in approximately 70–80% of healthy
individuals under age 65, but the protection rate is much lower in those most susceptible to infection, namely infants, the elderly, and
immunocompromised individuals. Although four antiviral drugs have been approved in the United States for treatment and/or prophylaxis of
influenza, their use is limited because of concerns about side effects, compliance, and the possible emergence of resistant virus. We found
that short interfering RNAs (siRNAs) specific for conserved regions of the influenza virus genome are potent inhibitors of influenza virus
replication in both cell lines and embryonated chicken eggs. In this review, we discuss the potential value of siRNAs for preventing and
treating influenza virus infections in humans and the challenges that have to be overcome to realize their potential.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Influenza virus; siRNA; Prophylaxis; Therapy; Delivery

1. Influenza virus infection is a significant a template for synthesis of viral proteins. Transcription of
public health problem vRNA also produces complementary RNA (cRNA), which
differs from mRNA by lacking the 5 cap and the 3 poly A
Influenza viruses are enveloped, single-stranded RNA tail and serves as a template for synthesizing more vRNA
viruses of the Orthomyxoviridae family (Lamb and Krug, for new virion production.
2001). They are classified as influenza virus types A, B, and In humans, influenza A virus infection occurs in the upper
C based on differences in their nucleoproteins and matrix respiratory tract and in the lungs (Lamb and Krug, 2001).
proteins. Among the three types, influenza A virus is most Its threat as a pathogen depends on some of the virus’s
pathogenic. Influenza A virus contains eight RNA seg- unique properties. First, the virus is easily spread by aerosol.
ments of negative polarity in its genome (Lamb and Krug, Second, small changes (antigenic drift) in the virus’ prin-
2001). Three of the segments encode the three compo- cipal antigens HA and NA are frequent, enabling the virus
nents of RNA-dependent RNA transcriptase complex (PA, to readily escape protective immunity induced by previous
PB1, and PB2). Three additional RNA segments code for exposure to a different variant of the virus (Webster et al.,
(i) hemagglutinin (HA), a major surface glycoprotein, (ii) 1992). Third, new strains of virus, markedly different anti-
neuraminidase (NA), another major surface glycoprotein, genically, can be generated by reassortment (antigenic shift)
and (iii) nucleocapsid protein (NP), a scaffolding and RNA (Webster et al., 1982). When influenza viruses from two
binding protein. Each of the remaining two RNA segments species, such as human and swine, infect the same cell, hy-
encodes two proteins, either M1 and M2 or NS1 and NS2, brid (reassortant) viruses containing RNA segments from
which function either as viral structural proteins or as non- the two species can emerge (Webby and Webster, 2001).
structural proteins in the viral life cycle. The RNA segments The hybrid virus, with the resulting major antigenic shift,
present in the fully assembled virus particle are referred can be especially virulent, because it is not restrained by
to as virion RNA (vRNA). Transcription of vRNA by the host immunity to previously prevalent strains of the virus.
virus-encoded transcriptase produces mRNA that serves as The 1918 pandemic, during which over 20 million people
were estimated to have died worldwide was thought to be
∗ Corresponding author. Tel.: +1-617-258-6173; caused by a hybrid virus arising from reassortment of swine
fax: +1-617-258-6172. and human viruses (Patterson and Pyle, 1991; Taubenberger
E-mail address: jchen@mit.edu (J. Chen). et al., 2001). The potential for the emergence of a highly

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.013
38 Q. Ge et al. / Virus Research 102 (2004) 37–42

virulent strain of influenza virus, either naturally occurring subtypes. Each year, a decision must be reached before the
or deliberately constructed, was clearly demonstrated by the flu season as to which virus strains to include in the vaccine;
sudden appearance of a lethal form of the virus in Hong it then takes 6 months to produce enough vaccine. In the
Kong in 1997, which killed 6 out of 18 infected individuals face of a rapidly approaching epidemic, 6 months is proba-
(Claas et al., 1998; Yuen et al., 1998). Even infection with bly too long to be acceptable. Third, the expense and poten-
comparatively less virulent influenza strains is estimated tially serious side effects, such as Guillain-Barré syndrome,
to cause up to 40,000 deaths in the US in a typical year associated with annual reformulation of vaccine production
(Thompson et al., 2003). The mortality due to influenza virus and administration make this approach less than optimal.
infection generally is found in infants, adults over 65 years Four drugs have been approved for treating and/or pre-
of age, and immunocompromised individuals (Kemble and venting influenza virus infection in the US (Cheung and
Greenberg, 2003; Thompson et al., 2003). As the population Lieberman, 2002). Amantadine and rimantadine are in-
ages in industrialized countries, influenza virus infection and hibitors of the M2 ion-channel and can prevent viral un-
its associated complications and mortality will likely be- coating (Luscher-Mattli, 2000). Zanamivir and oseltamivir
come an even larger public health problem. Currently, the inhibit NA activity and prevent the release of influenza viri-
economic costs of influenza virus infection are estimated to ons from the cytoplasmic membrane. These drugs usually
be US$ 5 billion per annum in the US alone. reduce the incidence of influenza infection when used pro-
phylactically and reduce the duration of symptoms by 1–2
days when given within 1 or 2 days of infection (Stiver,
2. More effective influenza therapeutics are needed 2003). However, the widespread use of these drugs is lim-
ited by concerns about side effects, patients’ compliance,
The only effective protection from influenza virus infec- and the possible emergence of drug-resistant variants. For
tion now available is vaccination (Palese and Garcia-Sastre, example, in a family-based clinical study, 33% of recipients
2002). In the US, inactivated trivalent influenza virus of amantadine began shedding amantadine-resistant virus
is the commonly used form of the vaccine. Recently, a by day 5 of treatment (Hayden et al., 1991). For these rea-
cold-adapted influenza virus vaccine was approved and is sons, these drugs are prescribed only to highly susceptible
expected to be available in the coming flu season. The individuals.
cold-adapted virus is capable of undergoing limited repli-
cation in the upper respiratory tract, where the tempera-
ture is lower than 37 ◦ C, but not in the lung, where the 3. siRNA-based therapy is ideal for inhibiting influenza
temperature is higher (Maassab and Bryant, 1999). Thus, virus infection
the cold-adapted virus is administered intranasally as a
live-attenuated virus vaccine. Although this is expected to RNA interference (RNAi) is a process by which
induce local and perhaps longer-lasting protective immune double-stranded RNA (dsRNA) directs sequence-specific
responses (Palese and Garcia-Sastre, 2002), its efficacy at degradation of messenger RNA (mRNA) (Brantl, 2002;
the population level has yet to be determined. Currently, Sharp, 2001; Vaucheret et al., 2001). This phenomenon was
influenza vaccines are recommended for all individuals initially observed in plants (Baulcombe, 2002; Vaucheret
with chronic underlying diseases, those aged 65 or older, et al., 2001), in Caenorhabditis elegans (Fire et al., 1998),
and those who can transmit influenza virus to high-risk and, recently, in mammalian cells (Elbashir et al., 2001). In
individuals. plants, it is an evolutionarily conserved response to virus
Protection provided by vaccines, however, is limited. infection. Naturally occurring RNAi is initiated by the
First, the vaccinees’ age and immune status significantly af- dsRNA-specific endonuclease, called Dicer, which proces-
fect the vaccines’ effectiveness. Current vaccines, consisting sively cleaves long dsRNA into double-stranded fragments
of either killed virus or recombinant surface glycoproteins, between 21 and 25 nucleotides long, termed short inter-
can prevent illness in approximately 70–80% of healthy in- fering RNA (siRNA) (Elbashir et al., 2001). siRNAs are
dividuals under the age of 65, but only 30–40% in the elderly then incorporated into a protein complex that recognizes
(Patriarca et al., 1985). The protection rate is even lower in and cleaves target mRNAs. RNAi can be triggered in mam-
infants, immunocompromised individuals, and elderly per- malian cells by introducing synthetic 21-nucleotide siRNA
sons with chronic obstructive pulmonary disease, asthma, duplexes (Elbashir et al., 2001; Fire et al., 1998), bypass-
chronic heart disease, diabetes, chronic renal or hepatic dis- ing the requirement for Dicer-mediated processing of long
ease, neoplastic disease, or chronic connective tissue disease dsRNA. Because 21-nucleotide siRNAs are too short to
(Glezen et al., 1987; Nichol et al., 1998). Thus, the vaccines induce an interferon response in mammalian cells (Chi
are only moderately effective in those who most need pro- et al., 2003; Kumar and Carmichael, 1998; Semizarov et al.,
tection. Second, existing vaccines have to be reformulated 2003), yet still able to confer transient interference of gene
each year because the viral antigens (HA and NA) that elicit expression in a sequence-specific manner, they represent a
the neutralizing antibodies usually undergo changes, ren- new class of molecules that are likely to have significant
dering the previous year’s vaccine ineffective against new medical applications.
Q. Ge et al. / Virus Research 102 (2004) 37–42 39

RNAi appears to be ideal for inhibiting influenza virus in- PB2


5’ 3’

UTR
fection. First, influenza virus is an RNA virus, without any PB1
Coding sequence
PA
DNA intermediates during its entire life cycle (Lamb and NP siRNA
Krug, 2001). Besides mRNA, both vRNA and cRNA could M
No inhibition
Partial inhibition
also be potential targets for siRNA-mediated degradation. NS Strong inhibition

Second, the influenza virus genome consists of eight seg- Fig. 1. Comparison of relative potency of siRNAs specific for different
mented RNAs, encoding a total of 10 proteins. Each pro- influenza viral genes in inhibiting influenza virus production. Locations
tein is either an integral component of the viral structure or of siRNAs relative to their respective viral genes (PB2, PB1, etc.) are
plays a critical role during the virus life cycle. Interfering depicted. The relative potency of siRNAs in inhibiting influenza virus
with the production of any one of them is likely to have se- production is based on results in MDCK cells and chicken embryos. UTR:
untranslated regions.
vere consequences on viral replication and production. Thus,
there are multiple siRNA targets, and combinations of siR-
NAs to different targets may be used simultaneously. The in the culture supernatants at different times after infection
use of two or more siRNAs simultaneously may be required (Ge et al., 2003). The ability of siRNAs to inhibit influenza
to prevent the emergence of resistant virus, analogous to the virus production was also verified in embryonated chicken
use of drug “cocktails” for treating other infectious diseases eggs. We found that: (i) some siRNAs can potently inhibit
(caused by Mycobacteria, HIV, etc.). Third, influenza virus influenza virus production in cultured cells; (ii) influenza
naturally infects epithelial cells in the upper respiratory tract virus production can be inhibited by siRNAs specific for
and the lungs in humans. Thus, siRNAs can be administered different viral genes, especially those encoding NP, PA, and
by inhalation, which would not only be convenient but may PB1; (iii) siRNA inhibition can still occur in cells that were
also result in much higher local siRNA concentrations than infected with virus prior to siRNA introduction; (iv) the
could be achieved by parenteral injection. Considering that amount of siRNA required for significant inhibition is small
the number of virions is probably small at the onset of a (sub-nanomoles); and (v) siRNAs that potently inhibited in-
natural infection, sufficient amounts of siRNA may be de- fluenza virus production in cultured cells also inhibited virus
livered to epithelial cells in the upper airways and the lungs production in chicken embryos (Fig. 1).
to inhibit virus replication or production, thus potentially Because siRNAs are double-stranded, the antisense strand
achieving preventive and/or therapeutic effects. Finally, un- is complementary to mRNA and cRNA, and the sense strand
like vaccines that require the recipients to have a relatively is complementary to vRNA. Thus, besides mRNA, siRNA
normal immune system, siRNA-based treatment does not might also interfere with vRNA and cRNA. To investigate
depend on a functional immune system and should be as ef- these possibilities, we used two different approaches: First,
fective in the elderly or immuno-compromised individuals we used NP-specific siRNA in which either the sense (+) or
as in immunocompetent individuals. antisense (−) strand was modified. The modification, which
substituted the 2 -hydroxyl group with a 2 -O-methyl group
at every nucleotide residue, does not affect base-pairing for
4. siRNAs specific for nucleocapsid and transcriptase duplex formation, but the modified RNA strand no longer
are particularly potent in inhibiting influenza virus supports RNA interference. We found that inhibition of in-
production fluenza virus production required a wildtype antisense (−)
strand of siRNA duplex, indicating that the target of RNA
Among influenza A viruses, 15 HA subtypes and 9 NA interference is either mRNA (+) or cRNA (+) or both (Ge
subtypes are known. There are also extensive differences in et al., 2003).
nucleotide sequences of other genes among influenza virus Second, we examined the effect of siRNA on the accu-
isolates from different species. To design siRNAs that re- mulation of the corresponding mRNA, cRNA, and vRNA
main effective despite antigenic drifts and shifts, we have using reverse transcription followed by real time PCR (Ge
focused on regions of the viral genome that are conserved et al., 2003). We found that M-specific siRNA specifically
among different subtypes and strains of virus from human, inhibited the accumulation of M mRNA. However, siRNA
chicken, duck, equine, and swine (the influenza sequence specific for NP or PA abolished the accumulation of not
database, www.flu.lanl.gov). We designed a total of 20 siR- only the corresponding mRNA but also vRNA and cRNA
NAs specific for NP, PA, PB1, PB2, M, and NS genes, but not (Fig. 2). These siRNAs also broadly inhibited the accu-
for HA and NA because they contain no stretch of 21 con- mulation of other viral RNAs. This broad inhibition was
served nucleotides, a result of extensive variations in these virus-specific as it did not significantly affect RNAs tran-
genes among different virus isolates from humans and other scribed from cellular genes. The virus-specific inhibition
species. was also observed in Vero cells, which lack the interferon
The ability of synthetic siRNAs to inhibit influenza virus ␣, ␤, and ␻ genes, indicating that the broad inhibition was
production was determined in cultured cells by using graded not a result of an interferon response that shuts off all
amounts of siRNAs, introducing siRNAs into the cells ei- transcription in the infected cells. In addition, the broad
ther before or after virus infection, and assaying virus titers inhibition was not a result of a general degradation of
40 Q. Ge et al. / Virus Research 102 (2004) 37–42

(A) NP cRNA (B) NP cRNA 3

Virion NP vRNA New virion Virion NP vRNA 3 New virion 4

NP mRNA NP mRNA Other viral RNAs 3


NP siRNA
1
NP protein Degradation NP protein 2

Fig. 2. NP-specific siRNAs inhibit influenza virus production through both direct and indirect mechanisms. (A) Following influenza virus infection,
vRNA is transcribed into mRNA, which serves as a template for synthesis of viral proteins. Transcription of vRNA also produces cRNA, which in turn
serves as a template for synthesis of more vRNA for new virion production. Transcription of NP RNAs and synthesis of NP protein are depicted as an
example. (B) In the presence of NP-specific siRNA, NP mRNA is targeted for degradation (step 1). The resulting decrease in NP mRNA level results in
a decrease in NP protein synthesis (step 2). As NP protein normally binds to vRNA and cRNA and is required for their transcription and replication,
the decrease in NP protein level then indirectly results in a decrease in NP vRNA and cRNA synthesis as well as other viral RNA synthesis (step 3).
The broad shutoff of viral RNA and protein synthesis results in a severe reduction of new virion production (step 4). siRNAs specific for viral RNA
transcriptase also directly target specific mRNA for degradation and indirectly interfere with all viral RNA accumulation.

virus-specific RNAs, because activation of protein kinase R and to have a long-lasting effect. These requirements could
was not affected by the presence of siRNA in infected cells. be met by intranasal administration of siRNA, which might
Instead, these findings reveal a critical role of newly be able to achieve a high enough local siRNA concentration
synthesized NP and PA proteins in viral transcription to result in sufficient uptake of siRNA by epithelial cells. In
and replication. NP protein is required for elongation and rapidly dividing cells in cultures, sufficient amount of siRNA
antitermination of nascent cRNA and vRNA transcripts can be introduced to mediate effective gene silencing that
(Beaton and Krug, 1986; Shapiro and Krug, 1988). Without persisted for five cell divisions (McManus et al., 2002). In
newly synthesized NP, further viral transcription and repli- humans, epithelial cells in the respiratory tract do not divide
cation are blocked, as is new virion production. Similarly, significantly. Thus, it might be possible to achieve preven-
without newly synthesized RNA transcriptase, further viral tion of influenza infection by inhalation of siRNA once per
transcription and replication are evidently inhibited. These week or even less often.
findings are consistent with studies showing that siRNAs In infants, the elderly, and in immunocompromised in-
target the degradation of mRNAs but not vRNA of respira- dividuals, influenza virus infection can result in secondary
tory syncytial virus (RSV) (Bitko and Barik, 2001). siRNA infections, severe complications, and death. For these seri-
specific for RSV P gene, which encodes a subunit of the ously ill patients, currently there is no effective treatment.
RNA-dependent RNA polymerase, inhibits not only virus siRNA-based intervention may be able to fill this spe-
P protein expression but also all other viral protein expres- cific gap. Besides intranasal administration, other treatment
sion (Bitko and Barik, 2001). Both the targeted mRNA routes, such as intravenous injection, may be reasonable
degradation and the resulting global inhibition of other viral in these life-threatening situations. Besides synthetic siR-
RNA transcription make the NP- and PA-specific siRNAs NAs, siRNA transcribed from viral or DNA vectors might
especially potent inhibitors of influenza virus infection. also prove to be useful therapeutically and justifiable under
some circumstances.

5. Potential siRNA-based prophylaxis and therapy of


influenza infection in humans 6. Delivery: the challenge of siRNA-based intervention

Influenza virus normally infects epithelial cells in the up- siRNAs are negatively charged and do not readily cross
per airway and the lungs. In healthy individuals, virus is cell membrane. An effective siRNA-mediated prevention
cleared about 7 days after infection, and tissue damage is re- and treatment of influenza virus infection requires efficient
paired about 10–12 days after infection. Studies have shown non-toxic means to deliver siRNAs into epithelial cells of
that antiviral drugs are effective only when they are admin- the respiratory tract or other cell types of interest for other
istered prior to virus infection or within 1 or 2 days of in- diseases. Because of the structural and chemical similarities
fection (Hayden et al., 1991; Stiver, 2003). It is possible that between siRNA and DNA, methodologies that have been, or
siRNA may also have to be administered prior to infection are being, developed for DNA delivery (gene therapy) will
or during the early phase of infection in order to achieve pro- likely be useful for siRNA delivery. Compared to plasmid
phylactic or therapeutic effect. For prevention or treatment DNAs, siRNAs are much smaller and therefore probably
of influenza infection on a population-wide scale, siRNA easier to introduce into cells than DNA molecules. In addi-
would have to be convenient to use, relatively inexpensive, tion, siRNAs can function in the cytosol whereas DNA has
Q. Ge et al. / Virus Research 102 (2004) 37–42 41

to enter the nucleus to initiate transcription (Simeoni et al., Recently, synthetic siRNAs and shRNAs transcribed from
2003). Thus, it is possible that effective siRNA delivery will DNA templates were shown to potently inhibit gene ex-
be more easily achievable than DNA delivery. pression in mice (McCaffrey et al., 2002, 2003). These re-
Currently, there are two classes of DNA delivery vehicles: sults suggest that siRNA can be introduced into the animals
genetically engineered viruses and synthetic carriers (non- and mediate effective gene silencing. However, the hydrody-
viral carriers). Adenovirus, lentivirus, and retrovirus have namic intravascular infusion that was used to deliver siRNA
been widely investigated for gene delivery because of their or DNA into mouse liver can result in transient right-sided
high infection efficiency (Thomas et al., 2003). These virus heart failure and liver injury (Zhang et al., 2000), making it
vectors have also been successfully used to encode short unlikely to be applicable to humans. Development of tech-
hairpin RNAs (shRNAs) for expressing siRNAs in cultured nologies for safe, efficient, and target cell-specific delivery
cells and in experimental animals (Hemann et al., 2003; of siRNA will be the key to successful utilization of siRNA
Rubinson et al., 2003; Shen et al., 2003; Xia et al., 2002). Al- in medical applications.
though these viral vectors are efficient research tools, there
will likely be serious concerns about their medical use in
humans. For example, the viruses tend to infect many cell 7. Conclusions
types and may induce strong immune responses against in-
fected cells, resulting in the elimination of the infected cells. Influenza virus infection is a severe public health prob-
Some viruses can also integrate into the host genome and lem with significant personal, social, and economical conse-
might result in tumorigenesis. In particular, the idea of us- quences. More effective approaches are urgently needed to
ing one virus to prevent or treat infection caused by another treat influenza virus infections, especially in infants, the el-
virus may be difficult to gain regulatory approval, perhaps derly, and immunocompromised individuals. siRNA-based
under some circumstances. intervention seems ideal for this purpose and may meet this
Synthetic carriers, such as peptides, cationic lipids, and specific need. siRNAs specific for nucleocapsid and tran-
cationic polymers, have also been investigated for DNA scriptase have been shown to be particularly potent in in-
delivery. The most widely investigated non-viral carriers hibiting influenza virus production in cell lines and chicken
for DNA transfection are cationic polymers (Han et al., embryos. The challenge of successful siRNA-based therapy
2000). Cationic polymers bind electrostatically to DNA in the near future is to develop efficient and safe means to
and condense large plasmid DNA molecules into smaller deliver siRNA into cells of interest.
DNA/polymer complexes for more efficient endocytosis.
The DNA/cationic polymer complexes also act as bioad-
hesives because of their electrostatic interaction with nega- Acknowledgements
tively charged sialic acid residues of cell surface glycopro-
teins (Soane et al., 1999). The ability of cationic polymers We thank Drs. Peter Palese and Adolfo Garcia-Sastre for
to bind to negatively charged siRNA and still interact with kindly allowing Q.G. to visit their laboratories, Drs. Christo-
the negatively charged cell surface may facilitate endo- pher Basler, Astrid Flandorfer, Luis Martinez-Sobrido, and
cytic uptake of siRNAs. In addition, some polymers, such Yurie Nakaya for teaching Q.G. various techniques for han-
as imidazole group-modified polylysine, can apparently dling influenza virus; and members of the Chen and Eisen
disrupt the acidic endosomes and, therefore, facilitate re- laboratories for helpful discussions. This work was sup-
lease of DNA or RNA into the cytosol (Putnam et al., ported in part by grants from the National Institutes of Health
2001). AI40146, AI44478, and AI50631 (to J.C.), AI44477 and
The advantages of nonviral carriers include greater con- CA60686 (to H.N.E.).
trol of the molecular composition of the final products, flex-
ibility in the amount of siRNAs to be delivered, and reduced
safety concerns. In addition, some polymers are also low
in immunogenicity. However, the delivery efficiency of the References
nonviral carriers is generally low compared to viral vectors.
Relatively large doses of DNA, and perhaps siRNA, are usu- Anderson, D.G., Lynn, D.M., Langer, R., 2003. Semi-automated synthesis
and screening of a large library of degradable cationic polymers for
ally required for animal experiments as well as clinical trials
gene delivery. Angew Chem. Int. Ed. Engl. 42, 3153–3158.
in humans (Donnelly et al., 2003). It is reported that approx- Baulcombe, D., 2002. RNA silencing. Curr. Biol. 12, R82–R84.
imately 106 copies of plasmid DNA molecules are needed Beaton, A.R., Krug, R.M., 1986. Transcription antitermination during in-
to transfect a single cell, with ∼102 –104 copies having to fluenza viral template RNA synthesis requires the nucleocapsid pro-
make it into the nucleus for transcription (Tachibana et al., tein and the absence of a 5 capped end. Proc. Natl. Acad. Sci. U.S.A.
83, 6282–6286.
2002). Nevertheless, rapid development of novel polymers
Bitko, V., Barik, S., 2001. Phenotypic silencing of cytoplasmic genes
or modification of existing polymers or cationic lipids will using sequence-specific double-stranded short interfering RNA and its
likely yield carriers that can deliver DNA and siRNA into application in the reverse genetics of wild type negative-strand RNA
cells efficiently (Anderson et al., 2003). viruses. BMC Microbiol. 1, 34.
42 Q. Ge et al. / Virus Research 102 (2004) 37–42

Brantl, S., 2002. Antisense-RNA regulation and RNA interference. nursing homes. Reduction in illness and complications during an in-
Biochim. Biophys. Acta 1575, 15–25. fluenza A (H3N2) epidemic. JAMA 253, 1136–1139.
Cheung, M., Lieberman, J.M., 2002. Influenza: update on strategies for Patterson, K.D., Pyle, G.F., 1991. The geography and mortality of the
management. Contemp. Pediatr. 19, 82–114. 1918 influenza pandemic. Bull. Hist. Med. 65, 4–21.
Chi, J.T., Chang, H.Y., Wang, N.N., Chang, D.S., Dunphy, N., Brown, Putnam, D., Gentry, C.A., Pack, D.W., Langer, R., 2001. Polymer-based
P.O., 2003. Genomewide view of gene silencing by small interfering gene delivery with low cytotoxicity by a unique balance of side-chain
RNAs. Proc. Natl. Acad. Sci. U.S.A. 100, 6343–6346. termini. Proc. Natl. Acad. Sci. U.S.A. 98, 1200–1205.
Claas, E.C., A.D., O., van Beek, R., De Jong, J.C., Rimmelzwaan, G.F., Rubinson, D.A., Dillon, C.P., Kwiatkowski, A.V., Sievers, C., Yang, L.,
Senne, D.A., Krauss, S., Shortridge, K.F., Webster, R.G., 1998. Human Kopinja, J., Rooney, D.L., Ihrig, M.M., McManus, M.T., Gertler,
influenza A H5N1 virus related to a highly pathogenic avian influenza F.B., Scott, M.L., Van Parijs, L., 2003. A lentivirus-based system to
virus. Lancet 351, 472–477. functionally silence genes in primary mammalian cells, stem cells
Donnelly, J., Berry, K., Ulmer, J.B., 2003. Technical and regulatory hurdles and transgenic mice by RNA interference. Nat. Genet. 33, 401–
for DNA vaccines. Int. J. Parasitol. 33, 457–467. 406.
Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl, Semizarov, D., Frost, L., Sarthy, A., Kroeger, P., Halbert, D.N., Fesik,
T., 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference S.W., 2003. Specificity of short interfering RNA determined through
in cultured mammalian cells. Nature 411, 494–498. gene expression signatures. Proc. Natl. Acad. Sci. U.S.A. 100, 6347–
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello, 6352.
C.C., 1998. Potent and specific genetic interference by double-stranded Shapiro, G.I., Krug, R.M., 1988. Influenza virus RNA replication in vitro:
RNA in Caenorhabditis elegans. Nature 391, 806–811. synthesis of viral template RNAs and virion RNAs in the absence of
Ge, Q., McManus, M., Nguyen, T., Shen, C.-H., Sharp, P.A., Eisen, H.N., an added primer. J. Virol. 62, 2285–2290.
Chen, J., 2003. RNA interference of influenza virus production by Sharp, P.A., 2001. RNA interference—2001. Genes Dev. 15, 485–
directly targeting mRNA for degradation and indirectly inhibiting all 490.
viral RNA transcription. Proc. Natl. Acad. Sci. U.S.A. 100, 2718– Shen, C., Buck, A.K., Liu, X., Winkler, M., Reske, S.N., 2003. Gene
2723. silencing by adenovirus-delivered siRNA. FEBS Lett. 539, 111–
114.
Glezen, W.P., Decker, M., Perrotta, D.M., 1987. Survey of underlying
Simeoni, F., Morris, M.C., Heitz, F., Divita, G., 2003. Insight into the
conditions of persons hospitalized with acute respiratory disease during
mechanism of the peptide-based gene delivery system MPG: impli-
influenza epidemics in Houston, 1978–1981. Am. Rev. Respir. Dis.
cations for delivery of siRNA into mammalian cells. Nucleic Acids
136, 550–555.
Res. 31, 2717–2724.
Han, S.-O., Mahato, R.I., Sung, Y.K., Kim, S.W., 2000. Development of
Soane, R.J., Frier, M., Perkins, A.C., Jones, N.S., Davis, S.S., Illum,
biomaterials for gene therapy. Mol. Ther. 2, 302–317.
L., 1999. Evaluation of the clearance characteristics of bioadhesive
Hayden, F.G., Sperber, S.J., Belshe, R.B., Clover, R.D., Hay, A.J., Pyke, S.,
systems in humans. Int. J. Pharm. 178, 55–65.
1991. Recovery of drug-resistant influenza A virus during therapeutic
Stiver, G., 2003. The treatment of influenza with antiviral drugs. CMAJ
use of rimantadine. Antimicrob. Agents. Chemother. 35, 1741–1747.
168, 49–56.
Hemann, M.T., Fridman, J.S., Zilfou, J.T., Hernando, E., Paddison, P.J.,
Tachibana, R., Harashima, H., Ide, N., Ukitsu, S., Ohta, Y., Suzuki, N.,
Cordon-Cardo, C., Hannon, G.J., Lowe, S.W., 2003. An epi-allelic
Kikuchi, H., Shinohara, Y., Kiwada, H., 2002. Quantitative analysis of
series of p53 hypomorphs created by stable RNAi produces distinct
correlation between number of nuclear plasmids and gene expression
tumor phenotypes in vivo. Nat. Genet. 33, 396–400.
activity after transfection with cationic liposomes. Pharm. Res. 19,
Kemble, G., Greenberg, H., 2003. Novel generations of influenza vaccines. 377–381.
Vaccine 21, 1789–1795. Taubenberger, J.K., Reid, A.H., Janczewski, T.A., Fanning, T.G., 2001.
Kumar, M., Carmichael, G.G., 1998. Antisense RNA: function and fate Integrating historical. Phil. Trans. R. Soc. Lond. B: Biol. Sci. 356,
of duplex RNA in cells of higher eukaryotes. Microbiol. Mol. Biol. 1829–1839.
Rev. 62, 1415–1434. Thomas, C.E., Ehrhardt, A., Kay, M.A., 2003. Progress and problems with
Lamb, R.A., Krug, R.M., 2001. Orthomyxoviridae: the viruses and their the use of viral vectors for gene therapy. Nat. Rev. Genet. 4, 346–358.
replication. In: Knipe, D.M., Howley, P.M. (Eds.), Fundamental Vi- Thompson, W.W., Shay, D.K., Weintraub, E., Brammer, L., Cox, N.,
rology, pp. 725–770. Anderson, L.J., Fukuda, K., 2003. Mortality associated with influenza
Luscher-Mattli, M., 2000. Influenza chemotherapy: a review of the present and respiratory syncytial virus in the United States. JAMA 289, 179–
state of art and of new drugs in development. Arch. Virol. 145, 2233– 186.
2248. Vaucheret, H., Beclin, C., Fagard, M., 2001. Post-transcriptional gene
Maassab, H.F., Bryant, M.L., 1999. The development of live attenuated silencing in plants. J. Cell Sci. 114, 3083–3091.
cold-adapted influenza virus vaccine for humans. Rev. Med. Virol. 9, Webby, R.J., Webster, R.G., 2001. Emergence of influenza A viruses.
237–244. Phil. Trans. R. Soc. Lond. B: Biol. Sci. 356, 1817–1828.
McCaffrey, A.P., Meuse, L., Pham, T.T., Conklin, D.S., Hannon, G.J., Webster, R.G., Bean, W.J., Gorman, O.T., Chambers, T.M., Kawaoka, Y.,
Kay, M.A., 2002. RNA interference in adult mice. Nature 418, 38–39. 1992. Evolution and ecology of influenza A viruses. Microbiol. Rev.
McCaffrey, A.P., Nakai, H., Pandey, K., Huang, Z., Salazar, F.H., Xu, H., 56, 152–179.
Wieland, S.F., Marion, P.L., Kay, M.A., 2003. Inhibition of hepatitis Webster, R.G., Laver, W.G., Air, G.M., Schild, G.C., 1982. Molecular
B virus in mice by RNA interference. Nat. Biotechnol. 21, 639–644. mechanisms of variation in influenza viruses. Nature 296, 115–121.
McManus, M.T., Haines, B.B., Dillon, C.P., Whitehurst, C.E., van Parijs, Xia, H., Mao, Q., Paulson, H.L., Davidson, B.L., 2002. siRNA-mediated
L., Chen, J., Sharp, P.A., 2002. Small interfering RNA-mediated gene gene silencing in vitro and in vivo. Nat. Biotechnol. 20, 1006–1010.
silencing in T lymphocytes. J. Immunol. 169, 5754–5760. Yuen, K.Y., Chan, P.K., Peiris, M., Tsang, D.N., Que, T.L., Shortridge,
Nichol, K.L., Wuorenma, J., von Sternberg, T., 1998. Benefits of influenza K.F., Cheung, P.T., To, W.K., Ho, E.T., Sung, R., Cheng, A.F., 1998.
vaccination for low-, intermediate-, and high-risk senior citizens. Arch. Clinical features and rapid viral diagnosis of human disease associated
Intern. Med. 158, 1769–1776. with avian influenza A H5N1 virus. Lancet 351, 467–471.
Palese, P., Garcia-Sastre, A., 2002. Influenza vaccines: present and future. Zhang, G., Song, Y.K., Liu, D., 2000. Long-term expression of human
J. Clin. Invest. 110, 9–13. alpha1-antitrypsin gene in mouse liver achieved by intravenous ad-
Patriarca, P.A., Weber, J.A., Parker, R.A., Hall, W.N., Kendal, A.P., Breg- ministration of plasmid DNA using a hydrodynamics-based procedure.
man, D.J., Schonberger, L.B., 1985. Efficacy of influenza vaccine in Gene Ther. 7, 1344–1349.
Virus Research 102 (2004) 43–51

RNA silencing of rotavirus gene expression


Carlos F. Arias, Miguel A. Dector, Lorenzo Segovia, Tomás López, Minerva Camacho,
Pavel Isa, Rafaela Espinosa, Susana López∗
Departamento de Génetica del Desarrollo y Fisiologı́a Molecular, Instituto de Biotecnologı́a, Universidad Nacional Autónoma de México,
Avenida Universidad 2001, Col. Chamilpa, Cuernavaca, Morelos 62210, Mexico

Abstract

RNA interference (RNAi) is a double-stranded RNA (dsRNA)-triggered mechanism for suppressing gene expression, which is conserved
in evolution and has emerged as a powerful tool to study gene function. Rotaviruses, the leading cause of severe diarrhea in young children,
are formed by three concentric layers of protein, and a genome composed of 11 segments of dsRNA. Here, we show that the RNAi machinery
can be triggered to silence rotavirus gene expression by sequence-specific short interfering RNAs (siRNAs). RNAi is also useful for the study
of the virus-cell interactions, through the silencing of cellular genes that are potentially important for the replication of the virus. Interestingly,
while the translation of mRNAs is readily stopped by the RNAi machinery, the viral transcripts involved in virus genome replication do not
seem to be susceptible to RNAi. Since gene silencing by RNAi is very efficient and specific, this system could become a novel therapeutic
approach for rotavirus and other virus infections, once efficient methods for in vivo delivery of siRNAs are developed. Although the use of
RNAi as an antiviral therapeutic tool remains to be demonstrated, there is no doubt that this technology will influence drastically the way
postgenomic virus research is conducted.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Rotavirus; RNA interference; siRNAs; Antiviral agents; Reoviridae

1. Introduction trol the spread of rotavirus infections, there is an urgent need


to develop effective vaccination and therapeutic strategies.
Acute, infectious diarrhea is the most common cause of Fundamental to these developments is a basic understanding
morbidity and mortality among young children living in de- of the molecular mechanisms by which rotaviruses interact
veloping countries, accounting for as many as one billion with their host cell.
illnesses and between 2.5 and 3.2 million deaths annually Rotaviruses, a genus of the Reoviridae family, are formed
(Parashar and Glass, 2003). Rotaviruses are the leading eti- by three concentric layers of protein that enclose a genome
ologic agent of severe diarrheal disease in infants and young composed of 11 segments of double stranded RNA (dsRNA),
children worldwide. In developed countries, rotaviruses have ranging in size from approximately 660–3300 base pairs
been detected in 30–50% of infants hospitalized with acute (bp). The total genomic size is about 18,500 bp. The inner-
diarrhea. In less developed countries, rotaviruses are also most layer of the virion is formed by 120 molecules of pro-
the most frequently detected pathogen in children with se- tein VP2, which surrounds the viral genome. Twelve copies
vere gastroenteritis. While the mortality from rotavirus dis- each of VP1, the RNA polymerase, and VP3, a guanylyl-
ease in developed countries is very low, rotavirus causes an transferase and methylase, constitute the core of the virus.
estimated 500,000–600,000 deaths each year (Parashar and The addition of 260 trimers of VP6 on top of the VP2
Glass, 2003). The frequency of rotavirus infection is remark- layer produces double-layered particles (DLPs). The outer-
ably similar in both settings. Since rotaviruses play such an most layer, characteristic of triple-layered particles (TLPs),
important role in severe dehydrating gastroenteritis, and be- is composed of two proteins, VP4 and VP7. The smooth
cause even advanced levels of hygiene seem unable to con- external surface of the virus is made up of 780 copies of
glycoprotein VP7, organized as trimers, while 60 spike-like
∗ Corresponding author. Tel.: +52-777-3291615; structures, formed by dimers of VP4, extend about 12 nm
fax: +52-777-3172388. from the VP7 surface. The mature virus particle is ap-
E-mail address: susana@ibt.unam.mx (S. López). proximately 100 nm in diameter and contains 132 porous

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.014
44 C.F. Arias et al. / Virus Research 102 (2004) 43–51

channels, which allow the influx of compounds in aqueous the cell’s cytoplasm through the class I channels located at
solution to the inside of the capsid and the efflux of newly the icosahedral five-fold vertices of the particle (Pesavento
formed mRNAs (Pesavento et al., 2003). et al., 2003).
VP4 has essential functions in the virus life cycle, includ- The RNA transcripts direct the synthesis of six structural
ing receptor binding and cell penetration. The role of VP7 and six non-structural viral proteins (i.e., function as mR-
during the initial interactions of the virus with the cell is less NAs) and also serve as RNA templates (RNA(+)) for the
clear, although it has been recently shown that VP7 interacts synthesis of the RNA negative strands (RNA(−)), to form
with cell surface molecules at a step subsequent to the initial the dsRNA genome segments. Once a critical mass of viral
attachment of the virus through the spike protein (Graham proteins is accumulated into structures known as viroplasms,
et al., 2003; Zárate et al., 2002). After the virus attaches to core replication intermediate (RI) particles assemble. The
the cell surface, it has to penetrate the plasma membrane to synthesis of RNA(−) has been proposed to occur concur-
productively infect the cell. This penetration is increased by, rently with the packaging of RNA(+) into the core RIs, and
and most probably dependent on, trypsin treatment of the is highly coordinated in such a way that packaging and repli-
virus that results in the specific cleavage of VP4 to polypep- cation lead to the formation of cores containing one copy
tides VP8 and VP5. The cleavage of VP4 does not affect of each of the 11 genome segments of dsRNA (Patton and
cell binding and is rather associated with the entry of the Spencer, 2000). In addition to the virus RNA polymerase,
virus into the cell (Arias et al., 2001; Estes, 2001). the non-structural proteins NSP2 and NSP5 are thought to
During, or shortly after cell entry, the infecting TLP is be essential for the early steps of morphogenesis. All these
uncoated, loosing the two proteins of the outer layer, and processes lead to the production of transcriptionally ac-
yielding a DLP, which is transcriptionally active. VP1 syn- tive, dsRNA-containing double-layered RI particles. These
thesizes the primary viral transcripts, which are extruded into particles are responsible for an enhanced second round of

Fig. 1. Replication cycle of rotaviruses. The different steps in the replication cycle of the virus are indicated by numbers—1: attachment of the virion to
the cell surface; 2: penetration and uncoating of the virus particle to yield DLPs; 3: primary transcription of the genomic dsRNA; 4: synthesis of viral
proteins; 5: assembly of core RIs and negative strand RNA synthesis; 6: assembly of double-layered RIs; 7: secondary transcription from double-layered
RIs; 8: secondary, enhanced synthesis of viral proteins; 9: secondary, increased assembly of core RIs and negative strand RNA synthesis; 10: secondary,
increased assembly of double-layered RIs; 11: budding of double-layered RIs through the membrane of the endoplasmic reticulum (ER), and acquisition
of a membrane envelope; 12: loss of the membrane envelope and generation of mature triple-layered virions.
C.F. Arias et al. / Virus Research 102 (2004) 43–51 45

transcription, which results in a second wave of assembly Rotaviruses have a segmented dsRNA genome, which is
of double-layered RI particles, which then bud through the replicated in the cytoplasm of cells. The dsRNA genome,
membrane of the ER. During this budding, which is medi- however, is masked at all times during the virus replication
ated by the interaction of the double-layered RIs with the ER cycle, being found always associated with subviral particles
membrane-associated rotavirus protein NSP4, the particles and not free in the cellular cytosol. This probably prevents
acquire a transient membrane envelope (Estes, 2001). The the rotavirus genome to be detected and targeted by gen-
transiently enveloped particles contain, in addition to NSP4, eral defense mechanisms of the cell. In addition, the virus
the virus surface proteins VP4 and VP7, as well as minor replication cycle is highly lytic and rapid, being completed
amounts of other non-structural proteins (Poruchynsky and in about 12 h. The features of rotavirus replication repre-
Atkinson, 1991). The lipid envelope is then removed by a sented a challenge for the RNAi machinery to silence the
largely unknown mechanism, to yield mature TLPs (Fig. 1). virus gene expression and to block the replication of the
Apart from glycoprotein NSP4, the other five nonstruc- virus.
tural proteins (NSP1 to NSP3, NSP5, and NSP6) have the We showed that a siRNA with a sequence homologous to
ability to bind RNA (Patton, 1995) and are thought to be in- the gene encoding the virus spike protein VP4 (siRNAVP4 )
volved in the replication of the viral genome, although their efficiently inhibited the synthesis of VP4 to a barely de-
precise function is not known. Despite the efforts of many tectable level in cells transfected with the siRNA, whereas
laboratories, at present there is no reverse genetics system all other structural proteins remained unaltered (Dector
available for rotaviruses. Thus, the in vivo characterization of et al., 2002). Similarly, the yield of progeny virus that
rotavirus gene function has been limited so far to the charac- resulted from an infection carried out in the presence of
terization of antibody escape variants, temperature-sensitive siRNAVP4 was reduced to 15–25% that of a control in-
mutants, and reassortants. fection performed in the presence of an unrelated siRNA
(homologous to the nuclear proteins laminA/C). The resid-
ual infectious virus produced in the presence of siRNAVP4
2. RNA interference and rotaviruses was mostly the result of virus replication in those cells that
were not transfected with the siRNA, the transfection effi-
RNA interference (RNAi) has become a powerful and ciency achieved being about 75%. As result of the inhibition
widely used tool for the analysis of gene functions. of VP4 synthesis, non-infectious TLPs, lacking the VP4
This evolutionarily conserved mechanism, triggered by protein (appearing as “spike-less” TLPs), were obtained
double-stranded RNA (dsRNA), specifically suppresses (Dector et al., 2002).
gene expression by selectively degrading mRNAs matching We have also addressed the question of the duration of
the sequence of the dsRNA that triggered the response, an effective RNAi response in a rotavirus infection. For
without affecting the expression of other genes. During this, we transfected MA104 cells with either a siRNA to
RNAi, long dsRNA molecules are processed into 21–25 bp VP4 or to VP7, and then infected with rotavirus at differ-
RNAs known as short-interfering RNAs (siRNAs) that serve ent times post-transfection. The inhibition of the synthe-
as guides for enzymatic cleavage of complementary RNAs, sis of both viral proteins could be achieved even when the
thus offering an exquisite mode to specifically knockdown cells were infected 7 days after transfection (shown up to
the expression of a particular gene. day 5 in Fig. 2A), with the consequence of a significant
This phenomenon has been observed in invertebrates and decrease on the yield of viral progeny (Fig. 2B). The in-
plants (Hannon, 2002; Sharp, 2001; Zamore, 2001); how- hibitory effect could probably last even longer, however,
ever, the demonstration of an RNAi-like response in somatic after the fifth day cells in culture were notoriously dam-
mammalian cells had been hampered by dsRNA-induced aged, even in the untransfected controls, so that it was dif-
mechanisms, which non-specifically inhibit gene expres- ficult to have reproducible results at later times. Of inter-
sion. In mammalian cells, dsRNA fragments longer than est, we consistently found that the highest inhibition was
30 bp induce components of the interferon response, in- achieved when the cells were infected 72 h after transfec-
cluding the dsRNA-dependent protein kinase (PKR), the tion with the siRNA, suggesting that some of the elements
2 ,5 -oligoadenylate synthetase (OAS) and the non-specific of the RNAi response could be induced or activated by
RNAse L (Katze et al., 2002). These factors mediate a gener- the presence of the siRNA, increasing their effective con-
alized inhibition of gene expression through the non-specific centration inside the cell. Our observations are in agree-
shutdown of protein synthesis and mRNA degradation, what ment with other studies carried out with different viruses
eventually leads to cell apoptosis. The recent demonstration and different cell lines, where the RNAi effect was re-
that synthetic 21-nt siRNA duplexes effectively inhibit mam- ported to last also about 5 days (Flores-Jasso et al., 2004;
malian endogenous gene expression in a sequence-specific Ge et al., 2004; Kapadia et al., 2003). It is not clear, how-
manner without activating the nonspecific dsRNA responses ever, whether this effect is due to the fact that the siRNAs
(Caplen, 2002; Elbashir et al., 2001) offers great opportuni- are stable within the cells, or whether they are slowly re-
ties to use this pathway of gene silencing to study the func- leased from the membrane-associated transfection mixture
tion of mammalian virus genes. (Gitlin and Andino, 2003).
46 C.F. Arias et al. / Virus Research 102 (2004) 43–51

Fig. 3. Rotavirus gene expression is efficiently inhibited even if RNAi is


induced after viral infection. MA104 cells in 48-well plates were infected
for 1 h with rotavirus RRV, at an MOI of 3. At the indicated times
Fig. 2. The RNAi response lasts up to 7 days. MA104 cells in 48-well post-infection (hours post-infection, h.p.i.), the cells were transfected with
plates were transfected with an siRNA directed to VP4 (4), VP7 (7), a siRNA to VP4 (4), or lam A/C(C) as control, for 8 h. At 12 h.p.i. the
or lamin A/C (C), and after 8 h the lipofectamine–siRNA mixture cells were labeled with [35 S]-methionine for 1 h, lysed, and the proteins
was removed and replaced by medium. At the indicated times (days separated by gel electrophoresis and analyzed by fluorography (A), or by
post-transfection, d.p.t.), the cells were infected with rotavirus RRV at an Western blot using a monoclonal antibody to VP4 (B). The dots in (A)
MOI of 1 and 12 h post-infection (p.i.), the cells were harvested and pro- indicate the migration of viral proteins other than VP4.
cessed for electrophoresis or infectivity assays. (A) Western blot stained
with a rabbit polyclonal antibody to rotavirus TLPs. Arrows indicate the
positions of VP4, VP6, and VP7. (B) The yield of progeny virus ob-
tained after transfection with the siRNA to VP4 was determined by an the RNA interference of the cell by producing proteins that
immunoperoxidase assay, as described (Lizano et al., 1991). Data are ex- suppress this response at different levels (Ding et al., 2004;
pressed as the percentage of the infectivity obtained when the cells were Li et al., 2002; Roth et al., 2004; Vance and Vaucheret, 2001).
transfected with the control siRNA to lamin A/C. Although it is not yet known whether the RNAi has antiviral
functions in mammalian cells, it is tempting to speculate that
The results obtained in our work indicate that rotaviruses one of the rotavirus proteins could act as a suppressor of a
are sensitive to the RNAi response of the cell, as has been cellular RNAi response triggered by the viral infection.
described for viruses belonging to other families, such as po- In a previous publication we described that the synthesis
liovirus, hepatitis B and C viruses, human immunodeficiency of a viral protein in MA104 cells lipofected with a siRNA
virus, respiratory syncytial virus, vesicular stomatitis virus, specific for a viral protein 48–72 h prior to infection, was
papillomavirus, herpes virus, influenza virus, dengue virus, silenced in a sequence-specific manner (Dector et al., 2002,
and baculovirus, some of which are discussed in accom- unpublished results), suggesting that rotavirus infection can-
panying articles of this Issue (Adelman et al., 2002; Bitko not suppress the interfering process. However, since in those
and Barik, 2001; Capodici et al., 2002; Coburn and Cullen, experiments the siRNA was added to the cells before virus
2002; Ge et al., 2003; Gitlin et al., 2002; Hall and infection, we reasoned that for a virus-encoded suppressor
Alexander, 2003; Hamasaki et al., 2003; Hu et al., 2002; of RNAi, there might have not been enough time for its syn-
Jacque et al., 2002; Jia and Sun, 2003; Jiang and Milner, thesis before the RNAi response (triggered by the siRNA)
2002; Kapadia et al., 2003; Klein et al., 2003; Martinez inhibited the virus replication. To address this issue, we eval-
et al., 2002; McCaffrey et al., 2003; Novina et al., 2002; uated the capacity of the RNAi response to silence rotavirus
Randall et al., 2003; Shlomai and Shaul, 2003;Valdes et al., gene expression when the siRNAs were transfected into cells
2003; Wilson et al., 2003). after infection with the virus. We found that the synthesis
of VP4 was silenced even when the siRNAVP4 was added
at 4 h post-infection (p.i.), a time point at which viral pro-
3. Is there a suppression of the RNAi response during tein synthesis is already predominant in the infected cells
rotavirus infection? (Fig. 3). These results suggest that the RNAi machinery is
active and effective even in the presence of previously syn-
It has been found that many plant viruses, and at least thesized rotaviral proteins, and thus that rotaviruses do not
one insect virus (flock house virus) are able to counteract encode an RNAi suppressor. However, in order to formally
C.F. Arias et al. / Virus Research 102 (2004) 43–51 47

demonstrate that rotaviruses do not code for suppressors of ulovirus, human immunodeficiency virus, and retroviruses
RNAi, further experiments need to be done. For example, it in this issue, and for herpes virus, papillomavirus and the
is necessary to determine if rotavirus infection could block hepatitis delta agent (Chang and Taylor, 2003; Hall and
an RNAi response triggered by short hairpin RNAs, which Alexander, 2003; Jia and Sun, 2003).
have been shown to specifically induce the RNAi system. In the case of rotaviruses, one could explore the function
These short hairpin RNAs are substrates for the cellular en- of the viral proteins in three different conceptual areas: (i)
zyme Dicer (Brummelkamp et al., 2002), which is respon- proteins that do not participate in the replication of the virus
sible for processing long dsRNA molecules into siRNAs. genome, but are involved in the late steps of virus morpho-
Also, it could be helpful to assay candidate viral proteins genesis; (ii) proteins involved directly or indirectly in the
as RNAi suppressors in a heterologous system, like plants. replication of the genomic RNA; and (iii) proteins that, al-
With such an approach, it was recently reported that the ␴3 though not directly involved in virus replication, contribute
protein of reovirus, which is a dsRNA binding protein, has to make the replication cycle more efficient. Some viral pro-
a supressor activity that blocks the synthesis of GFP in an teins could certainly have activities that could fit into more
assay for suppressor silencing in tobacco plants (Lichner than one of the areas described.
et al., 2003; Roth et al., 2004). Silencing the viral genes that encode proteins involved in
the late steps of morphogenesis, like VP4 and VP7, should
allow the replication cycle to reach the synthesis of the
4. RNA interference to study the function of rotavirus second wave of double-layered RIs. However, after this
proteins point the morphogenesis of the virus should be impaired.
In agreement with this hypothesis, we have shown that the
The segmented nature of the genome of the viruses in silencing of the VP4 gene allowed the synthesis of abun-
the Reoviridae family makes them particularly amenable to dant double-layered RIs, that efficiently bud through the ER
analysis by RNAi. In the case of rotaviruses, each of the membrane, eventually loosing the transient lipid membrane
11 segments of dsRNA is transcribed into a single mRNA, and incorporating the outer protein layer formed by VP7,
each encoding a single protein (with the exception of one to form spike-less TLPs. Of interest, the VP7 outer layer
bicistronic segment). This makes it possible to silence the assembled on the spike-less TLPs was found by cryoelec-
expression of individual genes without affecting the expres- tron microscopy to have an structure identical to that of the
sion of the others. The analysis of the phenotypes generated outer layer in wild-type TLPs (B.V.V. Prasad, unpublished
should allow the characterization of the function of proteins results), clearly showing that the assembly of VP7 on DLPs
encoded by the silenced genes. is a process independent of the assembly of VP4. In addition,
This sort of individual gene function analysis is more dif- these findings indicated that VP4 is neither required for the
ficult to carry out in viruses with a positive strand RNA budding process nor for the removal of the lipid membrane
genome, since, as it has been shown for poliovirus, hep- from the enveloped particles, as had been suggested (Estes,
atitis C (Randall and Rice, 2004; Saleh et al., 2004), and 2001). Similarly, silencing the VP7 gene prevented the for-
SARS-coronavirus (Zhang et al., 2003), their genome is mation of TLPs, and caused DLPs to accumulate (Patton,
targeted by the RNAi machinery, inhibiting the replication 2003; Camacho et al., manuscript in preparation). A priori,
of the virus. Since the genome of these viruses functions one would expect that inhibition of the synthesis of NSP4,
as mRNA to direct the synthesis of precursor polyproteins the ER membrane receptor for DLPs, should also lead to an
which are post-translationally cleaved to yield the individual accumulation of DLPs. However, since it is known that this
viral polypeptides, any siRNA directed against the mRNA protein alters the calcium homeostasis of cells (Tian et al.,
(represented by either the genomic RNA, or the subgenomic 1994, 1995), its absence might have additional effects on the
mRNAs in some virus families) would result in a decreased virus replication cycle. Silencing the NSP4 and VP7 genes
synthesis of the entire polyprotein, preventing the study should allow us to establish directly the role of these pro-
of the role of individual polypeptides. In this regard, it is teins in the translocation of DLPs into the lumen of the ER
of interest that in viruses with a non-segmented, negative and in the removal of the intermediary lipid envelope.
strand RNA genome, like respiratory syncytial virus, vesic- RNAi should also be useful to study in vivo the role of
ular stomatitis virus, and human parainfluenza virus, the ge- proteins that have been suggested to be involved in the early
nomic RNA as well as the replication intermediate RNA(+), stages of the virus replication cycle, like the viral polymerase
are protected from the action of RNAi, while only the mR- VP1, the protein that forms the innermost shell of the virion
NAs are degraded (Barik, 2004; Bitko and Barik, 2001). VP2, and the nonstructural proteins NSP2 and NSP5, all
This also seems to be the case for influenza A viruses which of which are thought to be necessary to generate core RIs
have a genome composed of eight single-stranded RNA seg- containing newly formed genomic dsRNA segments (Patton
ments of negative polarity (Ge et al., 2003). The functions and Spencer, 2000). One would expect that silencing the
of proteins of DNA viruses and retroviruses, whose genome expression of either of these genes would block the virus
is in general transcribed into monocistronic mRNAs, is also secondary transcription, with the concomitant inhibition of
amenable to dissection by RNAi, as has been shown for bac- the synthesis of all viral proteins, as it has been found for
48 C.F. Arias et al. / Virus Research 102 (2004) 43–51

VP1 and NSP5 (Campagna et al., 2003; T. López and M. of these proteins in the virus replication cycle should be
Arias, manuscript in preparation). A more detailed anal- amenable to analysis by RNAi.
ysis of these data should allow us to define the role of Knockdown of the caveolin-1 gene and other genes en-
each protein in the formation of functional cores. Also, coding proteins involved in different types of cell endocyto-
since most viral proteins do not have a random distribu- sis, like those encoding clathrin and Eps15, should also help
tion within the cell, the inhibition of the synthesis of par- to study the role of these proteins in virus entry. Using this
ticular proteins should help to identify the role they might approach, together with pharmacological and other genetic
have in the overall intracellular organization of the viral approaches, we have recently described that rotavirus cell
polypeptides. entry occurs through a caveolae- and clathrin-independent
In addition, RNAi could serve to approach the character- endocytosis, (Sánchez-SanMartı́n et al., submitted).
ization of the role of viral proteins that have been proposed
to favor the replication cycle of the virus through, for in-
stance, inhibiting the cellular protein synthesis (NSP3), in- 6. RNAi induces the degradation of only a subset of the
creasing the translation rate of the viral proteins (Piron et al., viral transcripts
1998), counteracting cellular activities that might be noxious
to virus replication, such as the interferon response (NSP1) During rotavirus infection, the DLPs derived from the
(Graff et al., 2002), or promoting the cytopathic effect to infecting viruses synthesize 11 viral transcripts. Since these
facilitate the release of virus particles (NSP4) (Estes, 2003). transcripts function both as mRNA and as RNA(+), it was
It is foreseen that in the near future many new studies expected that siRNAs directed to a given transcript would
using this methodology will help to unravel the role of all impair the synthesis of both the corresponding protein and
rotaviral proteins in vivo, both in cell culture and in ani- the corresponding dsRNA segment. However, we and others
mal models. The feasibility of inhibiting virus replication have found that when silencing rotavirus genes encoding
in complete animals has already been shown for hepatitis C proteins not involved in the replication of the viral genome,
using the mouse as a model (McCaffrey et al., 2002). such as VP4 and VP7, the newly assembled particles, either
spike-less TLPs (in the case of silencing VP4) or DLPs (in
the case of silencing VP7), contain all 11 RNA segments
5. RNAi to study rotavirus–host cell interactions in equimolar amounts (Dector et al., 2002; Patton, 2003;
Camacho et al., manuscript in preparation), despite the fact
Recently, a number of cellular proteins have been sug- that the synthesis of the target protein is almost completely
gested to participate in the virus replication cycle. These ablated, indicating that the mRNA has been degraded.
proteins range from virus cellular receptors to chaperones The previous observations have been taken to suggest
and transcription factors. It is clear that RNAi will help to that viral transcripts establish two, functionally different and
elucidate the role of these cell molecules during the infec- physically separated pools (Patton, 2003; Camacho et al.,
tious process. manuscript in preparation), one that directs the synthesis of
The entry of rotaviruses into cells has been described as proteins, and the other that is used as template for repli-
a multistep process where at least four interactions between cation of the virus genome. The mRNAs are accessible in
the viral surface proteins and cellular surface molecules the cytoplasm to be translated (where they can be efficiently
occur. The molecules implicated as virus receptors include targeted by RNAi), whereas the RNA(+) molecules act-
gangliosides GM1 and GM3, integrins ␣2␤1, ␣x␤2, and ing as template for dsRNA replication could be either kept
␣v␤3, and the heat shock cognate protein hsc70 (Arias et al., within the so-called viroplasms (where the synthesis of the
2001). Silencing of individual receptors or combinations RNA(−) is thought to take place) or could be associated
of them should help to establish their role during virus in the cytoplasm with RNA binding proteins, making them
entry. In fact, preliminary experiments have shown that inaccessible to the RNAi machinery. Even though the ex-
inhibition of ␣v␤3 expression decreases the ability of istence of two separated viral transcripts pools would ap-
rotaviruses to infect epithelial cells (P. Isa, unpublished pear to be logical, the common belief is that viral tran-
results). scripts represent a single pool of RNA that could be used at
It has been shown that rotavirus infection induces the any time, and stochastically, for either of the two functions.
expression of a large set of genes, including those encoding The hypothesis that a mRNA pool could be physically sep-
heat shock proteins hsp90 and hsp70, as well as the glucose arated from the RNA(+) (replication) pool might explain
regulated proteins grp78 and grp94 (Cuadras et al., 2002; why the attempts to develop a reverse genetic system for ro-
Xu et al., 1998; L. Maruri, unpublished). These findings, taviruses have so far failed; the introduction of exogenous
together with the fact that cells that are poorly susceptible to transcripts that represent rotavirus mRNAs (obtained by in
rotavirus infection increase their susceptibility to the virus vitro synthesis), or the production inside the cell of these
up to 100-fold when subjected to a heat shock (T. Lopez, transcripts from recombinant vectors, might not reach the
unpublished results), suggest that stress proteins might be compartment where such molecules are used as templates for
important for the efficient replication of the virus. The role replication.
C.F. Arias et al. / Virus Research 102 (2004) 43–51 49

7. Perspectives for RNAi in rotavirus research of fluid and electrolyte balance until the infection resolves
(symtomatic treatment), specific interventions aimed at in-
The ability to knock down the expression of individ- hibiting the viral replication could be designed. The intro-
ual rotavirus genes should provide the basis to develop duction of siRNAs preventing the replication of the virus
phenotypic complementation systems in the short term. In (for instance silencing genes involved in the replication of
principle, given the high efficiency of RNAi to prevent the the viral genome) could shorten the duration of the illness
synthesis of a given viral protein, it is conceivable that an al- and possibly help to prevent dehydration, the principal cause
most homologous polypeptide (encoded by a gene having a of death of a rotavirus infection. Another attractive target
few nucleotide differences in the siRNA target site) could be for antiviral therapy could be NSP4, since this nonstructural
synthesized from a recombinant expression vector, to phe- protein, in addition to its important role during virus mor-
notypically rescue the activity of the missing protein. Once phogenesis, has been shown to function as a viral entero-
established, such system should allow the expression of toxin that contributes to the diarrheal illness (Estes, 2003).
modified versions of the protein to dissect its function. In the Although the use of RNAi as an antiviral therapeutic prin-
absence of a reverse genetics system, this approach should be ciple and method still remains to be demonstrated, there is
very useful to carry out functional genomics in rotaviruses, no doubt that this technology will influence drastically the
and in viruses of other genera of the Reoviridae. In addition, way postgenomic viral and cellular research is conducted.
it can be forseen that the studies employing RNAi will soon
extend to the analysis of a number of cellular genes whose
protein products influence the replication of the virus in a Acknowledgements
positive or negative manner, and more in the mid term, to the
analysis of viral gene function in complete animal models. We thank Ulrich Desselberger for critical reading the
One of the limiting steps using RNAi to silence viral manuscript. The excellent technical assistance of Pedro
genes is the siRNA transfection efficiency. Thus, the RNAi Romero is acknowledged. This work was partially sup-
studies, including the phenotypic rescue system described ported by grants 55003662 and 55000613 from the Howard
above, will be greatly improved and simplified if all cells Hughes Medical Institute, and by grant G37621N from the
in a culture could harbor the silencing siRNA. This could National Council for Science and Technology—Mexico.
be achieved by constructing stable cell lines expressing the
siRNA (either as two complementary strands that will hy-
bridize inside the cell, or as precursor short hairpin RNAs; References
Miyagishi et al., 2004), or alternatively, by developing bet-
ter systems to deliver the siRNAs into the cells, for instance, Adelman, Z.N., Sanchez-Vargas, I., Travanty, E.A., Carlson, J.O., Beaty,
using lentiviruses or flavivirus replicons as vectors to direct B.J., Blair, C.D., Olson, K.E., 2002. RNA silencing of dengue virus
the intracellular synthesis of the siRNAs. These approaches type 2 replication in transformed C6/36 mosquito cells transcribing
an inverted-repeat RNA derived from the virus genome. J. Virol. 76,
should be evaluated carefully to avoid the induction of the 12925–12933.
non-specific interferon response by the siRNAs, as has been Arias, C.F., Guerrero, C.A., Mendez, E., Zarate, S., Isa, P., Espinosa,
recently reported (Bridge et al., 2003; Sledz et al., 2003). R., Romero, P., Lopez, S., 2001. Early events of rotavirus infection:
The sequence specificity of RNAi is remarkable; siRNAs the search for the receptor(s). Novartis Found. Symp. 238, 47–60,
that differ from their target sequence in one or more bases, discussion 60–43.
Barik, S., 2004. Control of nonsegmented negative-strand RNA virus
depending on the position of the mismatch, do not efficiently replication by siRNA. Virus Res., this issue.
silence the expression of that gene. This fact has been re- Bitko, V., Barik, S., 2001. Phenotypic silencing of cytoplasmic genes
garded at as a drawback for the use of siRNAs as potential using sequence-specific double-stranded short interfering RNA and its
therapeutic antiviral compounds, since variant viruses could application in the reverse genetics of wild type negative-strand RNA
escape the RNAi response, as has been shown for poliovirus viruses. BMC Microbiol. 1, 34.
Bridge, A.J., Pebernard, S., Ducraux, A., Nicoulaz, A.L., Iggo, R., 2003.
in cell culture (Gitlin et al., 2002). However, based on the Induction of an interferon response by RNAi vectors in mammalian
high specificity of RNAi, an interesting potential applica- cells. Nat. Genet. 34, 263–264.
tion could be the use of siRNAs directed to a specific region Brummelkamp, T.R., Bernards, R., Agami, R., 2002. A system for stable
of a gene, to select viral escape mutants that will contain expression of short interfering RNAs in mammalian cells. Science
nucleotide changes in the specific targeted region. SiRNAs 296, 550–553.
Campagna, M., Vascotto, F., Eichwald, C., Burrone, O.R., 2003. Interfering
could then be used as region-specific mutagens to generate with rotavirus NSP5. In: Proceedings of 8th International Symposium
pre-designed mutants. on Double-Stranded RNA Viruses, Lucca, Italy.
On the other hand, the inhibition of viral gene expres- Caplen, N.J., 2002. A new approach to the inhibition of gene expression.
sion by siRNAs offers the potential for a novel therapeu- Trends Biotechnol. 20, 49–51.
tic approach for infections by rotaviruses and other viruses, Capodici, J., Kariko, K., Weissman, D., 2002. Inhibition of HIV-1 infection
by small interfering RNA-mediated RNA interference. J. Immunol.
once efficient methods for in vivo delivery of siRNAs have 169, 5196–5201.
been developed. In addition to the standard management of Chang, J., Taylor, J.M., 2003. Susceptibility of human hepatitis delta virus
rotavirus infection symptoms, directed towards restoration RNAs to small interfering RNA action. J. Virol. 77, 9728–9731.
50 C.F. Arias et al. / Virus Research 102 (2004) 43–51

Coburn, G.A., Cullen, B.R., 2002. Potent and specific inhibition of human hepatitis B virus replication in vivo by nucleoside analogues and
immunodeficiency virus type 1 replication by RNA interference. J. siRNA. Gastroenterology 125, 9–18.
Virol. 76, 9225–9231. Li, H., Li, W.X., Ding, S.W., 2002. Induction and suppression of RNA
Cuadras, M.A., Feigelstock, D.A., An, S., Greenberg, H.B., 2002. Gene silencing by an animal virus. Science 296, 1319–1321.
expression pattern in Caco-2 cells following rotavirus infection. J. Lichner, Z., Silhavy, D., Burgyan, J., 2003. Double-stranded RNA-binding
Virol. 76, 4467–4482. proteins could suppress RNA interference-mediated antiviral defences.
Dector, M.A., Romero, P., Lopez, S., Arias, C.F., 2002. Rotavirus gene J. Gen. Virol. 84, 975–980.
silencing by small interfering RNAs. EMBO Rep. 3, 1175–1180. Lizano, M., Lopez, S., Arias, C.F., 1991. The amino-terminal half of
Ding, S.W., Li, H., Lu, R., Li, F., Li, W.X., 2004. RNA silencing: a rotavirus SA114fM VP4 protein contains a hemagglutination domain
conserved antiviral immunity of plants and animals. Virus Res., this and primes for neutralizing antibodies to the virus. J. Virol. 65, 1383–
issue. 1391.
Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl, Martinez, M.A., Clotet, B., Este, J.A., 2002. RNA interference of HIV
T., 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference replication. Trends Immunol. 23, 559–561.
in cultured mammalian cells. Nature 411, 494–498. McCaffrey, A.P., Meuse, L., Pham, T.T., Conklin, D.S., Hannon, G.J.,
Estes, M.K., 2001. Rotaviruses and their replication. In: Knipe, D.N., Kay, M.A., 2002. RNA interference in adult mice. Nature 418, 38–
Howley, P.M. (Eds.), Virology, fourth ed. Lippincott Williams and 39.
Wilkins, Philadelphia, PA, pp. 1747–1785. McCaffrey, A.P., Nakai, H., Pandey, K., Huang, Z., Salazar, F.H., Xu, H.,
Estes, M.K., 2003. The rotavirus NSP4 enterotoxin: current status and Wieland, S.F., Marion, P.L., Kay, M.A., 2003. Inhibition of hepatitis
challenges. In: Desselberger, U., Gray, J. (Eds.), Viral Gastroenteritis. B virus in mice by RNA interference. Nat. Biotechnol. 21, 639–
Elsevier Science, Amsterdam, pp. 207–224. 644.
Flores-Jasso, C.F., Valdes, V.J., Sampieri, A., Valadez-Graham, V., Miyagishi, M., Matsumoto, S., Taira, K., 2004. Generation of an RNA
Recillas-Targa, F., Vaca, L., 2004. Silencing structural and nonstruc- expression library against the human genome. Virus Res., this issue.
tural genes in baculovirus by RNA interference. Virus Res., this issue. Novina, C.D., Murray, M.F., Dykxhoorn, D.M., Beresford, P.J., Riess, J.,
Ge, Q., Eisen, H.N., Chen, J., 2004. Use of siRNAs to prevent and treat Lee, S.K., Collman, R.G., Lieberman, J., Shankar, P., Sharp, P.A.,
influenza virus infection. Virus Res., this issue. 2002. siRNA-directed inhibition of HIV-1 infection. Nat. Med. 8,
Ge, Q., McManus, M.T., Nguyen, T., Shen, C.H., Sharp, P.A., Eisen, 681–686.
H.N., Chen, J., 2003. RNA interference of influenza virus production Parashar, U.D., Glass, R.I., 2003. Viral causes of gastroenteritis. In: Des-
by directly targeting mRNA for degradation and indirectly inhibiting selberger, U., Gray, J. (Eds.), Viral Gastroenteritis. Elsevier Science,
all viral RNA transcription. Proc. Natl. Acad. Sci. U.S.A. 100, 2718– Amsterdam, pp. 9–22.
2723.
Patton, J.T., 1995. Structure and function of the rotavirus RNA-binding
Gitlin, L., Andino, R., 2003. Nucleic acid-based immune system: the
proteins. J. Gen. Virol. 76 (Pt 11), 2633–2644.
antiviral potential of mammalian RNA silencing. J. Virol. 77, 7159–
Patton, J.T., 2003. Rotavirus genome replication: sites and signals. In:
7165.
Proceedings of 8th International Symposium on Double-Stranded RNA
Gitlin, L., Karelsky, S., Andino, R., 2002. Short interfering RNA confers
Viruses, Lucca, Italy.
intracellular antiviral immunity in human cells. Nature 418, 430–434.
Patton, J.T., Spencer, E., 2000. Genome replication and packaging
Graff, J.W., Mitzel, D.N., Weisend, C.M., Flenniken, M.L., Hardy, M.E.,
of segmented double-stranded RNA viruses. Virology 277, 217–
2002. Interferon regulatory factor 3 is a cellular partner of rotavirus
225.
NSP1. J. Virol. 76, 9545–9550.
Pesavento, J.B., Estes, M.K., Prasad, B.V.V., 2003. Structural organization
Graham, K.L., Halasz, P., Tan, Y., Hewish, M.J., Takada, Y., Mackow,
of the genome in rotavirus. In: Desselberger, U., Gray, J. (Eds.),
E.R., Robinson, M.K., Coulson, B.S., 2003. Integrin-using rotaviruses
Viral Gastroenteritis, first ed., vol. 9. Elsevier Science, Amsterdam,
bind a2b1 integrin a2 I domain via VP4 DGE sequence and recognize
pp. 115–128.
aXb2 and aVb3 by using VP7 during cell entry. J. Virol. 77, 9969–
9978. Piron, M., Vende, P., Cohen, J., Poncet, D., 1998. Rotavirus RNA-binding
Hall, A.H., Alexander, K.A., 2003. RNA interference of human papil- protein NSP3 interacts with eIF4GI and evicts the poly(A) binding
lomavirus type 18 E6 and E7 induces senescence in HeLa cells. J. protein from eIF4F. EMBO J. 17, 5811–5821.
Virol. 77, 6066–6069. Poruchynsky, M.S., Atkinson, P.H., 1991. Rotavirus protein rearrange-
Hamasaki, K., Nakao, K., Matsumoto, K., Ichikawa, T., Ishikawa, H., ments in purified membrane-enveloped intermediate particles. J. Virol.
Eguchi, K., 2003. Short interfering RNA-directed inhibition of hepatitis 65, 4720–4727.
B virus replication. FEBS Lett. 543, 51–54. Randall, G., Grakoui, A., Rice, C.M., 2003. Clearance of replicating
Hannon, G.J., 2002. RNA interference. Nature 418, 244–251. hepatitis C virus replicon RNAs in cell culture by small interfering
Hu, W.Y., Myers, C.P., Kilzer, J.M., Pfaff, S.L., Bushman, F.D., 2002. RNAs. Proc. Natl. Acad. Sci. U.S.A. 100, 235–240.
Inhibition of retroviral pathogenesis by RNA interference. Curr. Biol. Randall, G., Rice, C.M., 2004. Interfering with hepatitis C virus RNA
12, 1301–1311. replication. Virus Res., this issue.
Jacque, J.M., Triques, K., Stevenson, M., 2002. Modulation of HIV-1 Roth, B.M., Pruss, G.J., Vance, V., 2004. Plant viral suppressors of RNA
replication by RNA interference. Nature 418, 435–438. silencing. Virus Res., this issue.
Jia, Q., Sun, R., 2003. Inhibition of gammaherpesvirus replication by Saleh, M., Van Rij, R.P., Andino, R., 2004. RNA silencing in viral
RNA interference. J. Virol. 77, 3301–3306. infections: insights from poliovirus. Virus Res., this issue.
Jiang, M., Milner, J., 2002. Selective silencing of viral gene expression Sharp, P.A., 2001. RNA interference—2001. Genes Dev. 15, 485–490.
in HPV-positive human cervical carcinoma cells treated with siRNA, Shlomai, A., Shaul, Y., 2003. Inhibition of hepatitis B virus expres-
a primer of RNA interference. Oncogene 21, 6041–6048. sion and replication by RNA interference. Hepatology 37, 764–
Kapadia, S.B., Brideau-Andersen, A., Chisari, F.V., 2003. Interference of 770.
hepatitis C virus RNA replication by short interfering RNAs. Proc. Sledz, C.A., Holko, M., de Veer, M.J., Silverman, R.H., Williams, B.R.,
Natl. Acad. Sci. U.S.A. 100, 2014–2018. 2003. Activation of the interferon system by short-interfering RNAs.
Katze, M.G., He, Y., Gale Jr, M., 2002. Viruses and interferon: a fight Nat. Cell Biol. 9, 834–839.
for supremacy. Nat. Rev. Immunol. 2, 675–687. Tian, P., Estes, M.K., Hu, Y., Ball, J.M., Zeng, C.Q., Schilling, W.P.,
Klein, C., Bock, C.T., Wedemeyer, H., Wustefeld, T., Locarnini, S., Dienes, 1995. The rotavirus nonstructural glycoprotein NSP4 mobilizes Ca2+
H.P., Kubicka, S., Manns, M.P., Trautwein, C., 2003. Inhibition of from the endoplasmic reticulum. J. Virol. 69, 5763–5772.
C.F. Arias et al. / Virus Research 102 (2004) 43–51 51

Tian, P., Hu, Y., Schilling, W.P., Lindsay, D.A., Eiden, J., Estes, M.K., to an endoplasmic reticulum-localized virion component. J. Virol. 72,
1994. The nonstructural glycoprotein of rotavirus affects intracellular 9865–9872.
calcium levels. J. Virol. 68, 251–257. Zamore, P., 2001. RNA interference: listening to the sound silence. Nat.
Valdes, V.J., Sampieri, A., Sepulveda, J., Vaca, L., 2003. Using Struct. Biol. 8, 746–750.
double-stranded RNA to prevent in vitro and in vivo viral infections Zárate, S., Espinosa, R., Romero, P., Arias, C.F., López, S., 2002. Ro-
by recombinant baculovirus. J. Biol. Chem. 278, 19317–19324. tavirus interacts with avb3 through a novel binding site. In: Proceed-
Vance, V., Vaucheret, H., 2001. RNA silencing in plants—defense and ings of Meeting of the Howard Hughes Medical Institute International
counterdefense. Science 292, 2277–2280. Research Scholars, Palm Cove, Australia.
Wilson, J.A., Jayasena, S., Khvorova, A., Sabatinos, S., Rodrigue-Gervais, Zhang, R., Guo, Z., Lu, J., Meng, J., Zhou, C., Zhan, X., Huang, B.,
I.G., Arya, S., Sarangi, F., Harris-Brandts, M., Beaulieu, S., Richard- Yu, X., Huang, M., Pan, X., Ling, W., Chen, X., Wan, Z., Zheng,
son, C.D., 2003. RNA interference blocks gene expression and RNA H., Yan, X., Wang, Y., Ran, Y., Liu, X., Ma, J., Wang, C., Zhang,
synthesis from hepatitis C replicons propagated in human liver cells. B., 2003. Inhibiting severe acute respiratory syndrome-associated
Proc. Natl. Acad. Sci. U.S.A. 100, 2783–2788. coronavirus by small interfering RNA. Chin. Med. J. 116, 1262–
Xu, A., Bellamy, A.R., Taylor, J.A., 1998. BiP (GRP78) and endoplasmin 1264.
(GRP94) are induced following rotavirus infection and bind transiently
Virus Research 102 (2004) 53–58

Control of HIV-1 replication by RNA interference


Nan Sook Lee, John J. Rossi∗
Division of Molecular Biology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA

Abstract

Small interfering RNAs (siRNAs) have been shown to direct sequence-specific inhibition of gene expression in mammalian cells. siRNAs
are RNA duplexes of 21–23 nucleotides (nts) with ∼2 nt 3 overhangs that can induce degradation of their homologous target mRNAs without
interferon responses in mammalian cells. The degradation of the target occurs at the post-transcriptional level, meaning a post-transcriptional
gene silencing (PTGS) mechanism called as RNA interference (RNAi). RNAi has emerged as an efficient method to inhibit gene expression
in mammalian cells with increasingly successful cases of knockdown of many specific genes. Recent works have shown that the use of
RNAi could inhibit HIV-1 replication by targeting viral or cellular genes. RNAi can be considered as a gene-specific therapeutic option for
controlling HIV-1 replication. However, the control of HIV-1 replication has become complex because of the limited effectiveness of existing
anti-HIV-1 agents and the high speed mutation rate of the HIV-1 genome. Careful assessments are required for the potential of RNAi as a
gene therapy approach for controlling HIV-1 replication. This review will discuss the status of the science using RNAi for controlling HIV-1
replication and will describe possible problems for therapeutic applications of RNAi-mediated technologies for HIV-1 behind this novel
mechanism.
© 2004 Elsevier B.V. All rights reserved.

Keywords: RNAi; siRNA; shRNA; HIV-1; PTGS

1. Introduction quence to the silenced gene (Elbashir et al., 2001; Fire et al.,
1998; Hutvagner and Zamore, 2002; Sharp, 2001; Zamore
Gene therapy has gained consideration as a possible treat- et al., 2000). RNAi is initiated by the dsRNA-specific en-
ment for acquired immunodeficiency syndrome (AIDS), ei- donuclease Dicer that promotes processive cleavage of long
ther as an alternative or as an addition to anti-retroviral dsRNA into small 21–25 nucleotide (nt) small interfering
chemotherapy. Several types of RNA gene therapies have RNAs (siRNAs) (Bernstein et al., 2001; Hutvagner et al.,
been developed and shown to inhibit HIV-1 replication in 2001; Ketting et al., 2001; Knight and Bass, 2001). The du-
mammalian cell cultures; these include antisense RNA, cat- plex siRNA is unwound, and one of the two strands is in-
alytic RNA (ribozyme) and high-affinity RNA ligands (ap- corporated into an RNA-induced silencing complex (RISC)
tamers or decoys). Several of these RNA-based approaches (Bernstein et al., 2001; Hammond et al., 2001) guiding it
have been safety tested in phase I clinical trials, although to a homologous target mRNA, which in turn is cleaved by
there are no reports yet of efficacy trials. Nevertheless, in an endonuclease in RISC. This powerful sequence-specific
vitro data suggest that their anti-viral efficiencies are vari- knockdown mechanism has rapidly gained wide acceptance
able and can often be overcome by increasing the multi- as a surrogate genetic tool and possible therapeutic modality
plicity of HIV infectious particles. The limitations have led in mammalian cells.
several investigators to explore the potential of RNA inter- It is known that dsRNA of ≥30 bp in size can trigger in-
ference (RNAi) as an anti-HIV therapy. terferon responses in mammalian cells that are intrinsically
RNAi is the process of sequence-specific, post-transcrip- sequence-nonspecific to the inducing dsRNA (Paddison
tional gene silencing (PTGS) in animals and plants triggered et al., 2002b). However, introduction of shorter siRNAs
by double-stranded RNA (dsRNA) that is homologous in se- (<30 bp) into mammalian cells leads to mRNA degrada-
tion with sequence specificity without activating the inter-
feron response (Elbashir et al., 2001). Synthetic duplexes
∗ Corresponding author. Tel.: +1-626-301-8360. of 21-nucleotide siRNAs with protruding 3 termini can
E-mail address: jrossi@coh.org (J.J. Rossi). mediate RNAi in a sequence-specific manner in cultured

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.015
54 N.S. Lee, J.J. Rossi / Virus Research 102 (2004) 53–58

mammalian cells. This siRNA technology takes advantage 2. Targeting HIV-1 with RNAi
of the fact that RNAi is a natural biological mechanism for
silencing genes in most eukaryotic organisms ranging from Introduction of siRNAs specific for HIV-1 into mam-
fission yeast through plants and mammals (Agami, 2002; malian cells could lead to viral RNA degradation and in-
Cottrell and Doering, 2003; Paddison and Hannon, 2002; hibition of HIV-1 gene expression and replication during
Scherr et al., 2003). RNAi functions as an antiviral mech- different stages of the viral life cycle (Fig. 1). HIV-1 has
anism in some organisms such as Drosophila and plants, ∼9 kbp genome that contains nine genes encoding 15 pro-
although the role of RNAi in controlling viral infection in teins (Greene and Peterlin, 2002). The first potential tar-
mammals is unknown. siRNA-mediated PTGS, therefore, get for siRNAs is the viral genomic RNA upon viral en-
offers a potentially powerful tool for inhibiting HIV repli- try and uncoating. At least one report demonstrates that
cation. This mechanism can be targeted to both viral and siRNA-mediated destruction of incoming HIV-1 can take
cellular transcripts. place (Jacque et al., 2002) although other studies of RNAi

Fig. 1. HIV-1 life cycle and potential intervention by RNAi. HIV-1 utilizes the CD4 receptor and the chemokine receptors CCR5/and or CXCR4 for
entry. Each of these receptors is a potential RNAi target to block viral entry. In lieu of targeting the receptor or co-receptor mRNAs, incoming genomic
viral RNA is a potential target as well, as are all classes of viral transcripts. RISC is the RNAi induced silencing complex that interacts with one of
the siRNA strands and positions the strand for complementary base pairing with the target RNA. A component of RISC endonucleolytically cleaves the
target RNA allowing the RISC complex to recycle.
N.S. Lee, J.J. Rossi / Virus Research 102 (2004) 53–58 55

inhibition of retroviral infection suggested that incoming ge- Lee et al., 2002a) (Fig. 2). The expression of siRNAs
nomic RNA may not be accessible to siRNAs (Hu et al., from psiRNAs appears to bypass the need for Dicer or
2002b; Waterhouse et al., 2001). Once the viral genomic other endonucleases to produce siRNAs (Lee et al., 2002a;
DNA has been integrated, the viral mRNA transcripts as Miyagishi and Taira, 2002). The same is true for ssiR-
well as the unspliced genomic length RNA are targets for NAs, but these must be used in multiple transfections to
RNAi. Early transcripts such as HIV-1 rev and tat are good maintain silencing. The psiRNA system can potentially be
targets for siRNAs since the Tat and Rev proteins encoded used in differentiated mammalian cells containing no or
by these RNAs are essential for subsequent expression of low amounts of Dicer (Kitabwalla and Ruprecht, 2002).
HIV-1 structural genes (Gag, Pol, and Env) and for the syn- However, shRNAs with different stem-loop structures ex-
thesis of full length viral genomic RNA. pressed from pshRNAs in mammalian cells require pro-
In addition to viral targets, the cellular chemokine recep- cessing to remove the loop. Depending upon the length
tors CCR5 and CXCR4, which function as co-receptors for of the duplex, these may utilize Dicer, but perhaps one or
HIV-1, have provided new therapeutic targets and a better more other endonucleases are involved in this processing
understanding of the progression of viral infection. CCR5, step. (Brummelkamp et al., 2002; Parrish et al., 2000; Paul
an HIV-1 co-receptor for M-tropic HIV-1 provides an attrac- et al., 2002; Sui et al., 2002; Yu et al., 2002; Zeng et al.,
tive cellular target for siRNAs since homozygous deletions 2002). The extent of siRNA-directed silencing from both
of CCR5 effectively confer protection from HIV-1 without psiRNAs and pshRNAs may be limited by the concentration
any serious deleterious effects in immune function (Samson of active siRNAs in the target cell. The stem-loop struc-
et al., 1996). At least one group has taken advantage of this tures of shRNAs mimic the naturally occurring stem-loop
target for RNAi mediated gene silencing demonstrating in structures found in micro-RNA precursors which are pro-
vitro knockdown of CCR5 by siRNAs provided marked pro- cessed by Dicer to yield small temporal RNAs (stRNA) or
tection from HIV-1 infection (Martinez et al., 2002). microRNA (miRNAs) (Lee et al., 2002b). Processing of
anti-HIV siRNAs from a miRNA precursor has recently
been demonstrated (Zeng and Cullen, 2002).
3. Exogenous delivery versus endogenous expression of Inhibition of HIV-1 infection using ssiRNAs, or siRNAs
si/shRNAs produced from transcription of psiDNAs and pshDNAs has
served as a proof-of-principle that siRNA technology can be
In mammalian systems, the sequence-specific anti-HIV-1 a possible therapeutic approach for the inhibition of HIV-1
effects have been observed following introduction of syn- replication in hematopoietic cells (Coburn and Cullen, 2002;
thetic siRNAs (ssiRNAs) via transfection into HIV-1 in- Hu et al., 2002a; Jacque et al., 2002; Lee et al., 2002a;
fectable cells (Coburn and Cullen, 2002; Gitlin et al., 2002; Novina et al., 2002). We have demonstrated that a psiRNA
Jacque et al., 2002; Novina et al., 2002), or via endogenous approach can be used to inhibit expression of HIV-1 rev
expression of 21–23 nt transcripts (psiRNAs) or hairpin and/or tat transcript (s) in both transient transfections (Lee
RNAs (pshRNAs) from DNA plasmids (Jacque et al., 2002; et al., 2002a) or from lentiviral transduced hematopoietic

Fig. 2. Gene expression systems for intracellular transcription of siRNAs or shRNAs. For purposes of simplicity, only the Pol III promoted transcripts
are depicted, but Pol II systems can be used as well provided that a polyA signal is included downstream of the shRNA genes. The Pol III transcripts
terminate within a string of 5–6 uridines providing a defined 3 end for the transcripts. When sense and antisense strands are transcribed from separate
promoters, these strands must anneal to form an siRNA. In contrast, linked shRNA strands readily form a duplex, but the loop joining these must be
processed to generate siRNAs.
56 N.S. Lee, J.J. Rossi / Virus Research 102 (2004) 53–58

progenitor cells (Banerjea et al., 2003). In this approach, can effectively transduce both dividing and non-dividing
the vectors contain two tandem human U6 snRNA promot- cells and provide long-term gene expression (Abbas-Terki
ers followed by 21-mers encoding sense and antisense siR- et al., 2002; Matta et al., 2003; Qin et al., 2003; Rubinson
NAs. In the co-transfection experiments, psiRNAs cotrans- et al., 2003; Tiscornia et al., 2003). Anti-CCR5-shRNA
fected with the HIV-1 pNL4-3 proviral DNA led to up to genes inserted in a lentiviral vector and introduced into hu-
4 logs of inhibition of HIV-1 p24 antigen expression (Lee man peripheral blood T lymphocytes (PBLs) resulted in a
et al., 2002a). The high degree of inhibition was achieved 10-fold reduction of CCR5 in PBLs and a five-fold reduc-
by simultaneously targeting two essential sites (rev and tat). tion of HIV-1 replication. This protection was long term
It was also demonstrated that synthetic siRNAs targeted to and sequence-specific (Qin et al., 2003). It has also been
HIV-1 rev and tat mRNAs inhibit HIV-1 gene expression demonstrated that anti-Rev siRNA genes expressed from
and replication in human T-cell lines and primary lympho- lentiviral vector-transduced hematopoietic precursor cells
cytes (Coburn and Cullen, 2002). Novina et al. (2002) re- provided in vitro derived macrophages and SCID-hu mouse
ported that silencing CD4 expression inhibited viral entry, differentiated T-lymphocytes with long term protection
syncytia formation and reduced free viral titers, although from HIV-1 infection and replication (Banerjea et al., 2003).
CD4 may not be a good therapeutic target because of the It should be pointed out that long term RNAi effected by
receptor’s critical role in T-cell function. ssiRNAs may also be possible in certain cell types. For in-
Gitlin et al. (2002) reported that siRNA against poliovirus stance non-dividing macrophages were shown to be resistant
provided intracellular anti-viral immunity to human cells, to HIV-1 challenge for over two weeks following treatment
promoting viral clearance and cell survival. They suggested with ssiRNAs targeting CCR5(Song et al., 2003). Interest-
that gene silencing by siRNAs functions as an adaptive, ingly, these same investigators demonstrated that the RNAi
RNA-based defense mechanism in mammalian cells. Jacque effect is more long-lived if there is a target RNA present
et al. (2002) reported similar results in their studies with since RNAi was lost in cells that were not continually chal-
HIV-1. They utilized intracellular T7 transcription from a lenged with viral gene expression.
plasmid DNA template to produce siRNAs in the cyto-
plasm of mammalian cells. Utilizing this system to produce
shRNAs targeting HIV-1 vif they were able to confer intra- 4. Possible problems for therapeutic applications
cellular immunity to HIV-1 in cell lines as well as primary
lymphcytes, in part by blocking reverse transcription of ge- While the results obtained to date should be considered
nomic RNA into proviral cDNA. Capodici et al. (2002) used preliminary in terms of application to humans, they do pro-
synthetic and in vitro transcribed siRNAs and obtained sim- vide strong justification for further investigating the use of
ilar suppression of HIV-1 specific proviral DNA formation RNAi for treatment of HIV-1 in a clinical setting. It has
and replication in cell lines and primary, activated CD4+ T largely been assumed that siRNAs appear to be under the
lymphocytes. In addition to demonstrating that siRNA can radar of the host interference response mechanism. It has
inhibit viral infection at pre- and post-integration stages in recently been demonstrated that some expressed shRNAs
mammalian cells, they also showed that primary CD4+ T can activate at least one of the arms of the human inter-
cells could be targeted by siRNAs using lipofectin, or by feron response mechanism (Bridge et al., 2003). Since many
non-lipid complexed fluorine-derivatized siRNAs. commonly used tumor cells cannot respond to interferon be-
Although transfected siRNAs can effectively inhibit viral cause of specific mutations of genes in the interferon path-
replication, the effects are largely transient (Novina et al., way (Stojdl et al., 2000), careful studies with primary cells
2002). Therefore, even if effective in vivo delivery is ob- need to be carried out to ensure that potentially serious side
tained, the siRNAs will have to be infused on a regular effects due to interferon induction are not occurring.
basis to treat chronic diseases such as AIDS. The use of An additional concern is that the success of RNAi against
engineered DNA vectors capable of long term production a viral pathogen is not automatically assured. One impor-
of siRNAs or shRNAs has facilitated the practical applica- tant factor is that not all viral mRNA sequences are equally
tion of RNAi in human primary cells (Brummelkamp et al., accessible to siRNAs. Upon HIV-1 infection, genomic viral
2002; Lee et al., 2002a; Miyagishi and Taira, 2002; RNA is introduced into the host cell cytoplasm in the form
Paddison et al., 2002a; Paul et al., 2002; Xia et al., 2002). of a nucleoprotein complex that can obscure the recogni-
The demonstrations that RNAi could be activated by ex- tion by siRNAs. Some viral RNA sequences might be buried
pressing siRNAs or shRNAs from eukaryotic promoters has within secondary structures or within highly folded regions.
prompted the inclusion of these expression systems into vi- It may often be necessary to test several different target sites
ral vectors that can be used to transduce cells with siRNA or before a potent RNAi/target combination can be identified.
shRNA genes. For treatment of HIV-1 infection, lentiviral For HIV-1 there is the compounding problem of rapid muta-
vector-based transduction of siRNA and shRNA encoding tion, rendering the virus resistant to therapeutic agents. Us-
genes into hematopoietic cells is particularly attractive from ing RNAi agents directed against multiple conserved RNA
the experimental point of view. Lentiviruses are effective target sequences in combination with targeting host factors
for delivery of siRNAs into mammalian cells because they may overcome this problem. Another concern is maintaining
N.S. Lee, J.J. Rossi / Virus Research 102 (2004) 53–58 57

siRNA expression long term in stably transduced primary Hu, W.Y., Myers, C.P., Kilzer, J.M., Pfaff, S.L., Bushman, F.D., 2002b.
cells. It is currently unknown whether or not silencing of Inhibition of retroviral pathogenesis by RNA interference. Curr. Biol.
12, 1301–1311.
Pol III-promoted siRNA and shRNA transcription will be a Hutvagner, G., McLachlan, J., Pasquinelli, A.E., Balint, E., Tuschl, T.,
problem. Finally, since RNAi is a native cellular mechanism Zamore, P.D., 2001. A cellular function for the RNA-interference
that most likely has an important, if not essential function enzyme Dicer in the maturation of the let-7 small temporal RNA.
in most cell types, there is the concern that usurping the Science 293, 834–838.
RNAi machinery with foreign siRNAs could have adverse Hutvagner, G., Zamore, P.D., 2002. RNAi: nature abhors a double-strand.
Curr. Opin. Genet. Dev. 12, 225–232.
developmental and functional consequences. Only by car- Jacque, J.M., Triques, K., Stevenson, M., 2002. Modulation of HIV-1
rying out the appropriate experiments in relevant cells and replication by RNA interference. Nature 418, 435–438.
animal models can the potential problems be addressed. Ketting, R.F., Fischer, S.E., Bernstein, E., Sijen, T., Hannon, G.J., Plasterk,
R.H., 2001. Dicer functions in RNA interference and in synthesis of
small RNA involved in developmental timing in C. elegans. Genes
Dev. 15, 2654–2659.
Acknowledgements Kitabwalla, M., Ruprecht, R.M., 2002. RNA interference—a new weapon
against HIV and beyond. N. Engl. J. Med. 347, 1364–1367.
This work was supported by NIH grants AI 29329, Knight, S.W., Bass, B.L., 2001. A role for the RNase III enzyme DCR-1
AI42552, and HL074704 to JJR and a GlaxoSmith Kline in RNA interference and germ line development in Caenorhabditis
elegans. Science 293, 2269–2271.
(GSK) grant to NSL.
Lee, N.S., Dohjima, T., Bauer, G., Li, H., Li, M.J., Ehsani, A., Salvaterra,
P., Rossi, J., 2002a. Expression of small interfering RNAs targeted
against HIV-1 rev transcripts in human cells. Nat. Biotechnol. 20,
500–505.
References Lee, Y., Jeon, K., Lee, J.T., Kim, S., Kim, V.N., 2002b. MicroRNA
maturation: stepwise processing and subcellular localization. EMBO
Abbas-Terki, T., Blanco-Bose, W., Deglon, N., Pralong, W., Aebischer, J. 21, 4663–4670.
P., 2002. Lentiviral-mediated RNA interference. Hum. Gene. Ther. 13, Martinez, M.A., Gutierrez, A., Armand-Ugon, M., Blanco, J., Parera, M.,
2197–2201. Gomez, J., Clotet, B., Este, J.A., 2002. Suppression of chemokine
Agami, R., 2002. RNAi and related mechanisms and their potential use receptor expression by RNA interference allows for inhibition of
for therapy. Curr. Opin. Chem. Biol. 6, 829–834. HIV-1 replication. AIDS 16, 2385–2390.
Banerjea, A., Li, M.J., Bauer, G., Remling, L., Lee, N.S., Rossi, J., Akkina, Matta, H., Hozayev, B., Tomar, R., Chugh, P., Chaudhary, P.M., 2003.
R., 2003. Inhibition of HIV-1 by lentiviral vector-transduced siRNAs in Use of lentiviral vectors for delivery of small interfering RNA. Cancer
T lymphocytes differentiated in SCID-hu mice and CD34+ progenitor Biol. Ther. 2, 206–210.
cell-derived macrophages. Mol. Ther. 8, 62–71. Miyagishi, M., Taira, K., 2002. U6 promoter-driven siRNAs with four
Bernstein, E., Caudy, A.A., Hammond, S.M., Hannon, G.J., 2001. Role uridine 3 overhangs efficiently suppress targeted gene expression in
for a bidentate ribonuclease in the initiation step of RNA interference. mammalian cells. Nat. Biotechnol. 20, 497–500.
Nature 409, 363–366. Novina, C.D., Murray, M.F., Dykxhoorn, D.M., Beresford, P.J., Riess, J.,
Bridge, A.J., Pebernard, S., Ducraux, A., Nicoulaz, A.L., Iggo, R., 2003. Lee, S.K., Collman, R.G., Lieberman, J., Shankar, P., Sharp, P.A.,
Induction of an interferon response by RNAi vectors in mammalian 2002. siRNA-directed inhibition of HIV-1 infection. Nat. Med. 8,
cells. Nat. Genet. 34, 263–264. 681–686.
Brummelkamp, T.R., Bernards, R., Agami, R., 2002. A system for stable Paddison, P.J., Caudy, A.A., Bernstein, E., Hannon, G.J., Conklin, D.S.,
expression of short interfering RNAs in mammalian cells. Science 2002a. Short hairpin RNAs (shRNAs) induce sequence-specific silenc-
296, 550–553. ing in mammalian cells. Genes Dev. 16, 948–958.
Capodici, J., Kariko, K., Weissman, D., 2002. Inhibition of HIV-1 infection Paddison, P.J., Caudy, A.A., Hannon, G.J., 2002b. Stable suppression of
by small interfering RNA-mediated RNA interference. J. Immunol. gene expression by RNAi in mammalian cells. Proc. Natl. Acad. Sci.
169, 5196–5201. U.S.A. 99, 1443–1448.
Coburn, G.A., Cullen, B.R., 2002. Potent and specific inhibition of human Paddison, P.J., Hannon, G.J., 2002. RNA interference: the new somatic
immunodeficiency virus type 1 replication by RNA interference. J. cell genetics. Cancer Cell 2, 17–23.
Virol. 76, 9225–9231. Parrish, S., Fleenor, J., Xu, S., Mello, C., Fire, A., 2000. Functional
Cottrell, T.R., Doering, T.L., 2003. Silence of the strands: RNA interfer- anatomy of a dsRNA trigger: differential requirement for the two
ence in eukaryotic pathogens. Trends Microbiol. 11, 37–43. trigger strands in RNA interference. Mol. Cell 6, 1077–1087.
Elbashir, S.M., Lendeckel, W., Tuschl, T., 2001. RNA interference is Paul, C.P., Good, P.D., Winer, I., Engelke, D.R., 2002. Effective expression
mediated by 21- and 22-nucleotide RNAs. Genes Dev. 15, 188–200. of small interfering RNA in human cells. Nat. Biotechnol. 20, 505–
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello, 508.
C.C., 1998. Potent and specific genetic interference by double-stranded Qin, X.F., An, D.S., Chen, I.S., Baltimore, D., 2003. Inhibiting HIV-1
RNA in Caenorhabditis elegans. Nature 391, 806–811. infection in human T cells by lentiviral-mediated delivery of small
Gitlin, L., Karelsky, S., Andino, R., 2002. Short interfering RNA confers interfering RNA against CCR5. Proc. Natl. Acad. Sci. U.S.A. 100,
intracellular antiviral immunity in human cells. Nature 418, 430–434. 183–188.
Greene, W.C., Peterlin, B.M., 2002. Charting HIV’s remarkable voyage Rubinson, D.A., Dillon, C.P., Kwiatkowski, A.V., Sievers, C., Yang, L.,
through the cell: Basic science as a passport to future therapy. Nat. Kopinja, J., Rooney, D.L., Ihrig, M.M., McManus, M.T., Gertler,
Med. 8, 673–680. F.B., Scott, M.L., Van Parijs, L., 2003. A lentivirus-based system to
Hammond, S.M., Boettcher, S., Caudy, A.A., Kobayashi, R., Hannon, G.J., functionally silence genes in primary mammalian cells, stem cells and
2001. Argonaute2, a link between genetic and biochemical analyses transgenic mice by RNA interference. Nat. Genet. 33, 401–406.
of RNAi. Science 293, 1146–1150. Samson, M., Libert, F., Doranz, B.J., Rucker, J., Liesnard, C., Farber,
Hu, W., Myers, C., Kilzer, J., Pfaff, S., Bushman, F., 2002a. Inhibition C.M., Saragosti, S., Lapoumeroulie, C., Cognaux, J., Forceille, C.,
of retroviral pathogenesis by RNA interference. Curr. Biol. 12, 1301. Muyldermans, G., Verhofstede, C., Burtonboy, G., Georges, M., Imai,
58 N.S. Lee, J.J. Rossi / Virus Research 102 (2004) 53–58

T., Rana, S., Yi, Y., Smyth, R.J., Collman, R.G., Doms, R.W., Vassart, pression in mammalian cells. Proc. Natl. Acad. Sci. U.S.A. 99, 5515–
G., Parmentier, M., 1996. Resistance to HIV-1 infection in caucasian 5520.
individuals bearing mutant alleles of the CCR-5 chemokine receptor Tiscornia, G., Singer, O., Ikawa, M., Verma, I.M., 2003. A general method
gene. Nature 382, 722–725. for gene knockdown in mice by using lentiviral vectors expressing
Scherr, M., Morgan, M.A., Eder, M., 2003. Gene silencing mediated by small interfering RNA. Proc. Natl. Acad. Sci. U.S.A. 100, 1844–1848.
small interfering RNAs in mammalian cells. Curr. Med. Chem. 10, Waterhouse, P.M., Wang, M.B., Lough, T., 2001. Gene silencing as an
245–256. adaptive defence against viruses. Nature 411, 834–842.
Sharp, P.A., 2001. RNA interference—2001. Genes Dev. 15, 485– Xia, H., Mao, Q., Paulson, H.L., Davidson, B.L., 2002. siRNA-mediated
490. gene silencing in vitro and in vivo. Nat. Biotechnol. 20, 1006–1010.
Song, E., Lee, S.K., Dykxhoorn, D.M., Novina, C., Zhang, D., Crawford, Yu, J.Y., DeRuiter, S.L., Turner, D.L., 2002. RNA interference by ex-
K., Cerny, J., Sharp, P.A., Lieberman, J., Manjunath, N., Shankar, P., pression of short-interfering RNAs and hairpin RNAs in mammalian
2003. Sustained small interfering RNA-mediated human immunode- cells. Proc. Natl. Acad. Sci. U.S.A. 99, 6047–6052.
ficiency virus type 1 inhibition in primary macrophages. J. Virol. 77, Zamore, P.D., Tuschl, T., Sharp, P.A., Bartel, D.P., 2000. RNAi:
7174–7181. double-stranded RNA directs the ATP-dependent cleavage of mRNA
Stojdl, D.F., Lichty, B., Knowles, S., Marius, R., Atkins, H., Sonenberg, at 21 to 23 nucleotide intervals. Cell 101, 25–33.
N., Bell, J.C., 2000. Exploiting tumor-specific defects in the interferon Zeng, Y., Cullen, B.R., 2002. RNA interference in human cells is restricted
pathway with a previously unknown oncolytic virus. Nat. Med. 6, to the cytoplasm. RNA 8, 855–860.
821–825. Zeng, Y., Wagner, E.J., Cullen, B.R., 2002. Both natural and designed
Sui, G., Soohoo, C., Affar el, B., Gay, F., Shi, Y., Forrester, W.C., micro RNAs can inhibit the expression of cognate mRNAs when
2002. A DNA vector-based RNAi technology to suppress gene ex- expressed in human cells. Mol. Cell 9, 1327–1333.
Virus Research 102 (2004) 59–64

RNA interference against retroviruses


Wen-Yuan Hu, Frederic D. Bushman∗ , Amara C. Siva
Infectious Disease Laboratory, The Salk Institute, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA

Abstract

Bang and Ellerman, and later Peyton Rous, reported the first identification of transmissible cancer-causing agents, which later turned out to
be avian retroviruses. Today avian retroviruses are important models for study of retrovirus replication and pathogenesis, and also important
pathogens of domestic fowl. Here we describe the use of RNA interference (RNAi) in live chick embryos to block replication of an avian
retrovirus. We also describe inhibition of ASLV and HIV replication in cell culture with RNAi.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Retrovirus; RNA interference; siRNA; ASLV; HIV

1. Introduction natively, RNAi can also be induced by direct transfection of


siRNAs, short RNAs mimicking the product of Dicer pro-
1.1. RNAi background cessing.
The siRNAs are incorporated into a multicomponent RNA
RNA interference (RNAi) is a process in which induced silencing complex (RISC), and act as guides for the
double-stranded RNA (dsRNA) triggers the silencing of targeting of mRNAs for cleavage and degradation (Martinez
gene expression in a sequence-specific manner. RNAi has et al., 2002a; Schwarz et al., 2002). The identities of all
been observed in organisms as diverse as plants, protozoa, RISC components have not been characterized to date. At
nematodes, fungi, insects, and mammals (Caplen et al., least one of the members of the Argonaute gene family is a
2000, 2001; Cogoni and Macino, 1999; Elbashir et al., component of this complex (for review, see Carmell et al.,
2001b; Fire et al., 1998; Hamilton and Baulcombe, 1999; 2002), and a micrococcal nuclease family member has been
Hammond et al., 2000; Jorgensen, 1990; Ngo et al., 1998; also been identified that may be the enzyme responsible for
Romano and Macino, 1992). Some of the components of mRNA degradation (Caudy et al., 2003).
the RNAi machinery are also involved in the processing In invertebrates, the RNA silencing machinery appears to
and function of microRNAs (miRNAs; also called small be able to amplify the degradation signal. A strand from an
temporal RNAs), a class of small endogenous noncoding siRNA bound to the target mRNA can apparently function
RNA molecules that carry out developmental gene control as a primer that extends a complementary strand along the
via translational repression (Doench et al., 2003; Palatnik targeted mRNA by an RNA-dependent RNA polymerase
et al., 2003; Zeng et al., 2000, 2003). (RdRP). Further processing of the dsRNA generated results
RNAi can be induced by introduction of dsRNA into in generation of new siRNAs (Lipardi et al., 2001; Plasterk,
cells in a variety of ways. These include endogenous syn- 2002; Sijen et al., 2001). However, such amplification has
thesis of complementary RNA strands, by infection with not been observed in mammalian cells to date.
viruses that produce dsRNA, and injection or transfection
of dsRNAs. Long dsRNA are processed to small interfering 1.2. RNAi in vertebrate cells
RNAs (siRNAs) of about 21–23 nt by the RNase III en-
zyme Dicer (for recent reviews of the RNAi mechanism, see Prior to 2001, there appeared to be little chance that
Dykxhoorn et al., 2003; Pickford and Cogoni, 2003). Alter- RNAi could be employed to inhibit the replication of ver-
tebrate viruses. Vertebrate cells were known to respond to
double-stranded RNA by inducing the interferon response.
∗ Corresponding author. Cells respond to interferon combined with a double-stranded
E-mail address: bushman@salk.edu (F.D. Bushman). RNA inducer by shutting down translation and other

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.016
60 W.-Y. Hu et al. / Virus Research 102 (2004) 59–64

responses designed to block viral replication and so block and Rossi in this volume), then studies of RNAi against
spread in the organism. However, key findings in the labo- avian retroviruses. These studies show that RNAi can inhibit
ratories of Tuschl and Fire revealed that the length of the viral replication in vivo and in cell culture, but leave the
double-stranded RNA determined the nature of the response question of its normal role unanswered. Regardless of its
(Caplen et al., 2001; Elbashir et al., 2001a). Long dsRNA normal function, it seems likely that RNAi can be adapted
induced the interferon response, while short dsRNAs in- to mitigate viral pathogenesis in vertebrates.
duced RNAi. This opened the door to studies of RNAi
against vertebrate viruses.
RNAi has been found to act as an antiviral response in 2. RNA interference against HIV in cell culture
plants. Plants inhibited for the RNAi response show exac-
erbated symptoms upon infection with some viruses. Fur- Inhibition of HIV-1 replication in cell culture was first re-
thermore, several plant viruses have been shown to encode ported by Garrus et al. (2001). In this study, Tsg101, a pro-
proteins that antagonize the RNAi system, providing strong tein involved in vacuolar protein sorting, was shown to be
evidence that RNAi is part of the normal plant response to required for HIV-1 budding through the targeting of Tsg101
infection (Al-Kaff et al., 1998; Marathe et al., 2000; Ratcliff mRNA by RNAi (Garrus et al., 2001). This beautiful in-
et al., 1999). Recently a protein inhibiting RNAi has been vestigation launched an explosion of studies using RNAi to
detected in Flock House Virus (FHV), a nodavirus that in- inhibit HIV replication (summarized in Table 1).
fects insects, implicating RNAi in defense against viruses in Many positions in the HIV genomic RNA have been tar-
insects as well (Li et al., 2002). The FHV B2 protein, the geted by siRNAs, including the gag and pol regions (encod-
inhibitor of RNAi, has been found to block RNA silencing ing viral structural proteins), and the genes for the regula-
in both plant and invertebrate cells, and can functionally re- tory proteins tat and rev. Cellular factors required for viral
place the suppressor of RNA silencing 2b protein of cucum- replication in addition to the Tsg101 coding region have also
ber mosaic virus (Li et al., 2002). been targeted, specifically the HIV receptor CD4 and the
Is RNAi a component of the normal vertebrate response coreceptors CXCR4 and CCR5 (references in Table 1).
to viral infection? This question is still unanswered. We and Several methods were used to initiate RNAi in these stud-
others have investigated this question by modeling inhibition ies. Some of these techniques in fact were first used in the
of viral replication by RNAi using retroviral models. This HIV model system, and each suggests a possible approach
review focuses on studies of avian retroviruses, for which we for using RNAi to treat HIV infection. Transfection of cells
have an in vivo model. Many publications have also reported with chemically synthesized siRNA was first used to de-
studies of inhibition of HIV. Below we first summarize the liver RNAi, but disadvantages of this method include that
HIV work (see also Bushman, 2003, and the article by Lee the RNAi effect is transient, and many primary cells are not

Table 1
Studies reporting inhibition of retroviral replication by RNAi
Target gene Means of inducing RNAi Cell line or tissue Reference

Viral gene
HIV-1 Tat, Rev siRNA transfection, siRNA electroporation 293T, Jurkat, PBMC Coburn and Cullen (2002)
HIV-1 Vif siRNA transfection Magi cells Jacque et al. (2002)
Nef, LTR TAR T7-shRNA (vector) transfection PBL
HIV-1 Gag, Pol siRNA transfection HOS Hu et al. (2002)
HIV-1 Gag, LTR siRNA (synthetic and transcribed from 293T, U87, PBMC Capodici et al. (2002)
T7 in vitro) transfection
HIV-1 Rev, Tat siRNA transfection, tandem U6-siRNA 293/EcR Lee et al. (2002)
(vector) transfection
HIV-1 Rev Tandem U6-siRNA (vector) transfection CD34+ hematopoetic Banerjea et al. (2003)
progenitor cells
HIV-1 Gag, Env Long dsRNA transfection COS, HeLa, PBMC Park et al. (2002)
HIV-1 Tat, RT, human siRNA transfection Magi cells, Jurkat cells Surabhi and Gaynor (2002)
NF-␬B p65 subunit
HIV-1 Gag, human CD4 siRNA transfection Magi cells Novina et al. (2002)
ASLV Gag siRNA transfection, siRNA in ovo DF1, chicken embryo Hu et al. (2002)
electroporation
Host genes
Human TSG101 siRNA transfection 293T Garrus et al. (2001)
Human CCR5 U6-shRNA on lentiviral vector transduction PBMC Li et al. (2003)
Human CCR5 U6-shRNA on lentiviral vector transduction Magi cells, PBL Qin et al. (2003)
Human CXCR4, CCR5 siRNA transfection U87, HeLa Martinez et al. (2002b)
W.-Y. Hu et al. / Virus Research 102 (2004) 59–64 61

readily transfected. RNAi can also be delivered via a DNA reduced (90%) 2 days after transfection as monitored by
vector encoding a short hairpin RNA expressed under the accumulation of the p19 matrix protein (Hu et al., 2002).
control of an RNA Pol III promoter (e.g. H1 and U6) or RNA Analysis of provirus formation in a spreading infection
Pol II promoter (e.g. CMV). Use of viral vectors has also also revealed that the two siGAG RNAs inhibited RCAS
been reported, including vectors based on adeno-associated replication. As controls, cultures were treated with siRNAs
virus, retroviruses, and lentiviruses. These three viruses have against irrelevant proteins or mock transfected, and these
the advantage of integrating the RNAi-initiating construct showed no significant inhibition. These data document that
in the host genome, thereby allowing inhibition to persist RNAi can efficiently inhibit RCAS replication.
during cell division.
3.3. Inhibition early or late?

3. RNA interference against avian retroviruses in cell There are two parts of the retroviral life cycle that could
culture potentially be targeted by RNAi. The incoming viral RNA
might be degraded by RNAi prior to completing reverse
3.1. ASLV history transcription, or the late viral mRNA and genomic RNA
transcribed from the integrated proviral DNA might also be
Ellerman and Bang (1908) demonstrated that a filterable cleaved. Several groups have investigated the viral RNAs
agent was responsible for causing chicken leukosis, a type attacked by RNAi, with somewhat different pictures emerg-
of leukemia/lymphoma. The cell-free transmission of solid ing.
tumors in chickens was reported by Rous (1911) at the Rock- We have analyzed this issue for both ASLV and HIV. To
efeller Institute in New York. It was subsequently observed model late expression from integrated proviruses, we trans-
that inoculation of RSV onto the chorioallantonic membrane fected DNA copies of the ASLV or HIV genomes along with
of the chick embryo resulted in the appearance of individual siRNAs against the retroviral genome or with control siR-
small tumors whose numbers could be correlated with vi- NAs. Inhibition was found to be efficient for both viruses.
ral concentrations (Keogh, 1938). Temin and Rubin (1958) Thus RNAi worked efficiently against the viral mRNAs and
demonstrated the oncogenic transformation of chick embryo genomic RNAs expressed from a DNA copy, modeling ac-
fibroblasts in culture by Rous sarcoma virus (RSV), and that tivity against the late viral RNAs produced by transcription.
single viral particles induced discrete foci of transformed What about RNAi against the viral RNA genome intro-
cells. Temin later used an avian retrovirus in his demon- duced into cells in the early steps of infection, i.e. initial fu-
stration that retroviral particles contain reverse transcriptase sion of the viral and cellular membranes? To study this step,
activity (Temin and Mizutani, 1970). Studies of the RSV cells were infected with ASLV or HIV and the production
also led to the identification of cellular homologs of the of early reverse transcription products measured. Because
oncogenes transduced by the acute transforming retroviruses the viral RNA genome serves as the template for DNA syn-
(Bishop, 1991). Today avian retroviruses are important mod- thesis, we reasoned that degradation of the incoming viral
els for retroviral replication and significant pathogens affect- RNA would result in reduced accumulation of viral DNA.
ing the poultry industry. However, no significant difference was seen in either ASLV
or HIV. Thus we concluded that the incoming genome was
3.2. RNAi against ASLV replication not a substrate for RNAi.
This conclusion has not been reached by all investiga-
We have used avian retroviruses as a model to study tors that examined this issue. Cullen and coworkers found
RNAi against retroviral replication. In these studies, siRNAs that the incoming genome could be a substrate for RNAi,
matching two sequences in the gag gene of Avian Sarcoma though not utilized as efficiently as the newly synthe-
Leucosis Virus (ASLV) were introduced into cells by trans- sized RNAs (Coburn and Cullen, 2002). Stevenson and
fection, one matching the p19 matrix (MA) coding region coworkers did not detect much difference in sensitivity to
and the other matching the p12 nucleocapsid (NC) coding RNAi in the early and late RNAs (Jacque et al., 2002).
region. As a model, cultured chicken DF-1 cells were stud- The reasons for these differences remain to be determined.
ied, and the RCAS version of ASLV was used. RCAS is Possibly different segments of the early RNAs are differ-
a replication-competent retroviral vector derived from RSV entially exposed in the viral core, so that the choice of
by removal of the src oncogene so as to allow introduc- RNA sequence to target determines whether or not they
tion of new genetic information in its place (Federspiel and are sensitive. Perhaps the detailed differences in exper-
Hughes, 1997; Hughes et al., 1987). imental protocols somehow dictate the outcome. It will
RCAS virus was introduced either by infecting DF-1 be interesting to work out these differences with further
cells or by transfecting them with RCAS proviral DNA. experimentation.
The siRNAs were introduced by cotransfection concur- Why would the incoming viral RNA genome be less sen-
rently. In the presence of specific siRNA, the production of sitive to RNAi? One possibility is that the incoming genomic
the RCAS virus in culture supernatants was dramatically RNA may be protected by viral proteins, thereby blocking
62 W.-Y. Hu et al. / Virus Research 102 (2004) 59–64

access of the RNAi machinery. Another possibility is that were analyzed at day 4 by sectioning and staining with an
incoming genomic RNA does not traffic through a cellular antibody against ASLV Gag protein p19.
compartment that harbors the RNAi machinery. For exam- Electroporation of RCAS DNA with no siRNA or control
ple, if the RNAi machinery resides at the ribosome or the siRNA resulted in virus spread from the electroporated half
nuclear pore, the incoming genomic RNA may not encounter of the spinal cord into the surrounding mesenchyme (due to
RNAi factors. In support of this, the RNAi machinery is re- secondary infection). At 2 days post-electroporation, ASLV
ported to be associated with the translational factor eIF2c, replication was inhibited efficiently (>90%) by either of the
suggesting ribosomal association (Doi et al., 2003; Sasaki siRNAs targeting gag: infection was evident in only a few
et al., 2003; Tabara et al., 1999). cells by anti-Gag staining. These results indicate that RNAi
can inhibit ASLV replication in chick embryos.

4. RNA interference against chicken retroviruses in an 4.3. Inhibiting pathogenesis by RSV


embryo electroporation model
We next asked whether RNAi could inhibit transforma-
4.1. RNAi in chicken embryos tion by RSV in the chick embryo model. RSV encodes the
oncogenic src gene, which disrupts signal transduction and
In ovo electroporation of chick embryos provides a sim- causes sarcomas in chickens. We electroporated embryos
ple and convenient system for studying gene activity during with DNA encoding RSV and different siRNAs. Three days
development (Nakamura and Funahashi, 2001). In ovo elec- after electroporation, none of chick embryos treated with
troporation involves the injection of nucleic acids into the RSV plus control siRNA survived. In contrast, 7 of 12 em-
central canal of the spinal cord, followed by electroporation bryos treated with the siGAG against p12 survived.
with five short (25-ms) square wave pulses at low voltages In order to examine the effect of RNAi on RSV patho-
(25 V). Nucleic acid enters the side of the embryo near the genesis, chick embryo sections were stained with different
positive electrode. The other half of the embryo is not elec- antibodies at 36 h post-electroporation. The mpm2 mono-
troporated and serves as a control. In previous studies, elec- clonal antibody detected M-phase phosphoproteins, thereby
troporation of dominant-negative gene constructs into chick identifying mitotic cells, and kip1 (cyclin-dependent ki-
embryos has allowed analysis of vertebrate gene function nase inhibitor p27) staining detected inhibition of cell
in the developing spinal cord (Thaler et al., 2002). We have cycle progression, showing cells arrested in G1. The Gag
found that RNA interference can be readily induced in the (p19) antibody was used to monitor the spread of RSV.
chick embryo spinal cord by in ovo electroporation, which In RSV-infected chick embryos, the neural tube was dis-
allowed the demonstration in vivo of inhibition of retroviral organized and normal cell cycle progression disrupted in
replication by RNAi. Pekarik et al. (2003) have also demon- the electroporated half. In contrast, normal development
strated the use of RNAi by in ovo electroporation. and properly controlled cell cycle progression were seen
Initially, to investigate whether RNAi took place in the when RSV replication was inhibited by RNAi, thereby
chick embryo, we used GFP as a target. A gfp encoding plas- documenting inhibition of pathogenesis.
mid was electroporated into the chicken neural tube together
with either specific (siGFP) or nonspecific (siLUC against
firefly luciferase) siRNAs. Two days after electroporation, 5. Applications of RNAi against animal retroviruses
in the absence of siRNA or in the presence of the control
siRNA bright green cells were detected in the half of the Today, avian retroviruses are a serious problem in the
spinal cord proximal to the positive electrode. In embryos poultry industry. Crowded conditions in poultry farms are
electroporated with the gfp plasmid and siGFP, a greater ideal for spread of epidemics of infectious disease, and retro-
than 90% reduction in GFP expression was observed. Elec- viruses remain a prominent threat. Vaccines are largely in-
troporation of siRNAs up to 3 ␮g/␮l showed no interruption effective, so new measures against disease caused by avian
of embryo development (Hu et al., 2002). These studies and retroviruses are needed.
those of Pekarik’s group establish that RNAi can operate in The three main disease-causing avian retrovirus of agri-
the developing chick spinal cord. cultural significance are the ASLV, reticuloendotheliosis
virus (REV), and lymphoproliferative disease virus (LPDV)
4.2. Inhibiting ALV infection by in ovo electroporation of turkeys (for review, see Payne, 1998). The ASLVs are
categorized into six subgroups based on their receptor
To investigate whether RNAi could inhibit replication of tropism, A–E and J. Of these, A, B, and J occur com-
RCAS in embryos, the two siRNAs against gag that had been monly. Although genetic resistance (due to recessive re-
shown above to be active in cell culture were used. siLUC sistance genes) to subgroups A–E have been observed in
was used as a nonspecific control. Two days after fertiliza- some chickens, all chicken strains are susceptible to sub-
tion, embryos were electroporated with a plasmid encod- group J (ALV-J). ALV-J induces myelocytic leukosis, a
ing the RCAS genome together with siRNAs. The embryos tumor-inducing infection of white blood cells from the bone
W.-Y. Hu et al. / Virus Research 102 (2004) 59–64 63

marrow. Control of ALV-J has become a major concern to Carmell, M.A., Xuan, Z., Zhang, M.Q., Hannon, G.J., 2002. The Arg-
the poultry industry as losses of breeder birds to ALV-J onaute family: tentacles that reach into RNAi, developmental control,
stem cell maintenance, and tumorigenesis. Genes Dev. 16, 2733–2742.
can be as high as 20–40% in severe outbreaks. The REV Caudy, A.A., Ketting, R.F., Hammond, S.M., Denli, A.M., Bathoorn,
family of retroviruses causes a variety of diseases including A.M., Tops, B.B.J., Silva, J.M., Myers, M.M., Hannon, G.J., Plasterk,
chronic lymphatomas in chickens, ducks, turkeys, geese, R.H.A., 2003. A micrococcal nuclease homologue in RNAi effector
pheasants, and quail. In turkeys, LPDV infection causes a complexes. Nature 425, 411–415.
lymphoproliferative disease. Coburn, G.A., Cullen, B.R., 2002. Potent and specific inhibition of human
immunodeficiency virus type 1 replication by RNA interference. J.
Furthermore, some poxviruses responsible for fowlpox Virol. 76, 9225–9231.
outbreaks in the United States have been found to harbor an Cogoni, C., Macino, G., 1999. Gene silencing in Neurospora crassa
intact REV provirus in their genomes (Kim and Tripathy, requires a protein homologous to RNA-dependent RNA polymerase.
2001; Singh et al., 2003). Evidently the REV virus used Nature 399, 166–169.
the poxvirus genome as an integration target rather than the Doench, J.G., Petersen, C.P., Sharp, P.A., 2003. siRNAs can function as
miRNAs. Genes Dev. 17, 438–442.
host cell chromosome, thereby incorporating the retroviral Doi, N., Zenno, S., Ueda, R., Ohki-Hamazaki, H., Ui-Tei, K., Saigo, K.,
genome in the poxvirus genome. The addition of REV to the 2003. Short-interfering-RNA-mediated gene silencing in mammalian
fowlpox genome has been implicated in increased poxvirus cells requires Dicer and eIF2C translation initiation factors. Curr. Biol.
replication in host chickens, and may facilitate infection of 13, 41–46.
previously vaccinated poultry. Dykxhoorn, D.M., Novina, C.D., Sharp, P.A., 2003. Killing the messenger:
short RNAs that silence gene expression. Nat. Rev. Mol. Cell. Biol.
An attractive potential technology to suppress retroviral 4, 457–467.
diseases of poultry would be germ line insertion of RNAi Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl,
constructs targeting either retroviral sequences or dispens- T., 2001a. Duplexes of 21-nucleotide RNAs mediate RNA interference
able host proteins involved in the infectious process. For in cultured mammalian cells. Nature 411, 494–498.
example, DNA hairpins initiating RNAi against the gag Elbashir, S.M., Lendeckel, W., Tuschl, T., 2001b. RNA interference is
mediated by 21 and 22-nucleotide RNAs. Genes Dev. 15, 188–200.
targets described above could be installed in the chicken
Ellerman, V., Bang, O., 1908. Experimentelle Leukamie bei Huhnern.
genome, blocking replication of pathogenic ASLVs. Such an Zentralbl. Bakteriol. Parasitenkd. Infectionskr. Hyg. Abt. Orig. 46,
approach, if successful, might be applied to inhibiting viral 595–609.
infection and pathogenesis in a wide range of domestic ani- Federspiel, M.J., Hughes, S.H., 1997. Retroviral gene delivery. Methods
mals. RNAi is also being investigated as a means of treating Cell. Biol. 52, 179–214.
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello,
HIV infection in humans, but development of RNAi as a
C.C., 1998. Potent and specific genetic interference by double-stranded
drug seems much more difficult—germ line modification of RNA in Caenorhabditis elegans. Nature 391, 806–811.
farm animals, in contrast, seems potentially feasible in the Garrus, J.E., von Schwedler, U.K., Pornillos, O.W., Morham, S.G., Zavitz,
near term. K.H., Wang, H.E., Wettstein, D.A., Stray, K.M., Cote, M., Rich, R.L.,
In summary, RNAi appears to be a promising new tech- Myszka, D.G., Sundquist, W.I., 2001. Tsg101 and the vacuolar protein
sorting pathway are essential for hiv-1 budding. Cell 107, 55–65.
nology for preventing and treating retroviral diseases, well
Hamilton, A.J., Baulcombe, D.C., 1999. A species of small antisense RNA
warranting a review that begins and ends with a bang! in posttranscriptional gene silencing in plants. Science 286, 950–952.
Hammond, S.M., Bernstein, E., Beach, D., Hannon, G.J., 2000. An
RNA-directed nuclease mediates post-transcriptional gene silencing in
Drosophila cells. Nature 404, 293–296.
References Hu, W.Y., Meyers, C.P., Kilzer, J.M., Pfaff, S.L., Bushman, F.D., 2002.
Inhibition of retroviral pathogenesis by RNA interference. Curr. Biol.
12, 1301–1311.
Al-Kaff, N.S., Covey, S.N., Kreike, M.M., Page, A.M., Pinder, R., Dale, Hughes, S.H., Greenhouse, J.J., Petropoulos, C.J., Sutrave, P., 1987.
P.J., 1998. Transcriptional and posttranscriptional plant gene silencing Adaptor plasmids simplify the insertion of foreign DNA into
in response to a pathogen. Science 279, 2113–2115. helper-independent retroviral vectors. J. Virol. 61, 3004–3012.
Banerjea, A., Li, M.J., Bauer, G., Remling, L., Lee, N.S., Rossi, J., Akkina, Jacque, J.M., Triques, K., Stevenson, M., 2002. Modulation of HIV-1
R., 2003. Inhibition of HIV-1 by lentiviral vector-transduced siRNAs in replication by RNA interference. Nature 418, 435–438.
T lymphocytes differentiated in SCID-hu mice and CD34+ progenitor Jorgensen, R., 1990. Altered gene expression in plants due to trans
cell-derived macrophages. Mol. Ther. 8, 62–71. interactions between homologous genes. Trends Biotechnol. 8, 340–
Bishop, J.M., 1991. Molecular themes in oncogenesis. Cell 64, 235–248. 344.
Bushman, F.D., 2003. RNA interference: applications in vertebrates. Mol. Keogh, E.V., 1938. Ectodermal lesions produced by the virus of Rous
Ther. 7, 9–10. sarcoma. Br. J. Exp. Pathol. 19, 1–9.
Caplen, N.J., Fleenor, J., Fire, A., Morgan, R.A., 2000. dsRNA-mediated Kim, T.J., Tripathy, D.N., 2001. Reticuloendotheliosis virus integration in
gene silencing in cultured Drosophila cells: a tissue culture model for the fowl poxvirus genome: not a recent event. Avian Dis. 45, 639–663.
the analysis of RNA interference. Gene 252, 95–105. Lee, N.S., Dohjima, T., Bauer, G., Li, H., Li, M.J., Ehsani, A., Salvaterra,
Caplen, N.J., Parrish, S., Imani, F., Fire, A., Morgan, R.A., 2001. Spe- P., Rossi, J., 2002. Expression of small interfering RNAs targeted
cific inhibition of gene expression by small double-stranded RNAs in against HIV-1 rev transcripts in human cells. Nat. Biotechnol. 20,
invertebrate and vertebrate systems. Proc. Natl. Acad. Sci. U.S.A. 98, 500–505.
9742–9747. Li, H., Li, W.X., Ding, S.W., 2002. Induction and suppression of RNA
Capodici, J., Kariko, K., Weissman, D., 2002. Inhibition of HIV-1 infection silencing by an animal virus. Science 296, 1319–1321.
by small interfering RNA-mediated RNA interference. J. Immunol. Li, M.J., Bauer, G., Michienzi, A., Yee, J.K., Lee, N.S., Kim, J., Li,
169, 5196–5201. S., Castanotto, D., Zaia, J., Rossi, J.J., 2003. Inhibition of HIV-1
64 W.-Y. Hu et al. / Virus Research 102 (2004) 59–64

infection by lentiviral vectors expressing Pol III-promoted anti-HIV Ratcliff, F.G., MacFarlane, S.A., Baulcombe, D.C., 1999. Gene silencing
RNAs. Mol. Ther. 8, 196–206. without DNA. RNA-mediated cross-protection between viruses. Plant
Lipardi, C., Wei, Q., Paterson, B.M., 2001. RNAi as random degradative Cell 11, 1207–1216.
PCR: siRNA primers convert mRNA into dsRNAs that are degraded Romano, N., Macino, G., 1992. Quelling: transient inactivation of gene
to generate new siRNAs. Cell 107, 297–307. expression in Neurospora crassa by transformation with homologous
Marathe, R., Anandalakshmi, R., Smith, T.H., Pruss, G.J., Vance, V.B., sequences. Mol. Microbiol. 6, 3343–3353.
2000. RNA viruses as inducers. Plant Mol. Biol. 43, 295–306. Rous, P., 1911. A sarcoma of the fowl transmissible by an agent separable
Martinez, J., Patkaniowska, A., Urlaub, H., Luhrmann, R., Tuschl, T., from the tumor cells. J. Exp. Med. 13, 397–411.
2002a. Single-Stranded Antisense siRNAs Guide Target RNA Cleavage Sasaki, T., Shiohama, A., Minoshima, S., Shimizu, N., 2003. Identification
in RNAi. Cell 110, 563–574. of eight members of the Argonaute family in the human genome small
Martinez, M.A., Gutierrez, A., Armand-Ugon, M., Blanco, J., Parera, M., star, filled. Genomics 82, 323–330.
Gomez, J., Clotet, B., Este, J.A., 2002b. Suppression of chemokine Schwarz, D.S., Hutvagner, G., Haley, B., Zamore, P.D., 2002. Evidence
receptor expression by RNA interference allows for inhibition of that siRNAs function as guides, not primers, in the Drosophila and
HIV-1 replication. AIDS 16, 2385–2390. human RNAi pathways. Mol. Cell. 10, 537–548.
Nakamura, H., Funahashi, J., 2001. Introduction of DNA into chick Sijen, T., Fleenor, J., Simmer, F., Thijssen, K.L., Parrish, S., Tim-
embryos by in ovo electroporation. Methods 24, 43–48. mons, L., Plasterk, R., Fire, A., 2001. On the role of RNA
Ngo, H., Tschudi, C., Gull, K., Ullu, E., 1998. Double-stranded RNA amplification in dsRNA-triggered gene silencing. Cell 107, 465–
induces mRNA degradation in Trypanosoma brucei. Proc. Natl. Acad. 476.
Sci. U.S.A. 95, 14687–14692. Singh, P., Schnitzlein, W.M., Tripathy, D.N., 2003. Reticuloendotheliosis
Novina, C.D., Murray, M.F., Dykxhoorn, D.M., Beresford, P.J., Riess, J., virus sequences within the genomes of field strains of fowlpox virus
Lee, S.K., Collman, R.G., Lieberman, J., Shankar, P., Sharp, P.A., display variability. J. Virol. 77, 5855–5862.
2002. siRNA-directed inhibition of HIV-1 infection. Nat. Med. 8, Surabhi, R.M., Gaynor, R.B., 2002. RNA interference directed against
681–686. viral and cellular targets inhibits human immunodeficiency virus type
Palatnik, J.F., Allen, E., Wu, X., Schommer, C., Schwab, R., Carrington, 1 replication. J. Virol. 76, 12963–12973.
J.C., Weigel, D., 2003. Control of leaf morphogenesis by microRNAs. Tabara, H., Sarkissian, M., Kelly, W.G., Fleenor, J., Grishok, A., Tim-
Nature 425, 257–263. mons, L., Fire, A., Mello, C.C., 1999. The rde-1 gene, RNA in-
Park, W.S., Miyano-Kurosaki, N., Hayafune, M., Nakajima, E., Matsuzaki, terference, and transposon silencing in C. elegans. Cell 99, 123–
T., Shimada, F., Takaku, H., 2002. Prevention of HIV-1 infection in 132.
human peripheral blood mononuclear cells by specific RNA interfer- Temin, H., Mizutani, S., 1970. RNA-dependent DNA polymerase in viri-
ence. Nucleic Acids Res. 30, 4830–4835. ons of Rous sarcoma virus. Nature 226, 1211–1213.
Payne, L.N., 1998. Retrovirus-induced disease in poultry. Poult. Sci. 77, Temin, H.M., Rubin, H., 1958. Characteristics of an assay for Rous
1204–1212. sarcoma virus and Rous sarcoma cells in tissue culture. Virology 6,
Pekarik, V., Bourikas, D., Miglino, N., Joset, P., Preiswerk, S., Stoeckli, 669–688.
E.T., 2003. Screening for gene function in chicken embryo using Thaler, J.P., Lee, S.K., Jurata, L.W., Gill, G.N., Pfaff, S.L., 2002. LIM
RNAi and electroporation. Nat. Biotechnol. 21, 93–96. factor Lhx3 contributes to the specification of motor neuron and in-
Pickford, A.S., Cogoni, C., 2003. RNA-mediated gene silencing. Cell terneuron identity through cell-type-specific protein-protein interac-
Mol. Life Sci. 60, 871–882. tions. Cell 110, 237–249.
Plasterk, R.H., 2002. RNA silencing: the genome’s immune system. Sci- Zeng, Y., Wagner, E.J., Cullen, B.R., 2000. Both natural and designed
ence 296, 1263–1265. micro RNAs can inhibit the expression of cognate mRNAs when
Qin, X.F., An, D.S., Chen, I.S., Baltimore, D., 2003. Inhibiting HIV-1 expressed in human cells. Mol. Cell. 9, 1327–1333.
infection in human T cells by lentiviral-mediated delivery of small Zeng, Y., Yi, R., Cullen, B.R., 2003. MicroRNAs and small interfering
interfering RNA against CCR5. Proc. Natl. Acad. Sci. U.S.A. 100, RNAs can inhibit mRNA expression by similar mechanisms. Proc.
183–188. Natl. Acad. Sci. U.S.A. 100, 9779–9784.
Virus Research 102 (2004) 65–74

RNA interference, arthropod-borne viruses, and mosquitoes


Irma Sanchez-Vargas, Emily A. Travanty, Kimberly M. Keene, Alexander W.E. Franz,
Barry J. Beaty, Carol D. Blair, Ken E. Olson∗
Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology,
Colorado State University, Fort Collins, CO 80523, USA

Abstract

RNA interference (RNAi) probably functions as an antiviral mechanism in most eukaryotic organisms. Variations in the activity of this
antiviral pathway in mosquitoes could explain, in part, why some mosquitoes are competent vectors of medically important, arthropod-borne
viruses (arboviruses) and others are not. There are three lines of evidence that show the RNAi pathway exists in Aedes species that transmit
arboviruses. The first is that recombinant Sindbis viruses expressing a RNA fragment from a genetically unrelated dengue-2 virus (DENV-2)
interfere with DENV-2 replication in Aedes aegypti mosquitoes by a mechanism similar to virus-induced gene silencing described in plants.
The second is that transfection of C6/36 (Aedes albopictus) cells with either double-stranded RNA or synthetic small interfering RNAs derived
from an arbovirus genome interferes with replication of the homologous virus. The third is that a hairpin DENV-2-specific RNA transcribed
from a plasmid can generate virus-resistant C6/36 cells. We hypothesize that genetically modified mosquitoes can be generated that transcribe
a flavivirus-specific dsRNA, triggering the RNAi response soon after ingestion of a blood meal. This could induce the RNAi pathway in the
midgut prior to establishment of virus infection and profoundly change vector competence. Towards this goal, we are developing transgenic
A. aegypti lines that are refractory to DENV by exploiting the RNAi pathway.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Mosquitoes; RNAi; dsRNA; siRNA; Dengue viruses; Arthropod-borne viruses

1. Introduction competent mosquito populations that support virus infec-


tion. Traditional mosquito control to lower the population
Mosquitoes transmit a number of medically important density of vectors and prevent arboviral disease has been
arthropod-borne viral pathogens to humans and other an- unsuccessful due to cost, expanding urbanization, increases
imals. Dengue viruses (DENV: family Flaviviridae, genus in human populations, pesticide resistance, and spreading
Flavivirus, species Dengue virus) cause an estimated of vector species due to globalization of trade and air travel.
50–100 million human cases of dengue fever (DF) every New strategies are needed to control these diseases.
year (Monath, 1994). The rapid spread of West Nile virus Dengue viruses arguably cause the most important
(family Flaviviridae, genus Flavivirus, species West Nile arthropod-borne viral infections. DENV serotypes 1–4 are
virus) in North America and outbreaks of Venezuelan equine transmitted to humans by Aedes (A.) aegypti and A. albopic-
encephalitis virus (family Togaviridae, genus Alphavirus, tus. Severe forms of DENV disease, DEN hemorrhagic fever
species Venezuelan equine encephalitis virus) in South (DHF) and DEN shock syndrome (DSS) are probably caused
America and Rift Valley fever virus (RVFV, family Bun- by the global trafficking of virulent genotypes of DENV
yaviridae, genus Phlebovirus, species Rift Valley fever virus) (Gubler, 1998) and sequential infection of individuals with
in the Middle East during the last decade further demon- heterotypic virus serotypes leading to antibody-mediated
strate the enormous public health problems associated with disease enhancement (Halstead, 1989). DHF/DSS are in-
arbovirus infections. The maintenance and transmission of creasing at an alarming rate due to the hyperendemnicity
arboviruses in nature is, in part, dependent on genetically of DENV in many parts of the world and the trafficking of
DENV serotypes to new geographic regions (Gubler, 1998).
∗ Corresponding author. Tel.: +1-970-491-4383; Dengue viruses, like other flaviviruses, are small, enve-
fax: +1-970-491-8323. loped particles (50 nm in diameter) containing a single-
E-mail address: kolson@lamar.colostate.edu (K.E. Olson). stranded, positive-sense RNA of approximately 11 Kb. The

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.017
66 I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74

genomic RNA is capped at the 5 end but not polyadeny- more complete understanding of arbovirus-mosquito vec-
lated at the 3 end. The virion RNA contains a single long tor interactions. Identifying molecules that modulate ar-
open reading frame encoding three structural proteins, cap- bovirus replication in mosquitoes and the characterization
sid (C), premembrane (prM), and envelope (E), and seven of mosquito genes that encode these molecules may lead
non-structural proteins, NS1, NS2A, NS2B, NS3, NS4A, to powerful new approaches to assess risk and attempt
NS4B, and NS5. The genomic RNA is translated into a new strategies to control arbovirus transmission (Beerntsen
polyprotein precursor that is co- and post-translationally et al., 2000; Blair et al., 2000).
processed by cellular and viral proteases into individual
proteins (Chambers et al., 1990).
Dengue viruses enter the mosquito when the adult fe- 2. Arboviruses, mosquitoes and RNA interference
male takes an infectious blood meal. In competent mosquito
vectors, viral amplification begins with the initial, unidirec- What could be the nature of arbovirus modulation in
tional, productive infection of the midgut epithelium cells. mosquito cells? A clue may lie in the replication strat-
Following replication of virus in the epithelial cells, virus egy used by positive-sense RNA viruses like DENV. Fla-
escapes the midgut and disseminates to secondary target or- vivirus RNA replication occurs in the cytoplasm of infected
gans such as the salivary glands. Further replication occurs cells and is asymmetric and semi-conservative (Chu and
in the salivary glands, and virus is eventually shed into the Westaway, 1985). RNA replicative intermediates (RI) are
salivary gland ducts. Transmission occurs by way of infected partially double-stranded RNAs (dsRNA) generated within
saliva in a subsequent bite. The time between initial infection infected cells (Westaway et al., 1997). Recently, Uchil
of the midgut and transmission of virus in saliva is termed and Satchidanandam (2003) provided biochemical evi-
the extrinsic incubation period (EIP). The EIP for DENV in dence that flaviviruses rearrange the architecture of cell
competent A. aegypti is usually 7–14 days (Chandler et al., membranes associated with virus replication to sequester
1998; Scott and Burrage, 1984). the dsRNAs in a double membrane barrier enclosing the
Transmission of virus to the vertebrate host is prevented viral replication complex (RC). The intricate mechanism
if one or more barriers to infection and dissemination are adopted by flaviviruses to hide the RI suggests that this
present in the mosquito (Hardy and Strauss, 1988; Woodring adaptation may have evolved to limit exposure of the RC
et al., 1996). Little is known about the molecular basis of to dsRNA-mediated host defenses such as protein kinase
these barriers. However, it is possible to speculate that a R (PKR), RNase L, ␣/␤ interferon, and RNA interference
midgut infection barrier (MIB) could occur if the arbovirus (RNAi). Simply put, the host cell and flaviviruses appear
fails to recognize critical receptors for entry into the midgut to have co-evolved defense-counter defense strategies.
epithelia, the virus is not processed correctly by proteolytic Mosquito cells do not appear to have PKR, RNase L, or ␣/␤
enzymes in the midgut lumen, or the virus fails to uncoat interferon pathways comparable to those found in mam-
during initial steps of infection. MIBs have been observed malian cells, but mosquito cells most likely have an RNAi
in certain A. aegypti populations after oral infection with a pathway. Therefore arbovirus interactions with mosquito
strain of DENV-2 that is infectious for other populations cells should be an excellent model for understanding how
(Bennett et al., 2002; Black et al., 2002). A. aegypti can animal viruses interact with RNAi pathway. Can RNAi act
also exhibit a midgut escape barrier (MEB). Here, virus in- as an antiviral pathway in mosquitoes, and if so, how do
fects the midgut epithelium but is unable to disseminate arboviruses overcome this pathway in competent mosquito
to other organs and remains restricted and sequestered in vectors?
the midgut cells of some populations of mosquitoes (Black RNAi is an evolutionarily conserved process through
et al., 2002). Although the molecular basis for MEBs is cur- which dsRNA induces the silencing of cognate gene expres-
rently unknown, one can hypothesize that virus replication sion (Bernstein et al., 2001; Carthew, 2001). RNAi results
in the midgut or secondary tissues is somehow modulated. in the degradation of dsRNA and of any mRNA present
A. aegypti with MEBs to DENV infections have also been in the organism with high sequence homology with the
described (Bennett et al., 2002; Black et al., 2002). dsRNA trigger (Bernstein et al., 2001). Synthetic dsRNA
The fields of molecular biology, vector biology, and can be introduced by transfection or can be produced intra-
bioinformatics are now converging to define the nature of cellularly by the introduction of viruses or transposons that
these barriers to infection. For example, the genomes of generate dsRNA during their replication cycle (Hammond
two insects in the order Diptera, the fruit fly Drosophila et al., 2001b). In addition, transcripts from short hairpin
(D.) melanogaster and the medically important mosquito sequences encoded within the genome of many organisms
vector of malaria, Anopheles (A.) gambiae, have now been appear to enter the RNAi pathway and function to reg-
sequenced and annotated (Adams et al., 2000; Holt et al., ulate the expression of endogenous protein-coding genes
2002). The genome sequence determination of a third (Grishok et al., 2001; Hutvagner et al., 2001; Hutvagner
dipteran, A. aegypti, is currently underway and should be and Zamore, 2002; Ketting et al., 2001; Knight and Bass,
available in the next several years. We can now use bioin- 2001). DsRNA is initially cleaved into small interfering
formatic and functional genomic analyses to develop a RNA (siRNA), 21–25 nucleotides in length, by a Dicer-1
I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74 67

Fig. 1. Proposed model for RNA-silencing pathway in insects (Drosophila). dsRNA is recognized by Dicer (Step 1) and cleaved into 21–25 nt dsRNA
fragments (siRNAs) forming siRNA-protein complexes (Step 2). Double-stranded siRNA molecules are unwound by the Dicer-associated helicase and
incorporated into RISC (Step 3). RISC is guided by the incorporated siRNAs to target RNA molecules of complementary sequence (Step 4). RISC then
cleaves target RNA (Step 5). Dicer: RNase III-like nuclease, helicase activity, ATP dependent. RISC: RNA-induced silencing complex (endonuclease).

protein (Fig. 1). The siRNAs are then incorporated into example, the tomato bushy stunt virus (family Tombusviri-
the RNA-induced silencing complex (RISC), which con- dae, genus Tombusvirus, species Tomato bushy stunt virus)
tains proteins of the Argonaute family (rde-4, rde-1 and p19 protein suppresses post-transcriptional gene silenc-
drh-1/2 in Caenorhabditis (C.) elegans or Argonaute 2 ing (PTGS) in plants by binding siRNAs generated after
in D. melanogaster) (Tabara et al., 2002; Williams and virus infection (Silhavy et al., 2002). Furthermore, the in-
Rubin, 2002; Zamore et al., 2000). siRNAs are unwound sect virus, Flock house virus (family Nodaviridae, genus
by an RNA helicase to act as guide sequences that direct Alphanodavirus, species Flock house virus) B2 protein
the RNA degradation machinery to the target mRNAs. The suppresses RNAi activity in both plants and Drosophila S2
RISC monitors the sequences of cytoplasmic RNA and is cells, emphasizing that an evolutionarily conserved RNAi
able to cleave the cognate target RNA in a sequence spe- pathway has a natural antiviral role (Li et al., 2002b).
cific, siRNA-dependent manner (Martinez et al., 2002). It The sequencing of the A. gambiae genome now makes
has been proposed for plants and lower eukaryotes such as it possible to identify genes that may be involved in the
C. elegans, that siRNAs also function as primers that are RNAi pathway and what role, if any, these genes have in ar-
extended on the targeted RNA by an RNA-dependent RNA bovirus infection. Recent studies have demonstrated that sev-
polymerase (RdRp) to amplify the dsRNA trigger (Lipardi eral members of the Dicer and Argonaute protein families,
et al., 2001; Sijen et al., 2001). Although RdRp-like activ- homologous to D. melanogaster proteins, are encoded in the
ity has been detected in D. melanogaster embryos, it does A. gambiae genome (Hoa et al., 2003). To identify the role of
not appear to play a significant role in RNAi within D. these proteins in RNAi, Hoa and colleagues initially showed
melanogaster and A. gambiae (Hoa et al., 2003; Roignant that transfection of A. gambiae cells with dsRNA derived
et al., 2003). from a 500 bp region of the firefly luciferase gene could effi-
What is the evidence that RNAi is an important antiviral ciently silence transient expression of a luciferase transgene.
defense strategy used by the host to modulate virus infec- A. gambiae Sua1B cells were pretreated with dsRNA derived
tions? First, there is a wealth of evidence that plant RNA from A. gambiae dcr1, dcr-2, or ago1-5 prior to silencing lu-
viruses act as both inducers and targets of RNAi (Ruiz ciferase expression. Pretreatment with dsRNA derived from
et al., 1998; Vance and Vaucheret, 2001). In addition, many A. gambiae dcr2, ago2, or ago3 consistently led to recovery
plant RNA viruses encode suppressors of RNAi, supporting of luciferase activity, by knocking down expression of com-
the hypothesis that RNAi is a viral defense system. For ponents of the RNAi pathway. That ago2 gene expression is
68 I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74

important in RNAi is not surprising since it is thought to be This is reminiscent of the use of RNA virus expression vec-
an important component of RISC (Hammond et al., 2001a). tors to silence host genes in plants. Nicotiana benthamiana
However, significant recovery of luciferase activity was not inoculated with modified tobacco mosaic virus (family To-
found in silenced Sua1B cells following pretreatment of cells bamoviridae, genus Tobamovirus, species tobacco mosaic
with dsRNA from A. gambiae dcr1, ago1, ago4, ago5, or virus) or potato virus X (genus Potexvirus, species potato
β-galactosidase genes. The gene dcr1 encodes the Dicer-1 virus X) carrying host-related inserts suppressed genes ho-
protein that is essential for cleaving dsRNAs into siRNAs mologous to the inserts (Kumagai et al., 1995; Ruiz et al.,
in a number of organisms. Why down-regulation of dcr1 1998). Some plant viruses also were able to induce silenc-
gene expression did not affect RNAi in A. gambiae cells ing of either transgenes or genes carried by genetically
is not easily explained, but may be due to the cell culture unrelated viruses having significant sequence identity with
system used in these experiments. We have recently used a the inoculated virus (Lindbo et al., 1993; Ratcliff et al.,
different strategy in A. gambiae mosquitoes to assign func- 1999). This phenomenon was termed virus-induced gene
tion to these genes by co-injecting 103 plaque forming units silencing (Angell and Baulcombe, 1997; Ruiz et al., 1998),
of O’nyong-nyong virus (ONNV; family Togaviridae, genus and the mechanism of action for VIGS was similar to
Alphavirus, SFV complex) with ∼200 ng of dsRNA de- post-transcriptional gene silencing or RNAi.
rived from dcr1, dcr2, ago2 or ago3 genes. ONNV is one We then conducted experiments to observe whether the
of the few arboviruses naturally transmitted by A. gambiae. mechanism by which recombinant Sindbis viruses induced
We have observed that virus replication is significantly in- resistance in mosquitoes to DENV-2 was similar to RNAi.
creased in mosquitoes co-injected with ONNV and dsRNA We engineered a Sindbis virus that contained a 290 base
from dcr1, dcr2, ago2, or ago3 and not with ␤-gal dsRNA, region of DENV-2 prM in either sense or antisense ori-
suggesting that these genes play an important role in RNAi entation and infected C6/36 cells with the recombinant
in the mosquito and that RNAi indeed functions as an antivi- virus. Infection with Sindbis virus with both orientations
ral pathway in A. gambiae (K.M. Keene, unpublished data). of DENV-2 prM RNA in the genome ablated the ability of
Also, we have shown that co-injection of dsRNA from the DENV-2 to infect cells, but not other serotypes of DENV.
nsp1 and nsp3 genome regions from ONNV significantly We observed by Northern blot analysis that intracellular
decreases the amount of virus found in the mosquito (K.M. concentrations of Sindbis genomic and subgenomic RNAs
Keene and K.E. Olson, unpublished data). carrying the DENV-2 prM RNA significantly declined by
It is very likely that an RNA silencing mechanism also 96 h post-infection, but cells retained profound resistance
exists in A. aegypti and A. albopictus mosquitoes. Aedes to DENV-2 challenge (Adelman et al., 2001). We reasoned
species of mosquitoes transmit a number of medically impor- that if an RNAi phenomenon were responsible for the ob-
tant arboviruses, such as DENV, yellow fever virus (family served resistance in C6/36 cells, then DENV-2 should be
Flaviviridae, genus Flavivirus, species yellow fever virus), unable to replicate in the cells even though the effector prM
Chikungunya virus (family Togaviridae, genus Alphavirus, sequences were undetectable. Three important features of
SFV complex), and RVFV. If RNAi is present, it might be a the resistance phenomenon suggested to us that recombi-
likely mechanism for modulating arbovirus replication in the nant Sindbis viruses were triggering an RNAi-like response
natural host and may explain, in part, why some mosquitoes in mosquito cells. (1) The interference observed was not re-
are refractory for transmission of arboviruses such as DENV. lated to the polarity of the inserted DENV sequence, because
Sindbis viruses with either sense or antisense DENV RNA
inserts conferred similar levels of resistance in mosquitoes
3. Evidence for RNA-mediated silencing of DENV when challenged with the homologous DENV serotype. (2)
replication in mosquito cells DENV-2 RNA failed to accumulate after challenge only
in cells infected with the recombinant Sindbis virus carry-
The first evidence that a functional RNAi pathway ex- ing the DENV-2 sequence. (3) The resistance phenotype
ists in Aedes mosquitoes came from studies to generate A. was not dependent on high concentrations of effector RNA
albopictus C6/36 cells that were resistant to DENV-2 infec- transcribed by Sindbis virus as one might expect with an
tion. We observed that transduction by recombinant Sindbis antisense RNA mechanism. Finally, we have now shown
viruses (family Togaviridae, genus Alphavirus, WEEV com- that the Sindbis virus used to transcribe the DENV-2 prM
plex) carrying the prM coding region of DENV-2 in their effector RNA triggers formation of RNA species that are
genome in either sense or antisense orientation made the 21–23 nucleotides in size and consistent with siRNAs, the
cells resistant to DENV-2, but not DENV-3 (Gaines et al., hallmark of RNAi (Olson et al., 2002). These small RNAs
1996; Olson et al., 2002). We then showed that this resis- hybridized with sense and antisense RNA probes derived
tance phenomenon could be reproduced in adult, female A. from either Sindbis or DENV insert RNA sequences.
aegypti by co-injecting 103 pfu/ml of recombinant Sindbis To test whether Sindbis viruses can silence endogenous
virus and DENV-2 (Olson et al., 1996). Eleven days after genes in A. aegypti, we engineered the Sindbis virus genome
injection, DENV-2 was absent only in mosquitoes that were to contain the coding region for early trypsin (TrypEarl)
co-infected with Sindbis viruses carrying the prM fragment. protein, an important regulator of the proteolytic cascade
I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74 69

demonstrated silencing of DENV-1 replication after trans-


fection of C6/36 cells with DENV-1 specific dsRNA. They
observed that different dsRNAs targeted to DENV-1 RNA
inhibited replication at similar levels, suggesting that degra-
dation of the whole genome RNA can be induced by cleav-
age within a region that corresponds to less that 1% of the
genome. However, the degree of inhibition reported using
Sindbis-induced silencing of DENV replication was greater
than reported for transfected dsRNA (Caplen et al., 2002).
Adelman et al. (2002) have shown that virus resistance
also can be generated in cultured C6/36 cells by transcription
of DENV-specific RNA that was designed to form a hair-
pin structure from a plasmid. The hairpin was transcribed
from the constitutive IE-1 promoter of Autographa califor-
nica baculovirus and consisted of the 567 base positive-sense
DENV-2 sequences encoding the prM protein followed by
a 290 base antisense RNA complementary to the first 290
bases of the prM RNA. After transformation with the plas-
mid, clonal cell lines were selected using an antibiotic resis-
Fig. 2. Detection of siRNA in midgut of mosquitoes infected with re- tance marker and challenged with DENV-2. The cell lines
combinant dsSIN viruses. Fourteen days after infection the midguts of 70 were classified as resistant to virus replication if fewer than
mosquitoes were obtained and small RNAs were purified following the 5% of cells in a culture expressed DENV-2 envelope (E)
protocol described by Hamilton and Baulcombe (1999) and hybridized
antigen 7 days after challenge. Cells in the line designated
with 32 P-labeled probe complementary to TrypEarl gene. Lane 1, 22 nt
RNA marker; lane 2, uninfected mosquitoes; lane 3, mosquitoes infected FB9.1 that was highly resistant to DENV-2 failed to accumu-
with dsSIN/GFP (MRE3 2J-GFP, negative control); lane 4, mosquitoes late DENV-2 genome RNA after challenge, indicating that
infected with dsSIN/ETas (MRE3 2J-ETas-500 bp); lane 5, mosquitoes in- viral RNA can be targeted by expression of a dsRNA-trigger
fected with dsSIN/FETas (MRE3 2J-FETas-800 bp); lane 6, mosquitoes (Fig. 3B). Most importantly, DENV-2 prM-specific, small
infected with dsSIN/FETs (MRE3 2J-FETs-800 bp). FETas: full length
RNAs 21–25-nt in length were detected in resistant FB9.1
TrypEarl gene antisense; FETs: full length TrypEarl gene sense; ETas:
fragment of TrypEarl gene antisense derived from the 3 500 bp of the cells using protocols described by Hamilton and Baulcombe
gene. (1999), consistent with RNA silencing as the mechanism
of resistance (Fig. 3A). These small RNA species were not
found in control cells. Surprisingly, in the FB9.1 cell line,
in the midguts of blood fed mosquitoes. A recombinant the rate of DENV-3 RNA and envelope protein accumulation
Sindbis virus was made that transcribed a 500 bp region also was greatly reduced when compared to infection of con-
of TrypEarl gene. Expression of TrypEarl RNA sequence trol cells (Fig. 3B). The viral interference appeared to be se-
from recombinant Sindbis viruses led to silencing of early quence specific because the replication of the more distantly
trypsin expression and the degradation of TrypEarl mRNA related flavivirus, West Nile virus, was not affected (unpub-
in mosquito midguts. The phenomenon appeared to be lished data). The cross-resistance to DENV-3 seen in the
post-transcriptional, because we were able to detect siRNAs FB9.1 cells is consistent with RNAi. When the prM coding
(21–25 nt) using a TrypEarl RNA probe in the midguts of regions from DENV-2 and DENV-3 are aligned, a number
these mosquitoes (Fig. 2). Inhibition of the expression of of short homologous regions are found; one of these regions
TrypEarl also affected dissemination of DENV-2 to head is 23 nt in length with 2 mismatches and if it is produced as
tissue. The experiments described above suggested that siRNA, it might direct the silencing of both DENV-2 and
virus-induced gene silencing (VIGS) occurs in mosquitoes DENV-3. According to Elbashir et al. (2001) mismatches
and that Sindbis virus expression vectors will be important are not tolerated at the middle of the siRNA as this is
tools for understanding the role of mosquito genes in de- the region that determines the cleavage site in the target
termining vector competence and virus transmission. The mRNA. The proposed DENV-2/DENV-3 siRNA does not
Sindbis expression system has already been used to knock- contain any mismatches in the vital cleavage area. Sequence
down expression of reporter genes in transgenic A. aegypti comparisons between DENV-2 and DENV-1, and DENV-2
mosquitoes and determine gene function of phenoloxidase and DENV-4 genomes reveal similar potential siRNA re-
in Armigeres subalbatus mosquitoes (Johnson et al., 1999; gions. Experiments are currently underway to determine if
Shiao et al., 2001). the cross-protection phenomenon extends to all 4 DENV
Others have presented evidence that a RNAi-like defense serotypes and if the cross-protection is indeed due to siR-
against DENV can be efficiently triggered in C6/36 mosquito NAs resulting from the near-homologous regions.
cells by dsRNA corresponding to a portion of the DENV-1 Recently, we have directly demonstrated the role of
or DENV-2 genome. The study by Caplen et al. (2002) DENV-2 specific siRNAs in inducing RNAi to DENV in
70 I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74

Fig. 3. Detection of siRNA in FB9.1 cells and DENV-2 and DENV-3 RNA accumulation in FB9.1 and H9.1 cells after DENV challenge. (A) Northern
blot hybridization to detect siRNA in FB9.1 cells. Small RNAs were obtained following the protocol described by Hamilton and Baulcombe (1999). Low
molecular weight RNAs were fractionated by 12% urea-PAGE, blotted and hybridized with 32 P-labeled sense and antisense RNA probes complementary
to prM gene region of DENV-2 RNA. (a) Untransformed C6/36 cells; (b and d) H9.1 control cells; (c and e). FB9.1 cells. The arrow shows DENV-2
prM-specific, small RNAs 21–25-nt in length. (B) Slot blot analysis comparing DENV-2 and DENV-3 RNA accumulation in FB9.1 and H9.1 cells after
DENV challenge. Total cellular RNA from each cell line on days 1–14 following DENV challenge was blotted and probed for DENV RNA. 32 P-labeled
probe complementary to the NS2b-NS3 gene region of DENV-2 RNA was used for detecting DENV-2 and a 32 P-labeled probe complementary to the C
gene region was used for detecting DENV-3. FB9.1: C6/36 cell line transformed with DENV-2 prM hairpin-expression plasmid. H9.1: C6/36 cell line
transformed with vector plasmid (control).

mosquito cells. We observed that transfection of C6/36 this interference will extend to DENV-3. We also gener-
cells with synthetic 21 bp siRNA designed from a region of ated siRNAs in vitro by digesting a dsRNA derived from a
DENV-2/DENV-3 prM homology (Dharmacon Research DENV-2 prM sequence with recombinant human Dicer-1
Inc., Lafayette, CO) resulted in interference with DENV-2 protein (Gene Therapy Systems). After transfection of the
replication (Fig. 4A). Experiments are underway to see if siRNAs into C6/36 cells, the cells were challenged with

Fig. 4. Effect of siRNA or d-siRNA on DENV-2 mRNA stability. (A) Effect of siRNA on DENV-2 mRNA stability. C6/36 cells were transfected with
TransIT-TKO (Mirus, Corp. Madison, WI) as a negative control or in conjunction with either luciferase GL2 siRNA duplex or DENV siRNA duplex
(12 mM). After 48 h post-transfection, cells were infected with DENV-2. Total cellular RNA was isolated from transfected cells on day 6 following DENV
challenge, and was then blotted and probed for DENV RNA using a 32 P-labeled probe complementary to the NS2b-NS3 gene region of DENV-2. Lane
1, DENV-2 alone, 6 dpi; lane 2, control luciferase GL2 siRNA and DENV-2, 6 dpi; lane 3, DENV siRNA and DENV-2, 6 dpi. (B). Effect of a pool of
22 bp siRNA (d-siRNA) generated by cleavage of in vitro transcribed dsRNA corresponding to DENV-2 C-prM-M (485bp) using recombinant human
Dicer enzyme (Gene Therapy Systems San Diego, CA). C6/36 cells were transfected with Gene Silencer (Gene Therapy Systems) as a negative control
either in conjunction with d-siRNA/GFP or d-siRNA/DENV-2-M. After 48 h post-transfection, cells were infected with DENV-2. Total cellular RNA
was isolated from transfected cells on day 6 following DENV challenge, and was then blotted and probed for DENV RNA using a 32 P-labeled probe
complementary to the NS2b-NS3 gene region of DENV-2. Lane 1, DENV-2 alone; lane 2, d-siRNA/GFP and DEN2 6 dpi; lane 3, d-siRNA/ DENV-2-M
and DEN-2 6 dpi. The bottom panel shows the ethidium bromide stained rRNA.
I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74 71

DENV-2 and failed to accumulate viral RNA. Cells trans- as a disease control method, but as we learn more about
fected with siRNAs derived from GFP mRNA accumulated arbovirus-vector interactions we will learn more about the
normal concentrations of DENV-2 RNA after challenge feasibility of this type of approach.
(Fig. 4B). These experiments demonstrate that direct ap-
plication of siRNAs targeted to DENV RNA can cause
degradation of DENV genomic RNA in mosquito cells and 5. Conclusions
suggest that siRNAs may be an important tool for antiviral
therapy of patients infected with flaviviruses (Song et al., An important opportunity for investigation is to determine
2003). how arboviruses overcome the RNAi pathway. Arboviruses
could replicate faster than the ability of RNAi to overcome
the virus. Alphaviruses such as Sindbis may use this strat-
4. RNAi and new strategies for arbovirus disease control egy. We know that Sindbis viruses replicate and generate
virus in C6/36 cells within 12 h post infection. We also know
How can the information presented so far be used to de- that Sindbis viruses can induce an RNAi-like response in
velop new tools for predicting DENV disease outbreaks or mosquito cells at about 48 h post infection, at which time
controlling the spread of DENV disease in the future? Once we first detect the appearance of siRNAs. The appearance
we identify genes in the RNAi pathway that are involved in of siRNAs roughly correlates with decline in virus mRNAs,
the antiviral response we can, in theory, look for variations and detection of maximum concentration of siRNA occurs
in these genes in populations of vectors. If, as an example, at 120 h post-infection, at which time little viral RNA is de-
we can associate genetic variation in ago2 genes with vec- tected by Northern blot analysis (Fig. 5). In natural mosquito
tor competence for DENV-2 and incidence of transmission, hosts, the EIP for some alphaviruses is as short as 4 days
it may be possible to rapidly screen for and identify vector
populations with a high risk for transmitting the arbovirus
to humans. These vector populations can be targeted for
control or elimination.
We can also think about new control strategies that in-
duce a more robust antiviral RNAi pathway in mosquitoes.
We have hypothesized that genetically modified mosquitoes
could be generated transcribing a flavivirus-specific dsRNA
that triggers the RNAi response soon after ingestion of
a blood meal. This could induce the RNAi pathway in
midguts prior to (or concurrent with) the establishment of
virus infection and change a highly competent mosquito
vector for a flavivirus to a poorly competent vector or a
vector that completely eliminates the virus infection. Re-
ducing the number of infected vectors will in turn reduce
the incidence of human disease. Couple this strategy with a
genetic drive mechanism such as transposable elements, and
it may be possible to drive the resistance gene into vector
populations (Beerntsen et al., 2000; Ribeiro and Kidwell,
1994). Towards this goal, we are developing transgenic A.
aegypti lines that are refractory to DENV by exploiting the
RNAi pathway of mosquitoes (Adelman et al., 2002; Olson
et al., 2002). We are currently generating genetically mod-
ified A. aegypti that transcribe a dsRNA derived from the
DENV-2 prM sequence. The dsRNA is transcribed from a Fig. 5. Detection of Sindbis virus-specific siRNA in C6/36 cells. (A)
midgut-specific (e.g. carboxypeptidase) promoter to initiate Northern blot analysis of dsSIN virus transcripts. Total RNA from C6/36
resistance in the organ system first encountering virus fol- cells infected with TE3 2J virus were isolated at 1, 2, 3, 4, 5, 6 or 7
days post-infection, 32 P-labeled sense or antisense probes to E1 gene re-
lowing ingestion of an infected blood meal (Moreira et al., gion of Sindbis RNA were used for detection of Sindbis mRNA. The
2000). The DENV-2-derived prM cDNA fragments are top panel shows ethidium bromide stained rRNA. (B) RNA blot analysis
inserted as inverted-repeat sequences into a Mariner MosI of Sindbis-specific siRNA in uninfected C6/36 cells (mock) or in C6/36
transposable element used for germ line transformation. cells infected with TE3 2J virus. Small RNAs were obtained following
The inverted repeat sequences contain a 291 nt fragment of the protocol described by Hamilton and Baulcombe (1999). Low molec-
ular weight RNAs were fractionated by 12% urea-PAGE, blotted and hy-
prM region in sense and antisense orientations separated by bridized with 32 P-labeled sense or antisense RNA probe complementary
a major intron of the A. aegypti Sialokinin I gene. Obvi- to E1 gene region of Sindbis RNA. The positions of the 10, 20 and 30
ously, much more work needs to be done to use this strategy nucleotide markers are shown at the left.
72 I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74

(Myles et al., 2003; Scott et al., 1984), suggesting that the D., Bolshakov, S., Borkova, D., Botchan, M.R., Bouck, J., Brokstein,
virus may replicate in and escape the midgut before the P., Brottier, P., Burtis, K.C., Busam, D.A., Butler, H., Cadieu, E.,
Center, A., Chandra, I., Cherry, J.M., Cawley, S., Dahlke, C., Dav-
RNAi response is underway. Arboviruses also could limit the enport, L.B., Davies, P., de Pablos, B., Delcher, A., Deng, Z., Mays,
number of dsRNA replication forms in an infected mosquito A.D., Dew, I., Dietz, S.M., Dodson, K., Doup, L.E., Downes, M.,
cell. Perhaps a threshold level of dsRNA is needed to trigger Dugan-Rocha, S., Dunkov, B.C., Dunn, P., Durbin, K.J., Evangelista,
RNAi. A corollary to this is that some arboviruses appear C.C., Ferraz, C., Ferriera, S., Fleischmann, W., Fosler, C., Gabrielian,
to sequester their dsRNA replicative forms from the cellular A.E., Garg, N.S., Gelbart, W.M., Glasser, K., Glodek, A., Gong, F.,
Gorrell, J.H., Gu, Z., Guan, P., Harris, M., Harris, N.L., Harvey, D.,
RNAi machinery (Uchil and Satchidanandam, 2003) sug- Heiman, T.J., Hernandez, J.R., Houck, J., Hostin, D., Houston, K.A.,
gesting a long evolutionary history of interaction between Howland, T.J., Wei, M.H., Ibegwam, C., et al., 2000. The genome
arboviruses and host dsRNA-mediated defense pathways. sequence of Drosophila melanogaster. Science 287, 2185–2195.
Viruses also may encode RNAi suppressors to over- Adelman, Z.N., Blair, C.D., Carlson, J.O., Beaty, B.J., Olson, K.E., 2001.
come the host defense. Examples of plant RNA and DNA Sindbis virus-induced silencing of dengue viruses in mosquitoes. In-
sect. Mol. Biol. 10, 265–273.
viruses encoding suppressors to down-regulate RNAi have
Adelman, Z.N., Sanchez-Vargas, I., Travanty, E.A., Carlson, J.O., Beaty,
now been described that presumably give the viruses an B.J., Blair, C.D., Olson, K.E., 2002. RNA silencing of dengue virus
advantage during their replication and spread in the plant type 2 replication in transformed C6/36 mosquito cells transcribing
host (Llave et al., 2000). As noted earlier, at least one ex- an inverted-repeat RNA derived from the virus genome. J. Virol. 76,
ample has been described of an insect virus (FHV) that 12925–12933.
suppresses RNAi in Drosophila S2 cells (Li et al., 2002a). Angell, S.M., Baulcombe, D.C., 1997. Consistent gene silencing in trans-
genic plants expressing a replicating potato virus X RNA. EMBO J.
Many virus proteins are multifunctional and some arboviral 16, 3675–3684.
proteins may have RNAi suppressor activity. This will be Beerntsen, B.T., James, A.A., Christensen, B.M., 2000. Genetics of
an important area of investigation in the future. Finally, mosquito vector competence. Microbiol. Mol. Biol. Rev. 64, 115–137.
plant geneticists have noted that developmental mutants of Bennett, K.E., Olson, K.E., Munoz Mde, L., Fernandez-Salas, I.,
plants with defined mutations in the Argonaute 1 protein Farfan-Ale, J.A., Higgs, S., Black 4th, W.C., Beaty, B.J., 2002. Varia-
tion in vector competence for dengue 2 virus among 24 collections of
are highly susceptible to virus infection, perhaps because
Aedes aegypti from Mexico and the United States. Am. J. Trop. Med.
the PTGS or RNAi pathway is defective (Williams and Hyg. 67, 85–92.
Rubin, 2002). This suggests that a functional RNAi pathway Bernstein, E., Denli, A.M., Hannon, G.J., 2001. The rest is silence. RNA
is essential in many organisms to counteract RNA virus in- 7, 1509–1521.
fection, and malfunctioning genes in the pathway could lead Black 4th, W.C., Bennett, K.E., Gorrochotegui-Escalante, N.,
to increased virus replication and pathology. It will be in- Barillas-Mury, C.V., Fernandez-Salas, I., de Lourdes Munoz, M.,
Farfan-Ale, J.A., Olson, K.E., Beaty, B.J., 2002. Flavivirus suscepti-
teresting to see whether vector competence for arboviruses
bility in Aedes aegypti. Arch. Med. Res. 33, 379–388.
can be associated with mutant gene products in the RNAi Blair, C.D., Adelman, Z.N., Olson, K.E., 2000. Molecular strategies for
pathway. We believe that further investigations of RNAi and interrupting arthropod-borne virus transmission by mosquitoes. Clin.
other RNA-mediated silencing pathways in mosquitoes will Microbiol. Rev. 13, 651–661.
be critical to the understanding of virus-vector interactions, Caplen, N.J., Zheng, Z., Falgout, B., Morgan, R.A., 2002. Inhibi-
vector competence, translational control of gene expression, tion of viral gene expression and replication in mosquito cells by
dsRNA-triggered RNA interference. Mol. Ther. 6, 243–251.
and vector developmental biology.
Carthew, R.W., 2001. Gene silencing by double-stranded RNA. Curr.
Opin. Cell Biol. 13, 244–248.
Chambers, T.J., Hahn, C.S., Galler, R., Rice, C.M., 1990. Flavivirus
Acknowledgements genome organization, expression, and replication. Annu. Rev. Micro-
biol. 44, 649–688.
Chandler, L.J., Blair, C.D., Beaty, B.J., 1998. La Crosse virus infection
This worked was supported by the National Institute of
of Aedes triseriatus (Diptera: Culicidae) ovaries before dissemination
Allergy and Infectious Diseases, grants AI34014, AI48740, of virus from the midgut. J. Med. Entomol. 35, 567–572.
TW1130, and the John T. and Catherine D. MacArthur Chu, P.W., Westaway, E.G., 1985. Replication strategy of Kunjin virus:
Foundation. evidence for recycling role of replicative form RNA as template in
semiconservative and asymmetric replication. Virology 140, 68–79.
Elbashir, S.M., Martinez, J., Patkaniowska, A., Lendeckel, W., Tuschl, T.,
2001. Functional anatomy of siRNAs for mediating efficient RNAi in
References Drosophila melanogaster embryo lysate. EMBO J. 20, 6877–6888.
Gaines, P.J., Olson, K.E., Higgs, S., Powers, A.M., Beaty, B.J., Blair,
Adams, M.D., Celniker, S.E., Holt, R.A., Evans, C.A., Gocayne, J.D., C.D., 1996. Pathogen-derived resistance to dengue type 2 virus in
Amanatides, P.G., Scherer, S.E., Li, P.W., Hoskins, R.A., Galle, R.F., mosquito cells by expression of the premembrane coding region of
George, R.A., Lewis, S.E., Richards, S., Ashburner, M., Henderson, the viral genome. J. Virol. 70, 2132–2137.
S.N., Sutton, G.G., Wortman, J.R., Yandell, M.D., Zhang, Q., Chen, Grishok, A., Pasquinelli, A.E., Conte, D., Li, N., Parrish, S., Ha, I., Baillie,
L.X., Brandon, R.C., Rogers, Y.H., Blazej, R.G., Champe, M., Pfeif- D.L., Fire, A., Ruvkun, G., Mello, C.C., 2001. Genes and mechanisms
fer, B.D., Wan, K.H., Doyle, C., Baxter, E.G., Helt, G., Nelson, C.R., related to RNA interference regulate expression of the small temporal
Gabor, G.L., Abril, J.F., Agbayani, A., An, H.J., Andrews-Pfannkoch, RNAs that control C. elegans developmental timing. Cell 106, 23–34.
C., Baldwin, D., Ballew, R.M., Basu, A., Baxendale, J., Bayraktaroglu, Gubler, D.J., 1998. Dengue and dengue hemorrhagic fever. Clin. Micro-
L., Beasley, E.M., Beeson, K.Y., Benos, P.V., Berman, B.P., Bhandari, biol. Rev. 11, 480–496.
I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74 73

Halstead, S.B., 1989. Antibody, macrophages, dengue virus infection, Lipardi, C., Wei, Q., Paterson, B.M., 2001. RNAi as random degradative
shock, and hemorrhage: a pathogenic cascade. Rev. Infect. Dis. 11, PCR: siRNA primers convert mRNA into dsRNAs that are degraded
S830–839. to generate new siRNAs. Cell 107, 297–307.
Hamilton, A.J., Baulcombe, D.C., 1999. A species of small antisense RNA Llave, C., Kasschau, K.D., Carrington, J.C., 2000. Virus-encoded suppres-
in posttranscriptional gene silencing in plants. Science 286, 950–952. sor of posttranscriptional gene silencing targets a maintenance step in
Hammond, S.M., Boettcher, S., Caudy, A.A., Kobayashi, R., Hannon, the silencing pathway. Proc. Natl. Acad. Sci. USA 97, 13401–13406.
G.J., 2001a. Argonaute 2, a link between genetic and biochemical Martinez, J., Patkaniowska, A., Urlaub, H., Luhrmann, R., Tuschl, T.,
analyses of RNAi. Science 293, 1146–1150. 2002. Single-stranded antisense siRNAs guide target RNA cleavage
Hammond, S.M., Caudy, A.A., Hannon, G.J., 2001b. Post-transcriptional in RNAi. Cell 110, 563–574.
gene silencing by double-stranded RNA. Nat. Rev. Genet. 2, 110–119. Monath, T.P., 1994. Dengue: the risk to developed and developing coun-
Hardy, W.R., Strauss, J.H., 1988. Processing the nonstructural polyproteins tries. Proc. Natl. Acad. Sci. USA 91, 2395–2400.
of Sindbis virus: study of the kinetics in vivo by using monospecific Moreira, L.A., Edwards, M.J., Adhami, F., Jasinskiene, N., James, A.A.,
antibodies. J. Virol. 62, 998–1007. Jacobs-Lorena, M., 2000. Robust gut-specific gene expression in trans-
Hoa, N.T., Keene, K.M., Olson, K.E., Zheng, L., 2003. Characterization of genic Aedes aegypti mosquitoes. Proc. Natl. Acad. Sci. USA 97,
RNA interference in an Anopheles gambiae cell line. Insect. Biochem. 10895–10898.
Mol. Biol. 33, 949–957. Myles, K.M., Pierro, D.J., Olson, K.E., 2003. A comparison of the trans-
Holt, R.A., Subramanian, G.M., Halpern, A., Sutton, G.G., Charlab, R., mission potential of two genetically distinct Sindbis viruses following
Nusskern, D.R., Wincker, P., Clark, A.G., Ribeiro, J.M., Wides, R., per os infetion of Aedes aegypti (Diptera: Culicidae). J. Med. Ento-
Salzberg, S.L., Loftus, B., Yandell, M., Majoros, W.H., Rusch, D.B., mol. 41, 95–106.
Lai, Z., Kraft, C.L., Abril, J.F., Anthouard, V., Arensburger, P., Atkin- Olson, K.E., Adelman, Z.N., Travanty, E.A., Sanchez-Vargas, I., Beaty,
son, P.W., Baden, H., de Berardinis, V., Baldwin, D., Benes, V., B.J., Blair, C.D., 2002. Developing arbovirus resistance in mosquitoes.
Biedler, J., Blass, C., Bolanos, R., Boscus, D., Barnstead, M., Cai, Insect Biochem. Mol. Biol. 32, 1333–1343.
S., Center, A., Chaturverdi, K., Christophides, G.K., Chrystal, M.A., Olson, K.E., Higgs, S., Gaines, P.J., Powers, A.M., Davis, B.S., Kam-
Clamp, M., Cravchik, A., Curwen, V., Dana, A., Delcher, A., Dew, I., rud, K.I., Carlson, J.O., Blair, C.D., Beaty, B.J., 1996. Genetically
Evans, C.A., Flanigan, M., Grundschober-Freimoser, A., Friedli, L., engineered resistance to dengue-2 virus transmission in mosquitoes.
Gu, Z., Guan, P., Guigo, R., Hillenmeyer, M.E., Hladun, S.L., Hogan, Science 272, 884–886.
J.R., Hong, Y.S., Hoover, J., Jaillon, O., Ke, Z., Kodira, C., Kokoza, Ratcliff, F.G., MacFarlane, S.A., Baulcombe, D.C., 1999. Gene silencing
E., Koutsos, A., Letunic, I., Levitsky, A., Liang, Y., Lin, J.J., Lobo, without DNA. RNA-mediated cross-protection between viruses. Plant
N.F., Lopez, J.R., Malek, J.A., McIntosh, T.C., Meister, S., Miller, J., Cell 11, 1207–1216.
Mobarry, C., Mongin, E., Murphy, S.D., O’Brochta, D.A., Pfannkoch, Ribeiro, J.M., Kidwell, M.G., 1994. Transposable elements as population
C., Qi, R., Regier, M.A., Remington, K., Shao, H., Sharakhova, M.V., drive mechanisms: specification of critical parameter values. J. Med.
Sitter, C.D., Shetty, J., Smith, T.J., Strong, R., Sun, J., Thomasova, Entomol. 31, 10–16.
D., Ton, L.Q., Topalis, P., Tu, Z., Unger, M.F., Walenz, B., Wang,
Roignant, J.Y., Carre, C., Mugat, B., Szymczak, D., Lepesant, J.A., An-
A., Wang, J., Wang, M., Wang, X., Woodford, K.J., Wortman, J.R.,
toniewski, C., 2003. Absence of transitive and systemic pathways al-
Wu, M., Yao, A., Zdobnov, E.M., Zhang, H., Zhao, Q., et al., 2002.
lows cell-specific and isoform-specific RNAi in Drosophila. RNA 9,
The genome sequence of the malaria mosquito Anopheles gambiae.
299–308.
Science 298, 129–149.
Ruiz, M.T., Voinnet, O., Baulcombe, D.C., 1998. Initiation and mainte-
Hutvagner, G., McLachlan, J., Pasquinelli, A.E., Balint, E., Tuschl, T.,
nance of virus-induced gene silencing. Plant Cell 10, 937–946.
Zamore, P.D., 2001. A cellular function for the RNA-interference
Scott, T.W., Burrage, T.G., 1984. Rapid infection of salivary glands in
enzyme Dicer in the maturation of the let-7 small temporal RNA.
Culiseta melanura with eastern equine encephalitis virus: an electron
Science 293, 834–838.
microscopic study. Am. J. Trop. Med. Hyg. 33, 961–964.
Hutvagner, G., Zamore, P.D., 2002. A microRNA in a multiple-turnover
Scott, T.W., Hildreth, S.W., Beaty, B.J., 1984. The distribution and devel-
RNAi enzyme complex. Science 297, 2056–2060.
opment of eastern equine encephalitis virus in its enzootic mosquito
Johnson, B.W., Olson, K.E., Allen-Miura, T., Rayms-Keller, A., Carlson,
vector, Culiseta melanura. Am. J. Trop. Med. Hyg. 33, 300–310.
J.O., Coates, C.J., Jasinskiene, N., James, A.A., Beaty, B.J., Higgs, S.,
1999. Inhibition of luciferase expression in transgenic Aedes aegypti Shiao, S.H., Higgs, S., Adelman, Z., Christensen, B.M., Liu, S.H., Chen,
mosquitoes by Sindbis virus expression of antisense luciferase RNA. C.C., 2001. Effect of prophenoloxidase expression knockout on the
Proc. Natl. Acad. Sci. USA 96, 13399–13403. melanization of microfilariae in the mosquito Armigeres subalbatus.
Insect Mol. Biol. 10, 315–321.
Ketting, R.F., Fischer, S.E., Bernstein, E., Sijen, T., Hannon, G.J., Plasterk,
R.H., 2001. Dicer functions in RNA interference and in synthesis of Sijen, T., Fleenor, J., Simmer, F., Thijssen, K.L., Parrish, S., Timmons,
small RNA involved in developmental timing in C. elegans. Genes L., Plasterk, R.H., Fire, A., 2001. On the role of RNA amplification
Dev. 15, 2654–2659. in dsRNA-triggered gene silencing. Cell 107, 465–476.
Knight, S.W., Bass, B.L., 2001. A role for the RNase III enzyme DCR-1 Silhavy, D., Molnar, A., Lucioli, A., Szittya, G., Hornyik, C., Tavazza,
in RNA interference and germ line development in Caenorhabditis M., Burgyan, J., 2002. A viral protein suppresses RNA silencing
elegans. Science 293, 2269–2271. and binds silencing-generated, 21- to 25-nucleotide double-stranded
Kumagai, M.H., Donson, J., della-Cioppa, G., Harvey, D., Hanley, K., RNAs. EMBO J. 21, 3070–3080.
Grill, L.K., 1995. Cytoplasmic inhibition of carotenoid biosynthesis Song, E., Lee, S.K., Wang, J., Ince, N., Ouyang, N., Min, J., Chen,
with virus-derived RNA. Proc. Natl. Acad. Sci. USA 92, 1679–1683. J., Shankar, P., Lieberman, J., 2003. RNA interference targeting Fas
Li, H., Li, W.X., Ding, S.W., 2002a. Induction and suppression of RNA protects mice from fulminant hepatitis. Nat. Med. 9, 347–351.
silencing by an animal virus. Science 296, 1319–1321. Tabara, H., Yigit, E., Siomi, H., Mello, C.C., 2002. The dsRNA bind-
Li, X., Scuderi, A., Letsou, A., Virshup, D.M., 2002b. B56-associated ing protein RDE-4 interacts with RDE-1, DCR-1, and a DExH-box
protein phosphatase 2A is required for survival and protects from helicase to direct RNAi in C. elegans. Cell 109, 861–871.
apoptosis in Drosophila melanogaster. Mol. Cell Biol. 22, 3674–3684. Uchil, P.D., Satchidanandam, V., 2003. Characterization of RNA synthesis,
Lindbo, J.A., Silva-Rosales, L., Proebsting, W.M., Dougherty, W.G., 1993. replication mechanism, and in vitro RNA-dependent RNA polymerase
Induction of a highly specific antiviral state in transgenic plants: activity of Japanese encephalitis virus. Virology 307, 358–371.
implications for regulation of gene expression and virus resistance. Vance, V., Vaucheret, H., 2001. RNA silencing in plants–defense and
Plant. Cell 5, 1749–1759. counterdefense. Science 292, 2277–2280.
74 I. Sanchez-Vargas et al. / Virus Research 102 (2004) 65–74

Westaway, E.G., Mackenzie, J.M., Kenney, M.T., Jones, M.K., Khromykh, Woodring, J.I., Higgs, S., Beaty, B.J., 1996. Natural cycles of vector-borne
A.A., 1997. Ultrastructure of Kunjin virus-infected cells: colocalization pathogens. In: Marquardt, W.C. (Ed.), The Biology of Disease Vectors.
of NS1 and NS3 with double-stranded RNA, and of NS2B with NS3, Universitiy of Colorado Press, Niwot, CO, pp. 51–72.
in virus-induced membrane structures. J. Virol. 71, 6650–6661. Zamore, P.D., Tuschl, T., Sharp, P.A., Bartel, D.P., 2000. RNAi:
Williams, R.W., Rubin, G.M., 2002. ARGONAUTE1 is required for ef- double-stranded RNA directs the ATP-dependent cleavage of mRNA
ficient RNA interference in Drosophila embryos. Proc. Natl. Acad. at 21–23 nucleotide intervals. Cell 101, 25–33.
Sci. USA 99, 6889–6894.
Virus Research 102 (2004) 75–84

Silencing structural and nonstructural genes in


baculovirus by RNA interference
C. Fabian Flores-Jasso a , Victor Julian Valdes a , Alicia Sampieri a ,
Viviana Valadez-Graham b , Felix Recillas-Targa b , Luis Vaca a,∗
a Departamento de Biologı́a Celular, Instituto de Fisiologı́a Celular, UNAM, Ciudad Universitaria, Mexico, D.F. 04510, Mexico
b Departamento de Genética Molecular, UNAM, Mexico, D.F. 04510, Mexico

Abstract

We review several aspects of RNAi and gene silencing with baculovirus. We show that the potency of RNAi in Spodoptera frugiperda
(Sf21) insect cells correlates well with the efficiency of transfection of the siRNA. Using a fluorescein-labeled siRNA we found that the siRNA
localized in areas surrounding the endoplasmic reticulum (ER). Both long (700 nucleotides long) and small (∼25 nucleotides long) interfering
RNAs were equally effective in initiating RNA interference (RNAi), and the duration of the interfering effect was indistinguishable. Even
though RNAi in Sf21 cells is very effective, in vitro experiments show that these cells fragment the long dsRNA into siRNA poorly, when
compared to HEK cells.
Finally, we show that in vivo inhibition of baculovirus infection with dsRNA homologous to genes that are essential for baculovirus infectivity
depends strongly on the amount of dsRNA used in the assays. Five hundred nanogram of dsRNA directly injected into the haemolymph of
insects prevent animal death to over 95%. In control experiments, over 96% of insects not injected with dsRNA or injected with an irrelevant
dsRNA died within a week. These results demonstrate the efficiency of dsRNA for in vivo prevention of a viral infection by virus that is very
cytotoxic and lytic in animals.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Systemic RNAi; Double stranded RNA; siRNA; Gene silencing; Baculovirus; Viral infection; In vivo inhibition

1. Introduction is a reality today. With RNAi one can knockdown as many


genes as one desires efficiently and selectively in cell cul-
Introduction of double stranded RNA (dsRNA) into a wide tures and even in living organisms. The most dramatic exam-
variety of cells and organisms results in depletion of the ple was provided with the nematode Caenorhabditis elegans,
homologue endogenous messenger RNA (mRNA) (Caplen where the term “genome-wide RNAi” has taken gene si-
et al., 2000; Chuang and Meyerowitz, 2000; Fire et al., 1998; lencing technology to a new level (Maeda et al., 2001).
Romano and Macino, 1992; Wianny and Zernicka-Goetz, Even though the mechanism by which RNAi operates is
2000). not yet completely understood, many researchers are taking
This phenomenon now known as RNA interference or advantage of this phenomenon in different ways. The rapid
RNAi in animals is related to the post transcriptional gene production of knockdown animals where genes of interest
silencing (PTGS) phenomena first discovered in plants as have been selectively silenced is just one of many possible
co-suppression (Napoli et al., 1990; Tabara, 2001; Vaucheret uses for RNAi (Voorhoeve and Agami, 2003).
et al., 2001). Another obvious use for RNAi is the selective silencing of
RNAi is rapidly becoming a powerful tool for gene silenc- viral genes essential for virulence (Lawrence, 2002; Shuey
ing (Voorhoeve and Agami, 2003). What only a few years et al., 2002). This objective is particularly important given
ago was the dream of many cell biologists, namely to have a the lack of good antiviral agents in the face of a large num-
way to explore the function of several genes simultaneously, ber of viral diseases afflicting humans. RNAi cannot only
be used as a possible antiviral therapy, but also as a pow-
erful tool to explore and better understand the basic aspects
∗ Corresponding author. Tel.: +52-5-622-5654; fax: +52-5-622-5611. of viral entry into host cells, disassembly, replication, par-
E-mail address: lvaca@ifc.unam.mx (L. Vaca). ticle assembly and export out of the infected cell. This tool

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.018
76 C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84

allows exploring simultaneously the role of several genes in ity and have produced recombinant baculovirus expressing
viral function, especially in viruses with large and complex exogenous proteins from a wide variety of organisms. As
genomes containing many putative proteins with unknown mentioned above, the polyhedrin gene is driven by its po-
function. tent promoter and thus, one can replace polyhedrin by the
In the present review we will describe some of the re- gene of interest, resulting in the production of large amounts
cent advances in RNAi using the baculovirus as a model of protein (Luckow and Summers, 1988). This protein pro-
system. We will also speculate about possible uses of RNAi duction system facilitates protein purification for functional
combined with baculovirus technologies to produce power- studies. Many proteins of different origins, including solu-
ful tools for gene function exploration. ble and plasma membrane proteins have been produced with
this system. The proteins produced in this form are func-
tional in the insect cell, an advantage that has positioned this
2. The Autographa californica nucleopolyhedrosis virus system as an excellent option to express and study the func-
tion of many G protein-coupled receptors and ionic channels
Baculoviruses comprise a large family of viruses which (Massotte, 2003).
naturally infect many different insect species (Jackson, Recently, new applications have been described for re-
1991). This type of virus has been the cause of severe eco- combinant or modified baculovirus. Special interest has been
nomic loses in the silk industry over many years. Literature directed to the use of recombinant baculovirus as gene ther-
published in the 16th century contains detailed descriptions apy vectors (Pieroni and La Monica, 2001). It has been
of the “wilting” disease in silkworms. A direct correlation shown that modified baculovirus containing suitable mam-
between wilting disease and baculovirus infection was not malian promoters can efficiently transfer genetic material
found until the beginning of the 19th century with the into mammalian cells from different tissues (Hu et al., 2003).
discovery of small crystalline bodies known as polyhedra, Another recent use for baculovirus is in the display of
obtained from infected silkworms, and the identification of peptides and proteins of interest. Because the modification
rod-shaped virions contained inside this structure. of the most abundant glycoprotein in the baculovirus nucle-
By far, the best studied member of this family is the A. ocapsid (gp64) is relatively easy, several groups have used
californica nucleopolyhedrovirus (AcNPV). This virus has recombinant baculovirus to display proteins and peptides
a large double stranded DNA genome with over 135 open fused to GP64 (Grabherr et al., 2001; Ojala et al., 2001).
reading frames (Ayres et al., 1994). The functions of most of Thus, baculovirus display has recently emerged as a new
the protein products encoded by this genome are unknown. powerful technique to identify ligands and in the presenta-
Virus morphogenesis is a rather complex process involv- tion of antigens (Ojala et al., 2001).
ing early, late and very late events (Yamagishi et al., 2003). Our group has explored recombinant baculovirus in com-
Some of the virions will bud out of the cell to infect adja- bination with RNAi technology to test two important con-
cent cells, while others will be occluded into a protein ma- cepts: (a) that RNAi is a feasible alternative to classic reverse
trix named polyhedra (Summers and Volkman, 1976). The genetics with baculovirus and (2) that interfering with genes
polyhedra are formed of the aggregates of polyhedrin, a essential for virulence prevents viral infections in vivo.
highly expressed protein produced during the late phase of Recently we have shown for the first time that injecting an-
baculovirus replication. Polyhedrin production is driven by imals with dsRNA corresponding to genes essential for bac-
the late and very strong polyhedrin promoter. This promoter ulovirus infection prevents insect death by over 95%.(Valdes
interacts selectively with baculovirus ␣-amanitin-resistant et al., 2003). The surviving insects do not show activity of
RNA polymerase. Occluded virions (contained inside the the reporter genes carried by the recombinant baculovirus,
polyhedra) will leave the cell after cell lysis. In the occluded indicating that virus replication was inhibited by the dsRNA
form, baculovirus is extremely stable and may maintain in- treatment (Valdes et al., 2003). We have explored the effect
fectivity for several years at room temperature. There are of dsRNA on two genes essential for baculovirus infectiv-
many unanswered questions regarding baculovirus morpho- ity. The first gene is gp64, the major protein found in bac-
genesis, for instance, the mechanism by which viruses are ulovirus nucleocapsid, and the second gene is ie1, an early
selected to be occluded or budded out of the infected cell. transcriptional activator essential for baculovirus replication.
The sorting system that operates to determine the fate of viri- We show that RNAi is very efficient and selective in si-
ons remains unclear, and the use of RNAi may help to elu- lencing structural (gp64) and nonstructural (ie1) baculovirus
cidate this and other intriguing questions about baculovirus genes.
morphogenesis in the future.
One of the most common systems used to study Ac-
NPV are cells originally isolated from larvae of the insect 3. RNAi is very potent and selective in insect cells
Spodoptera frugiperda (Sf21 and Sf9 cells). These cell lines
are extremely susceptible to AcNPV infection and in a few Introduction of dsRNA containing a sequence from ie1
days become highly lysed by the cytolytic viral effect. Many into Sf21 cells and living insects protects against baculovirus
groups have taken advantage of this kind of susceptibil- infection. Surprisingly, the protective effect of dsRNA was
C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84 77

very potent in both, cell cultures and whole organisms. Over showing systemic protection against a viral infection in an
95% of Sf21 cells transfected with 5 ␮g of dsRNA corre- animal by the injection of nude dsRNA.
sponding to the ie1 gene were not infected with recombinant The excellent protection conferred by the dsRNA in cell
baculovirus, even at multiplicity of infections (MOI) of five culture was most likely the result of the majority of the Sf21
and higher (Valdes et al., 2003). Furthermore, larvae from insect cells receiving dsRNA. To determine the transfection
the insect Tenebrio mollitor were systemically protected with efficiency in insect cells, we used a siRNA (21 nucleotides
as few as 0.5 ␮g of dsRNA that was directly injected in the long) conjugated with fluorescein to assess cell fluorescence
haemolymph (Valdes et al., 2003). This is the first report after transfection. Fig. 1A illustrates that the majority of

Fig. 1. Distribution of siRNA inside Sf21 insect cells. Sf21 insect cells were transfected with 0.5 ␮M of siRNA-fluorescein (QIAGEN, Hilden, Germany)
using cellfectin (INVITROGEN, Carlsbad, CA) and maintained in culture for several days as previously described (Valdes et al., 2003). Cells were
mounted on glass coverslips and observed with a confocal microscope (BIORAD, Hercules, CA). (A) Cells observed under low magnification to illustrate
that most of cells show red (brefeldin A) and green (siRNA) labeling. White arrow heads point to cells showing co-localization of brefeldin A and siRNA
(indicated by yellow color). (B) Magnification of an area in (A) to illustrate co-localization of brefeldin A and siRNA label. (C) Three-dimensional
reconstruction of two cells obtained a week after siRNA transfection showing siRNA distribution. Notice that the nuclei (Ni) of both cells do not contain
siRNA. (D) 90◦ rotation of the image in (C) to illustrate the siRNA distribution inside the cell. White arrows in (D) show cell limits. The scale bar
represents 50 ␮s for panel (A), 20 ␮s for (B and C) and 10 ␮s for (D). In all cases, cells were incubated for 5 min with 2 ␮M brefeldin A BODIPY
558/568 (MOLECULAR PROBES, Eugene, OR).
78 C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84

insect cells transfected with the fluorescein-labeled siRNA that the siRNA fluorescence was absent from the cell nuclei
fluoresce (shown in green). The transfected siRNA was ob- (Fig. 1C) and was not found dispersed throughout the cy-
served in well defined areas surrounding the endoplasmic tosol (Fig. 1D), but only in areas surrounding ER. In several
reticulum as illustrated in Fig. 1B. In this series of exper- cells we observed co localization of ER and siRNA fluo-
iments we used two selective markers of the endoplasmic rescence (Fig. 1A, arrow heads). These results indicate that
reticulum, the first one was brefeldin A (Nebenfuhr et al., siRNA is concentrated near the ER and does not cross the
2002) conjugated with BODIPY (illustrated in the figure nuclear membrane in Sf21 cells.
in red), and the second selective marker was thapsigargin A recent study has shown that Dicer, a type III RNase re-
(Inesi and Sagara, 1992), a specific inhibitor of the microso- sponsible for fragmenting long dsRNA into small interfering
mal calcium ATPase (data not shown). These results showed RNA (siRNA), is only present in the cytosol in an area that

Fig. 2. Efficient, long lasting RNAi in Sf21 insect cells. (A) GFP-positive cells measured with a cell sorter as previously described (Valdes et al.,
2003). No virus (NV) indicate uninfected cells (cell auto fluorescence), and cells transfected with dsRNA–gfp and infected with recombinant baculovirus
carrying GFP gene on days 2, 4, 6, and 9 after transfection. In all cases the GFP fluorescence was measured 48 h after infection with baculovirus, and
only transfection times varied. Cells infected with recombinant baculovirus and not exposed to dsRNA–gfp illustrate the maximum GFP fluorescence
obtained (no dsRNA). (B) Percentage of GFP-positive cells (mean ± standard deviation) obtained from 10,000 events measured with the cell sorter
(FACScalibur; Becton Dickinson). The percentage was calculated from the gray and white rectangles shown in (A) which separate GFP-positive from
GFP-negative cells (auto-fluorescence from uninfected cells). (C) Representative examples comparing the RNAi potency obtained with dsRNA–gfp (700
nucleotides) and siRNA–gfp (∼25 nucleotides) on the GFP production by recombinant baculovirus. Cells were infected with baculovirus 5 days after
transfection of the RNA. Infection was carried out in all cases with MOI of 1. Notice that both 700 nucleotide long dsRNA–gfp and the 25 nucleotide
long siRNA–gfp were equally efficient, inhibiting approximately 60% of GFP fluorescence. The mean of two independent observations is shown. All
cytometer measurements were performed as previously described (Valdes et al., 2003).
C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84 79

correlates well with markers of the ER (Provost et al., 2002). been reported in Drosophila embryos (Lipardi et al., 2001);
It remains to be explored whether the well defined spots on the other hand, have been reports that question the exis-
observed in Sf21 cells transfected with fluorescein-labeled tence of a RdRP in Drosophila and mammals (Schwarz et al.,
siRNA co-localize with Dicer. 2002; Stein et al., 2003). In fact, a single siRNA would be
Fluorescence had decayed only moderately a week after sufficient for priming the entire sequence of the gene of in-
siRNA transfection into Sf21 cells. We could not determine terest, thus producing full-length dsRNAs, which in turn can
whether the fluorescence decay observed after a week of enter a new dicing cycle to produce more siRNAs (Lipardi
transfection reflected siRNA degradation or simply fluores- et al., 2001). Whether Sf21 cells possess RdRP activity is
cein bleaching. Since in these experiments we used siRNA still an open question.
formed of 21 nucleotides, the data suggest that small inter- The duration of the fluorescence in cells transfected with
fering RNA (siRNA) may be stable inside insect cells for fluorescein-labeled siRNA correlates well with the persever-
over a week. Similar results have been observed in mam- ance of the RNAi effects on the inhibition of GFP production
malian cells, where siRNA protected against viral infection by recombinant baculovirus. This observation alone might
for several days with poliovirus (Gitlin et al., 2002), hep- be sufficient to explain the long lasting duration of RNAi in
atitis C virus (Kapadia et al., 2003) and influenza virus (Ge insect cells, but one cannot discard the involvement of an
et al., 2003). The stability of the siRNA, combined with the RdRP complex equivalent to the one reported in C. elegans.
efficient transfection observed in the experiments reported Future experiments may help to identify RdRP activity in
here, may explain the potent RNAi we have previously pub- Sf21 cells.
lished with insects (Valdes et al., 2003).
Although the fluorescence experiments suggested that
siRNAs are stable for over a week inside insect cells, it was 4. Dicer activity in insect and human cells
necessary to determine whether the RNAi effects driven by
siRNA could also be observed one week after transfection. Dicer is a type III RNase essential for RNAi. RNases
To determine if the siRNA could silence genes a week af- III are endonucleases with high specificity for dsRNA
ter transfection, insect cells were transfected with either long (Robertson et al., 1968). Dicer homologues have been iden-
dsRNA–gfp (700 nucleotides long) or the same dsRNA di- tified in bacteria, yeast, insects, nematodes and mammals
gested in vitro by recombinant human Dicer to obtain siRNA (Blaszczyk et al., 2001). This protein is responsible for
(21 nucleotides long). As illustrated in Fig. 2, interference cleaving (dicing) long dsRNA into small interfering dsRNA
of GFP production by recombinant baculovirus carrying (siRNA) fragments of ∼21–25 nucleotides long.
the GFP gene was observed even 9 days after transfecting A large body of experimental evidence strongly sug-
the cells with dsRNA–GFP. Interference efficiency decayed gests that siRNA is the functional unit in charge of direct-
slowly in correlation with the number of days after transfec- ing the degradation of the messenger RNA by the RISC
tion (Fig. 2B). Fifty percent GFP inhibition was obtained be- (RNA-induced silencing complex) (Ramaswamy and Slack,
tween days 6 and 9 post transfection. At day 9 only 30% GFP 2002). Therefore, the ability of different cells to produce
inhibition was obtained. This apparently small inhibition is, siRNA by fragmenting long dsRNA may correlate with
however, rather significant since we are not interfering with efficient RNAi.
baculovirus replication, but only with GFP production. To determine the Dicer activity in Sf21 insect cells, we
The results presented here indicate that the protective ef- performed experiments using long dsRNAs (700 nucleotides
fect of dsRNA–gfp remained active after 6–9 days after long) that were radiolabeled. This labeled dsRNA was used
transfection. Similar results were obtained using small inter- with cell extracts obtained from Sf21 insect and human em-
fering RNAs (siRNA–gfp). Both dsRNA (700 nt) and siRNA bryonic kidney (HEK) cells in dicing assays. As illustrated
(∼25 nt) were equally effective in preventing GFP produc- in Fig. 3, human cell extracts efficiently cleaved the long
tion in cells infected with recombinant baculovirus, as il- dsRNA (700 nt long) into ∼21 nucleotide siRNA fragments
lustrated in Fig. 2C. At day 5 post transfection, both long within 1 h. In comparison, Sf21 cell extracts showed re-
dsRNA and siRNA prevented 60% of GFP fluorescence. duced Dicer activity when compared to human cells, since
The long lasting effect of RNAi has not been extensively after 90 min only a small portion of the dsRNA had been
explored in different cells. In the nematode C. elegans RNAi processed into siRNA fragments (Fig. 3). The Dicing ac-
can last for a couple of generations (Fire et al., 1998). This tivity from cell extracts was compared with that of purified
lasting effect of RNAi is essentially explained by the fact that human Dicer. As shown in Fig. 3, overnight incubation of
this nematode has RNA-directed RNA polymerases (RdRP), the dsRNA with purified human Dicer resulted in fragmen-
which produce amplification of the dsRNA originally intro- tation of the dsRNA into siRNA. These experiments were
duced (Nishikura, 2001). In plants the RdRP gene (SDE-1) performed to compare the molecular weight of the products
is required only for transgene-induced PTGS, but not for resulting from cleaving with purified Dicer and cell extracts.
virus-induced PTGS (Dalmay et al., 2000). EGO-1, the or- As illustrated in the figure, HEK and Sf21 cell extracts
thologue in C. elegans of SDE-1 is essential for RNAi, but produced siRNA fragments of molecular weight equivalent
not in the germline (Smardon et al., 2000). RdRP activity has to the fragments produced by the purified enzyme. These
80 C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84

the Dicer from insect cells may not be fully functional in the
buffers used for these experiments. Another possibility may
be that insect cells have less Dicer protein than HEK cells.
Ongoing experiments may help to elucidate this point.
As shown in the previous section, both long dsRNA and
siRNA were effective after 7 days or more post transfection,
suggesting that double stranded RNA may be relatively re-
sistant to degradation in insect cells. The mechanisms by
which siRNAs degrade are still poorly explored. It is possi-
ble that degradation may take place after the double strand
is disrupted by a helicase or other yet unidentified mecha-
nism. We have maintained successfully dsRNA for several
months at −20 ◦ C. Not surprisingly, single strand RNA is
much more labile under these storage conditions and de-
grades relatively fast. Another explanation for the dsRNA
stability in Sf21 cells is that insects lack the interferon re-
sponse characteristic of higher eukaryotes (Clemens, 1997;
de Haro et al., 1996; Katze, 1995; Tan and Katze, 1999).
Thus, the stability of dsRNA could be enhanced because no
apoptosis will be induced by the presence of long dsRNA
in the cytosol of Sf21 cells.

5. Protecting living animals from a highly cytotoxic


baculovirus with dsRNA
Fig. 3. In vitro Dicer activity of Sf21 insect and human cells. In vitro RNA
cleavage experiments using extracts from Sf21 insect and HEK human We have observed systemic RNAi effects in larvae from
cells. Dicer reactions were stopped at 10, 30, 60, and 90 min as indicated
the insect T. mollitor. Injecting as few as 0.5 ␮g of dsRNA
below. Recombinant human Dicer was used in overnight experiments.
Control (C) indicates reaction being carried out in the absence of cell from ie1 directly into the haemolymph followed 24 h later
extracts or recombinant Dicer (Di) (INVITROGEN, Carlsbad, CA). MW by the injection of recombinant baculovirus prevents insect
indicates molecular weight markers. The horizontal thick arrow points to death in over 95% of the animals, as previously reported
the 20 nucleotide molecular weight marker. Methods were in brief: Either (Valdes et al., 2003). Surviving insects do not show beta
insect or human cells (∼3 × 107 ) were lysed with RIPA buffer (30 mM,
galactosidase activity or GFP fluorescence, the two reporter
Tris–HCl; 50 mM, NaCl; 0.1%, NP40; 20%, Glycerol; 3 mM, MgCl2 ; pH
6.8 final) and a cocktail of protease inhibitors. Assays were done at a final genes carried by the recombinant baculovirus.
concentration of 3 ␮g/␮l protein and contained 2 ␮g of dsRNA (700 nt We explored insect survival using different dsRNA con-
long) internally labeled with [32 P]-UTP (∼2 × 105 cpm/␮g). The final centrations and followed survival for up to 7 days. Fig. 4 il-
reaction volume was 20 ␮l. During experiments, Sf21 cells were incubated lustrates the results obtained with larvae injected with 0.05,
at 27 ◦ C and HEK cells were incubated at 37 ◦ C. Phenol/chloroform (5 ␮l)
0.1, and 0.5 ␮g dsRNA–ie1 24 h prior to the challenge with
were added to stop reactions. Assays were separated on 10% PAGE-8M
urea. Gels were dried, exposed to an amplifying screen and analyzed with recombinant baculovirus. The dsRNA–ie1 was synthesized
a PhosphorImager (AMERSHAM BIOSCIENCES, Sunnyvale, CA). from nucleotides 19-470 from the ie1 gene as previously
described (Valdes et al., 2003).
As illustrated in the figure, insect survival declined
observations indicate that the fragmentation of the long when the animals were injected with reducing amounts
dsRNA using the cells extracts result from the activity of of dsRNA–ie1. Dead insects previously injected with
a type III RNase. Increasing the amount of purified Dicer 0.1 ␮g dsRNA–ie1 showed a lighter blue color and reduced
used in the assay or reducing the amount of dsRNA both re- GFP fluorescence when compared to insects injected with
sulted in complete fragmentation of the substrates (data not 0.05 ␮g dsRNA–ie1. These results show that the expression
shown). in infected animals of the two reporter genes carried by
These results suggest that Sf21 cells are less efficient in the recombinant baculovirus (beta galactosidase and GFP)
cleaving long dsRNA than HEK cells. There are several ex- correlates well with the survival rate. This observation is
planations for this observation. For instance, it is possible more clearly illustrated in Fig. 4B, where the GFP fluo-
that the reduced Dicer activity of insect cell extracts may rescence intensity obtained from larvae injected with 0.05,
result of inadequate conditions for the assay. Although we 0.1, and 0.5 ␮g dsRNA–ie1 prior to baculovirus infection
explored different buffer solutions in which we changed pH, is plotted. Notice the correlation between dead insects col-
magnesium, temperature and ATP contents, all yielded sim- lected at day 7 after baculovirus infection and increased
ilar results. However, we cannot discard the possibility that GFP fluorescence.
C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84 81

Fig. 4. In vivo protection of insect larvae from baculovirus infection by the use of dsRNA. Panel (A) larvae from the insect T. mollitor (mealworm
beetle) injected in the haemolymph with the 0.05, 0.1, and 0.5 ␮g of dsRNA–ie1 or 0.5 ␮g dsRNA–gfp 24 h prior to the injection of 2 ␮l of recombinant
baculovirus AcNPV–gfp (viral stock with a titer of 1 × 10−7 pfu). To the left representative examples of larvae injected with 2 ␮l of 0.05% X-gal in
dimethyl sulfoxide on day 7 after baculovirus injection are shown. Notice the blue color in larvae infected with recombinant baculovirus and not protected
by the dsRNA–ie1. Notice also the lighter blue color obtained from the larva injected with 0.1 ␮g dsRNA–ie1. The middle panel shows confocal images
illustrating the GFP fluorescence from the head of larvae under the different experimental conditions mentioned above. Notice the lack of blue color
(beta galactosidase activity) and GFP fluorescence in larvae injected with 0.5 ␮g dsRNA–ie1. The plots to the right indicate the percentage of dead
insects at different days post infection with baculovirus. The numbers in the inserts indicate the number of dead larvae and the total larvae injected
for each condition (e.g. 57 dead larvae from 60 injected with 0.05 ␮g dsRNA–ie1). Panel (B), the plot to the left illustrates the number of dead larvae
obtained at day 7 post infection for the three different dsRNA–ie1 concentrations assayed. The plot to the right shows the GFP fluorescence intensity (in
arbitrary units, AU) obtained at day 7 post infection from the head of randomly selected larvae, injected with 0.5, 0.1, and 0.05 ␮g dsRNA–ie1 (n = 10
for each condition). Solid circles indicate dead larvae and open circles living larvae. Autofluorescence was subtracted from all experiments. For details
on experimental conditions see Valdes et al. (2003).
82 C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84

These results demonstrate that the protective effect of type virus or a recombinant virus, which are easy to obtain
dsRNA–ie1 is dose-dependent. When 0.05 ␮g dsRNA–ie1 and replicate, and use RNAi with insect cells to silence the
is used, only 3 out of 60 (5%) larvae injected survived bac- genes of interest. We have successfully silenced two genes
ulovirus infection (Fig. 4A, upper panel). All insects showed essential for infectivity in the AcNPV baculovirus, gp64 and
beta galactosidase activity and high GFP fluorescence. The ie1.
amount of GFP fluorescence obtained from these animals There are several important lessons we have learned from
was indistinguishable from that obtained with larvae not in- these experiments. A large body of evidence shows that
jected with dsRNA–ie1 (data not shown), suggesting that this GP64 is a protein essential for virus entry into insect cells
dosage of dsRNA is insufficient to protect insects from bac- (Robertson et al., 1974). Deletion of this protein resulted
ulovirus infection. Injecting larvae with 0.5 ␮g dsRNA–gfp in baculovirus that cannot infect cells and therefore cannot
prevented GFP fluorescence in infected animals but did not be replicated (Monsma et al., 1996). Thus studying the role
alter insect survival to the baculovirus infection. Insects in- of GP64 in baculovirus infectivity cannot be accomplished
jected with dsRNA–gfp turned blue upon X-gal injection, in- with traditional knockout technologies. We have used RNAi
dicating the presence of beta galactosidase in the baculovirus to produce viruses lacking GP64, by transfecting cells with
infected animals (Fig. 4A). These results demonstrate that dsRNA formed of 300 nucleotides of gp64 (dsRNA–gp64)
the in vivo RNAi effects are selective for the dsRNA se- following infection with recombinant baculovirus carrying a
quence used in the experiments, since we could interfere wild type copy of gp64. Virus produced by cells transfected
with GFP but not beta galactosidase. The high mortality rate with dsRNA–gp64 lacked this glycoprotein, as we have pre-
observed in animals injected with dsRNA–gfp demonstrates viously shown (Valdes et al., 2003). The virus produced by
that baculovirus replication and infectivity were not affected these cells could not bud out of the insect cells. Virions were
by irrelevant dsRNAs. identified inside the cells nuclei by electron microscopy, but
Since the dsRNA injection was into the haemolymph and they could not be collected from the supernatant of infected
baculovirus infection in insects is systemic, the protective cells even at 96 h post infection. These results strongly sug-
effects of dsRNA–ie1 strongly suggest that the effects of gest that GP64 glycoprotein is not only needed for virus en-
RNAi in these larvae are systemic. Systemic RNAi has been try (as previously shown by Monsma et al., 1996), but also
observed in plants (Palauqui et al., 1997) and several animals for budding out of infected cells.
including C. elegans (Fire et al., 1998; Winston et al., 2002) RNAi can also be used to identify novel proteins in the
and mice (Sorensen et al., 2003). More recently it has been genome of baculovirus. Recently, RNAi has been used
shown systemic and inheritable RNAi in the beetle Tribolium to identify a novel antiapoptotic gene (Op-iap3) from the
castaneum (Bucher et al., 2002). baculovirus Orgyia pseudotsugata M nucleopolyhedrovirus
(Means et al., 2003). Interfering with Op-iap3 resulted in
apoptosis in insect cells infected with this baculovirus, and
6. Conclusions virus production was markedly reduced due to early cell
death (Means et al., 2003). Thus RNAi provides a rapid
We have used the baculovirus AcNPV to explore two and efficient method for the screening of protein function
hypotheses: (a) that interfering with the synthesis of proteins in different members from the baculovirus family.
essential for baculovirus infectivity prevents in vitro and in
vivo viral infections and (b) that RNAi is a powerful tool to 6.2. Baculovirus as RNAi vectors
explore the morphogenesis of a complex virus.
We have shown, for the first time, the use of RNAi to Major efforts are directed worldwide to find a suitable
selectively inhibit viral genes essential for baculovirus vir- vector to induce RNAi in living organisms. Researchers
ulence. Furthermore, systemic application of dsRNA into a have injected nude siRNA or combined with lipids in mice
living animal protected insects from infection by this very (Sorensen et al., 2003). However, it has been difficult to
infective and cytolytic virus. These results demonstrate the obtain systemic distribution of the siRNA. More recently,
potential of using dsRNA as a therapeutic drug in the treat- several groups have developed viral vectors based on aden-
ment or prevention of viral infections in animals. ovirus (Shen et al., 2003) and lentivirus (Stewart et al., 2003)
carrying a polymerase III promoter (such as H1 or U6) and
6.1. Exploring the morphogenesis of a complex virus with a hairpin sequence to produce siRNAs. Although both aden-
RNAi ovirus and lentivirus are excellent vectors to transfer genetic
material in mammalian cells, the production of these vec-
Understanding the functional role of several genes from tors is time consuming, making the exploration of several
a complex genome the size of the AcNPV genome is not an siRNA sequences difficult. On the other hand, it is well es-
easy task. The production of knockout virus is a lengthy pro- tablished that baculovirus can transfer genetic material into
cess. On many occasions, the gene deletions result in virus mammalian cells (Hu et al., 2003), yet this virus is relatively
that cannot be replicated and therefore cannot be studied. easy to prepare, and production of high titer viral stocks is
The advantage of using RNAi is that one can have the wild achieved in a short period of time. Thus, the development
C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84 83

of a technology for RNAi based on baculovirus to trans- focuses on strategic infection and feeding. Biotechnol. Prog. 19, 373–
fer genetic material in mammalian cells is feasible. This is 379.
Inesi, G., Sagara, Y., 1992. Thapsigargin, a high affinity and global
one of many possible uses for baculovirus technology in the inhibitor of intracellular Ca2+ transport ATPases. Arch. Biochem.
RNAi field. Baculovirus can also be used as RNAi vectors Biophys. 298, 313–317.
in insects. One of the advantages of baculovirus is that they Jackson, J.A., 1991. Baculovirus expression systems and insect cells.
are highly infective and capable of producing large amounts Bioprocess Technol. 13, 402–413.
of dsRNA. The major disadvantage is that baculovirus are Kapadia, S.B., Brideau-Andersen, A., Chisari, F.V., 2003. Interference of
hepatitis C virus RNA replication by short interfering RNAs. Proc.
also highly cytolytic in insect cells, which complicates the Natl. Acad. Sci. U. S. A. 100, 2014–2018.
interpretation of the gene interference results. Katze, M.G., 1995. Regulation of the interferon-induced PKR: can viruses
cope? Trends Microbiol. 3, 75–78.
Lawrence, D., 2002. RNAi could hold promise in the treatment of HIV.
Lancet 359, 2007.
Acknowledgements Lipardi, C., Wei, Q., Paterson, B.M., 2001. RNAi as random degradative
PCR: siRNA primers convert mRNA into dsRNAs that are degraded
The excellent services from the Molecular Biology and to generate new siRNAs. Cell 107, 297–307.
Microscopy units and the library at the Instituto de Fisi- Luckow, V.A., Summers, M.D., 1988. Signals important for high-level
ologia Celular are greatly appreciated. Part of this work expression of foreign genes in Autographa californica nuclear poly-
hedrosis virus expression vectors. Virology 167, 56–71.
was produced with equipment donated by the Alexander Maeda, I., Kohara, Y., Yamamoto, M., Sugimoto, A., 2001. Large-
von Humboldt-Stiftung and a grant from Fundación Miguel scale analysis of gene function in Caenorhabditis elegans by
Alemán to LV. high-throughput RNAi. Curr. Biol. 11, 171–176.
Massotte, D., 2003. G protein-coupled receptor overexpression with the
baculovirus-insect cell system: a tool for structural and functional
studies. Biochim. Biophys. Acta 1610, 77–89.
Means, J.C., Muro, I., Clem, R.J., 2003. Silencing of the baculovirus
References
Op-iap3 gene by RNA interference reveals that it is required for pre-
vention of apoptosis during Orgyia pseudotsugata M nucleopolyhe-
Ayres, M.D., Howard, S.C., Kuzio, J., Lopez-Ferber, M., Possee, R.D., drovirus infection of Ld652Y cells. J. Virol. 77, 4481–4488.
1994. The complete DNA sequence of Autographa californica nuclear Monsma, S.A., Oomens, A.G., Blissard, G.W., 1996. The GP64 enve-
polyhedrosis virus. Virology 202, 586–605. lope fusion protein is an essential baculovirus protein required for
Blaszczyk, J., Tropea, J.E., Bubunenko, M., Routzahn, K.M., Waugh, cell-to-cell transmission of infection. J. Virol. 70, 4607–4616.
D.S., Court, D.L., Ji, X., 2001. Crystallographic and modeling studies Napoli, C., Lemieux, C., Jorgensen, R., 1990. Introduction of a chimeric
of RNase III suggest a mechanism for double-stranded RNA cleavage. chalcone synthase gene into petunia results in reversible co-suppression
Structure (Camb.) 9, 1225–1236. of homologous genes in trans. Plant Cell 2, 279–289.
Bucher, G., Scholten, J., Klingler, M., 2002. Parental RNAi in Tribolium Nebenfuhr, A., Ritzenthaler, C., Robinson, D.G., 2002. Brefeldin A: deci-
(Coleoptera). Curr. Biol. 12, R85–R86. phering an enigmatic inhibitor of secretion. Plant Physiol. 130, 1102–
Caplen, N.J., Fleenor, J., Fire, A., Morgan, R.A., 2000. dsRNA-mediated 1108.
gene silencing in cultured Drosophila cells: a tissue culture model for Nishikura, K., 2001. A short primer on RNAi: RNA-directed RNA poly-
the analysis of RNA interference. Gene 252, 95–105. merase acts as a key catalyst. Cell 107, 415–418.
Chuang, C.F., Meyerowitz, E.M., 2000. Specific and heritable genetic Ojala, K., Mottershead, D.G., Suokko, A., Oker-Blom, C., 2001. Specific
interference by double-stranded RNA in Arabidopsis thaliana. Proc. binding of baculoviruses displaying gp64 fusion proteins to mam-
Natl. Acad. Sci. U. S. A. 97, 4985–4990. malian cells. Biochem. Biophys. Res. Commun. 284, 777–784.
Clemens, M.J., 1997. PKR—a protein kinase regulated by double-stranded Palauqui, J.C., Elmayan, T., Pollien, J.M., Vaucheret, H., 1997. Systemic
RNA. Int. J. Biochem. Cell Biol. 29, 945–949. acquired silencing: transgene-specific post-transcriptional silencing is
Dalmay, T., Hamilton, A., Rudd, S., Angell, S., Baulcombe, D.C., 2000. transmitted by grafting from silenced stocks to non-silenced scions.
An RNA-dependent RNA polymerase gene in Arabidopsis is required EMBO J. 16, 4738–4745.
for posttranscriptional gene silencing mediated by a transgene but not Pieroni, L., La Monica, N., 2001. Towards the use of baculovirus as a
by a virus. Cell 101, 543–553. gene therapy vector. Curr. Opin. Mol. Ther. 3, 464–467.
de Haro, C., Mendez, R., Santoyo, J., 1996. The eIF-2alpha kinases and Provost, P., Dishart, D., Doucet, J., Frendewey, D., Samuelsson, B., Rad-
the control of protein synthesis. FASEB J. 10, 1378–1387. mark, O., 2002. Ribonuclease activity and RNA binding of recombi-
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver. S.E., Mello, nant human Dicer. EMBO J. 21, 5864–5874.
C.C., 1998. Potent and specific genetic interference by double-stranded Ramaswamy, G., Slack, F.J., 2002. siRNA. A guide for RNA silencing.
RNA in Caenorhabditis elegans. Nature 391, 806–811. Chem. Biol. 9, 1053–1055.
Ge, Q., McManus, M.T., Nguyen, T., Shen, C.H., Sharp, P.A., Eisen, Robertson, H.D., Webster, R.E., Zinder, N.D., 1968. Purification and
H.N., Chen, J., 2003. RNA interference of influenza virus production properties of ribonuclease III from Escherichia coli. J. Biol. Chem.
by directly targeting mRNA for degradation and indirectly inhibiting 243, 82–91.
all viral RNA transcription. Proc. Natl. Acad. Sci. U. S. A. 100, Robertson, J.S., Harrap, K.A., Longworth, J.F., 1974. Baculovirus mor-
2718–2723. phogenesis: the acquisition of the virus envelope. J. Invertebr. Pathol.
Gitlin, L., Karelsky, S., Andino, R., 2002. Short interfering RNA confers 23, 248–251.
intracellular antiviral immunity in human cells. Nature 418, 430–434. Romano, N., Macino, G., 1992. Quelling: transient inactivation of gene
Grabherr, R., Ernst, W., Oker-Blom, C., Jones, I., 2001. Developments in expression in Neurospora crassa by transformation with homologous
the use of baculoviruses for the surface display of complex eukaryotic sequences. Mol. Microbiol. 6, 3343–3353.
proteins. Trends Biotechnol. 19, 231–236 Schwarz, D.S., Hutvagner, G., Haley, B., Zamore, P.D., 2002. Evidence
Hu, Y.C., Tsai, C.T., Chang, Y.J., Huang, J.H., 2003. Enhancement and that siRNAs function as guides, not primers, in the drosophila and
prolongation of baculovirus-mediated expression in mammalian cells: human RNAi pathways. Mol. Cell 10, 537–548.
84 C.F. Flores-Jasso et al. / Virus Research 102 (2004) 75–84

Shen, C., Buck, A.K., Liu, X., Winkler, M., Reske, S.N., 2003. Gene Tabara, H., 2001. RNAi and PTGS in eukaryotes. Tanpakushitsu Kakusan
silencing by adenovirus-delivered siRNA. FEBS Lett. 539, 111–114. Koso 46, 2017–2024.
Shuey, D.J., McCallus, D.E., Giordano, T., 2002. RNAi: gene-silencing Tan, S.L., Katze, M.G., 1999. The emerging role of the interferon-induced
in therapeutic intervention. Drug Discov. Today 7, 1040–1046. PKR protein kinase as an apoptotic effector: a new face of death? J.
Smardon, A., Spoerke, J.M., Stacey, S.C., Klein, M.E., Mackin, N., Maine, Interferon Cytokine Res. 19, 543–554.
E.M., 2000. EGO-1 is related to RNA-directed RNA polymerase Valdes, V.J., Sampieri, A., Sepulveda, J., Vaca, L., 2003. Using
and functions in germ-line development and RNA interference in C. double-stranded RNA to prevent in vitro and in vivo viral infections
elegans. Curr. Biol. 10, 169–178. by recombinant baculovirus. J. Biol. Chem. 278, 19317–19324.
Sorensen, D.R., Leirdal, M., Sioud, M., 2003. Gene silencing by systemic Vaucheret, H., Beclin, C., Fagard, M., 2001. Post-transcriptional gene
delivery of synthetic siRNAs in adult mice. J. Mol. Biol. 327, 761– silencing in plants. J. Cell Sci. 114, 3083–3091.
766. Voorhoeve, P.M., Agami, R., 2003. Knockdown stands up. Trends Biotech-
Stein, P., Svoboda, P., Anger, M., Schultz, R.M., 2003. RNAi: mammalian nol. 21, 2–4.
oocytes do it without RNA-dependent RNA polymerase. RNA 9, 187– Wianny, F., Zernicka-Goetz, M., 2000. Specific interference with gene
192. function by double-stranded RNA in early mouse development. Nat.
Stewart, S.A., Dykxhoorn, D.M., Palliser, D., Mizuno, H., Yu, E.Y., An, Cell Biol. 2, 70–75.
D.S., Sabatini, D.M., Chen, I.S., Hahn, W.C., Sharp, P.A., Weinberg, Winston, W.M., Molodowitch, C., Hunter, C.P., 2002. Systemic RNAi in
R.A., Novina, C.D., 2003. Lentivirus-delivered stable gene silencing C. elegans requires the putative transmembrane protein SID-1. Science
by RNAi in primary cells. RNA 9, 493–501. 295, 2456–2459.
Summers, M.D., Volkman, L.E., 1976. Comparison of biophysical and Yamagishi, J., Isobe, R., Takebuchi, T., Bando, H., 2003. DNA microarrays
morphological properties of occluded and extracellular nonoccluded of baculovirus genomes: differential expression of viral genes in two
baculovirus from in vivo and in vitro host systems. J. Virol. 17, 962– susceptible insect cell lines. Arch. Virol. 148, 587–597.
972.
Virus Research 102 (2004) 85–96

RNA interference as a new biotechnological tool for the control of virus


diseases in plants
Francisco Tenllado, César Llave, José Ramón Dı́az-Ruı́z∗
Departamento de Biologı́a de Plantas, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, Madrid 28040, Spain

Abstract

RNA silencing occurs in a wide variety of organisms, including protozoa, fungi, plants and animals and involves recognition of a target
RNA and initiation of a sequence-specific RNA degradation pathway in the cytoplasm. In the last few years, there have been considerable
advances in our understanding of post-transcriptional gene silencing (PTGS). This mechanism is conceived as a natural antiviral defense
system in plants that is activated as a response to double-stranded RNA (dsRNA) formed during virus replication. To develop new approaches
for plant protection against virus diseases based on PTGS we have expanded previous findings on RNA interference (RNAi) in animals by
using dsRNA to specifically interfere with virus infection in plants. This approach differs from strategies based on transgenic expression of
RNAs but still relies on PTGS as a means to achieve pathogen-derived resistance (PDR). Our findings suggest that exogenously supplied
dsRNA could form the basis for the development of an environmentally safe, new biotechnological tool aimed at protecting crops against
virus diseases, provided that some limitations of the current status of the approach could be overcome.
© 2004 Elsevier B.V. All rights reserved.

Keywords: RNA interference; Post-transcriptional gene silencing; Double-stranded RNA; Virus diseases; Plant protection

1. Introduction specific step during virus multiplication in the plant. Many


papers have been devoted to RNA-mediated virus resistance,
A number of homology-dependent gene silencing phe- nearly all of which can be classified as a specific manifesta-
nomena have been reported in different organisms. They de- tion of PTGS in plants (reviewed by Vazquez Rovere et al.,
scribe specific variants of the same basic mechanism which 2002; Goldbach et al., 2003). In this particular case of PTGS,
acts mainly at the post-transcriptional level of gene ex- both the viral RNA and the virus-derived transgene mRNA
pression (Hammond et al., 2001). Post-transcriptional gene are targeted for degradation rendering plants that display
silencing (PTGS) was first reported in plants 13 years ago as protection against virus infection (Baulcombe, 1999).
a coordinated and reciprocal inactivation of host genes and A major breakthrough in the silencing history was the
transgenes encoding the same RNA (Napoli et al., 1990). discovery of a gene silencing response in the nematode
Shortly after, a similar phenomenon was observed in the Caenorhabditis elegans (Fire et al., 1998). This phe-
filamentous fungus Neurospora crassa and termed quelling nomenon, in which experimentally introduced double-
(Romano and Macino, 1992). Historically, the principle stranded RNA (dsRNA) leads to loss of expression of the
of pathogen-derived resistance (PDR) as a way to confer corresponding cellular gene by sequence-specific RNA
protection against viral infection has been strongly linked degradation, was called RNA interference (RNAi). RNAi
to genetic manipulation of plants in order to obtain trans- could be induced by injection of dsRNA into any site of
genic resistance (Sanford and Johnston, 1985). This well- the animal (Fire et al., 1998), by feeding the worms on
established principle involves the use of virus-derived genes engineered Escherichia coli producing dsRNA against the
or genome fragments with the purpose to interfere with a target gene (Timmons and Fire, 1998), by in vivo transcrip-
tion of dsRNA from transgene promoters (Tavernarakis
∗ Corresponding author. Present address: Centro de Investigaciones
et al., 2000), or simply by soaking the worms in a solution
Biológicas, Ramiro de Maeztu 9, 28040 Madrid, Spain.
containing the dsRNA (Tabara et al., 1998). Since this dis-
Tel.: +34-1-5611800; fax: +34-1-5627518. covery emerged in C. elegans, RNAi has been documented
E-mail address: jrdiazruiz@cib.csic.es (J.R. Dı́az-Ruı́z). in a wide variety of animals, ranging from protozoans, such

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.019
86 F. Tenllado et al. / Virus Research 102 (2004) 85–96

as Trypanosome, to metazoans, such as Drosophila and matin (Volpe et al., 2002) in different organisms. More-
mammals (Tijsterman et al., 2002). over, recent findings have pointed to an even more central
A growing body of biochemical and genetic data has role for RNA silencing in an RNA-based regulatory network
further demonstrated that plants, animals and yeasts share aimed at regulating gene expression in plants and animals
related mechanisms of specific degradation of RNAs in (Hutvágner and Zamore, 2002; Llave et al., 2002). Details
which double-stranded forms of RNA are initiator molecules on molecular mechanisms of PTGS/RNAi and its natural
(Dalmay et al., 2000; Fagard et al., 2000; Bernstein et al., role for endogenous gene regulation have been considered in
2001). In plants, dsRNA or self-complementary hairpin previous reviews (Hannon, 2002; Pasquinelli and Ruvkun,
RNA that are transcribed from engineered inverted repeats 2002; Denli and Hannon, 2003).
were shown to be potent inducers of a gene silencing re- The concept of obtaining resistance to viruses by transfor-
sponse when directed against trangenes (Hamilton et al., mation with genes derived from the genome of the pathogen
1998; Waterhouse et al., 1998; Johansen and Carrington, is a well-exploited and highly effective procedure to fight
2001). Furthermore, plants transformed with constructs that viruses as causal agents of diseases in plants. In the past,
produce RNAs capable of duplex formation induce virus im- we have genetically engineered Nicotiana plants using viral
munity with almost 100% efficiency when targeted against gene sequences with a view of providing the basis for a po-
viruses (Smith et al., 2000; Wang et al., 2000; Kalantidis tential control of virus diseases in crops (Peña et al., 1994;
et al., 2002). Tenllado et al., 1995, 1996). In particular, we were interested
As early as 1978, virus-specific dsRNA forms were iso- in tobamovirus strains which are able to infect commer-
lated from infected plant tissues (Dı́az-Ruı́z and Kaper, cial pepper cultivars with genetic resistances introduced by
1978). More recently, several lines of evidence indicate that classical breeding against common strains of tobamoviruses.
virus-induced gene silencing (VIGS), a particular manifes- Like most cases in PDR, transgenic resistance against Pepper
tation of PTGS in which viruses are both triggers and targets mild mottle virus (PMMoV) using replicase gene sequences
of silencing, has evolved into a viral surveillance system was based on PTGS (Tenllado and Dı́az-Ruı́z, 1999). How-
triggered by dsRNA produced during the intermediate steps ever, because most pepper varieties are recalcitrant to ge-
of virus replication (Ratcliff et al., 1999; Waterhouse et al., netic transformation, control of diseases caused by pepper
2001). Since dsRNA does not naturally occur in the cyto- strains of tobamoviruses using this strategy await future
plasm of plant cells, recognition of nucleic acid structures progress. One important aim of our current research is to
that are present only in infected cells seems to induce a develop new biotechnological approaches for plant protec-
natural line of defense against viral infections (Voinnet, tion against virus diseases based on PTGS. In this sense,
2001). Moreover, it has been speculated that silencing pro- we have expanded previous findings on RNAi in animals
cesses may also serve as an antiviral system in vertebrates by using dsRNA to specifically interfere with virus infec-
(Li et al., 2002). In the PTGS-associated antiviral response, tion in plants. This approach differs from strategies based
viral dsRNA is first processed by an RNase III-like nucle- on transgenic expression of RNAs but still relies on PTGS
ase, which was named Dicer in Drosophila melanogaster as a means to achieve PDR. Some of the results obtained in
and for which several homologs in Arabidopsis have been our laboratory are presented here.
found, into 21–25 nt dsRNAs (small interfering RNAs, siR-
NAs) that guide another nuclease complex (RNA-induced
silencing complex, RISC) to cleave the RNA genome of 2. RNAi in plants
the invading virus (Hamilton and Baulcombe, 1999; Tang
et al., 2003). As a counterdefensive strategy, many viruses The essence of RNAi technology is the delivery of dsRNA
encode proteins that suppress silencing at different points as a potent activator of RNA silencing into an organism,
of the PTGS pathway (Mlotshwa et al., 2002). Thus, the or cell, with the purpose of triggering sequence-specific
discovery that viruses are inducers, and targets, and may degradation of homologous target RNAs. As we mentioned
carry suppressors of PTGS adds further support to the in- above, dsRNA can be delivered by stably transforming
volvement of dsRNA-induced silencing processes as an plants with transgenes that express a self-complementary
innate defense mechanism against virus infection. RNA. The resulting transcript hybridizes with itself to
In the last few years, there have been considerable ad- form a hairpin structure that comprises a single-stranded
vances in our understanding of the molecular mechanism of loop region (usually corresponding to an intron) and a
PTGS. We now know that both PTGS and RNAi are man- base-paired stem, which mimics the dsRNA structure that
ifestations of a broader set of gene silencing phenomena induces RNAi. It has been shown that transgene constructs
common to most, if not all, eukaryotes. For instance, one encoding intron-spliced RNA with hairpin structure give
emerging theme is that besides an important biological role stable silencing with almost 100% efficiency when directed
in protecting the organism against exogenous nucleic acids, against viruses (Smith et al., 2000; Kalantidis et al., 2002).
PTGS-related processes are implicated in maintenance of The major technical limitation for this technology is that
genome stability (Ketting et al., 1999; Djikeng et al., 2001; many important plant crop species are difficult or impossi-
Hamilton et al., 2002) and in the formation of heterochro- ble to transform, precluding the constitutive expression of
F. Tenllado et al. / Virus Research 102 (2004) 85–96 87

constructs directing production of dsRNA. Moreover, ssRNA or dsRNA derived from the replicase gene of PM-
questions concerning the potential ecological impact of MoV (54-kDa-protein region) to inhibit local lesion forma-
virus-resistant transgenic plants have so far significantly tion elicited by PMMoV was evaluated using two different
limited their use (Tepfer, 2002). hypersensitive hosts, Nicotiana tabacum cv. Xanthi nc and
When the RNAi technology was first applied in mam- Capsicum chinense. We found that mechanical inoculation
malian cells, investigators had to tackle the fact that dsRNA of neither sense ssRNA, antisense ssRNA nor cDNA corre-
molecules longer than 30 base pairs trigger a non-specific sponding to the 54-kDa-protein region of the virus caused
interferon-mediated response that leads to general suppres- reduction in the number of local lesions elicited by PMMoV
sion of protein synthesis and ultimately cell death (Gil in plants. However, virus infectivity was completely blocked
and Esteban, 2000). Afterwards, an outstanding discovery by coinoculation of PMMoV particles with a 977-bp dsRNA
demonstrated that RNAi can also be initiated in different cul- derived from the 54-kDa-coding region (Fig. 1). Further-
tured mammalian cell lines by transfecting synthetic siRNA more, we found interference with virus infection only when
duplexes (Elbashir et al., 2001). Indeed, sequence-specific dsRNA molecules share sequence identity with the virus.
silencing at the post-transcriptional level can be achieved Coinoculation of PMMoV together with a 1483-bp dsRNA
in Drosophila embryos, C. elegans or cultured mammalian corresponding to most of the Helper component-Proteinase
cells by injection of synthetic dsRNA molecules. From (HC-Pro) gene of TEV (TEV-HC dsRNA), did not have any
this discovery emerged the notion that in vitro transcribed effect on PMMoV infectivity. Thus, these results clearly
dsRNA might also act as an activator of RNAi in plants. demonstrate that RNAi can be successfully triggered against
Moreover, plant cells seem to lack the mechanisms that plant viruses by mechanically inoculating in vitro tran-
trigger non-specific inhibition of gene expression by long scribed dsRNA to leaf cells together with the target virus.
dsRNA, such as the activation of a protein kinase–mediated Moreover, we also found that homologous dsRNA was
antiviral defense presents in mammalian cells (Cayley et al., competent to interfere with virus infection in a systemic host
1982). Thus, the use of synthetic siRNAs to induce RNAi infection. Nicotiana benthamiana plants that had been in-
is not obligatory in plants as it is in mammalian systems. oculated with mixtures of PMMoV particles and different
In our laboratory we have made intensive research efforts PMMoV-derived dsRNAs were resistant to systemic virus
to assess the potential of dsRNA-mediated interference in infection. We further confirmed that dsRNA molecules as
plant virus infections. As an initial approach, we investi- short as 596 bp from either the 54- or 30-kDa coding genes
gated whether direct delivery by mechanical inoculation of were effective in protecting plants against PMMoV infec-
dsRNA together with either virion particles or viral tran- tion. Not only did most plants not display disease symp-
scripts could interfere with infection in a sequence-specific toms during the completion of their life cycles, but more im-
manner (Tenllado and Dı́az-Ruı́z, 2001). We made use of portantly, neither viral RNA nor viral particles accumulated
three viruses belonging to the tobamovirus (PMMoV), po- to detectable levels in both inoculated and systemically in-
tyvirus (Tobacco etch virus, TEV) and alfamovirus (Alfalfa fected N. benthamiana leaves. We found that plants coinocu-
mosaic virus, AMV) groups, as representative examples lated with PMMoV particles and a homologous 315 bp long
in the diversity of positive-strand RNA viruses in plants. dsRNA displayed viral symptoms with a delay of 1–2 days
Sense single-stranded RNA (ssRNA) and antisense ssRNA compared to plants inoculated with PMMoV alone, with vi-
corresponding to different parts of the viral genomes were ral RNA accumulating at levels similar to the control. This
transcribed in vitro from partial cDNA clones and even- may indicate that the ability of dsRNA to specifically in-
tually annealed to each other to produce dsRNAs. In our terfere with virus infection seems to require a minimum
initial experiments, the ability of sense ssRNA, antisense length of dsRNA. In fact, RNAi has also been reported to

Fig. 1. Specific interference with PMMoV infection by dsRNA in a hypersensitive host. Response of N. tabacum cv. ‘Xanthi nc’ to PMMoV alone (left
halves of leaves) or to a combination of PMMoV plus either 54-kDa antisense (as) RNA (A), 54-kDa dsRNA (B) or TEV-HC dsRNA (C) (right halves
of the leaves). Similar number of local lesions was observed in both halves of the leaves in panels (A) and (C). No visible local response was observed
in the half-leaf inoculated with PMMoV plus 54-kDa dsRNA (B).
88 F. Tenllado et al. / Virus Research 102 (2004) 85–96

depend on the length of dsRNA in plants, C. elegans and of dsRNA required to ensure that most viral RNA pene-
Drosophila (Fire et al., 1998; Hammond et al., 2000). Re- trates into the damaged cells in combination with appropri-
combinant viruses carrying fragments of usually 300–800 ate quantities of dsRNA. From a mechanistic perspective, all
nucleotides derived from the target gene efficiently trigger evidence favors the hypothesis that dsRNA interferes with
RNAi of a transgene in plants, although fragments as short virus infection at the cell level in the inoculated leaves: (i)
as 23–60 nucleotides may also be effective (Thomas et al., dsRNA interferes with PMMoV infection on hypersensitive
2001). In these experiments, the source of siRNAs would hosts and (ii) in cases where the virus is able to overcome
be the sum of the sequence-specific RNA degradation prod- the protection conferred by dsRNA in a systemic host, there
ucts from the recombinant virus and the transgene mRNA, is a delay in symptom expression of several weeks, suggest-
conceivably a higher dose than produced in our experimen- ing a reduced number of competent infection foci in the in-
tal system. Neither sense ssRNA nor antisense ssRNA de- oculated leaves. Furthermore, delivery of virus and dsRNA
rived from PMMoV or nonhomologous dsRNA (TEV-HC into the same cells seems necessary to trigger interference,
dsRNA) protected N. benthamiana plants against systemic because only a short interval (24 h) between sequential in-
infection. oculation of dsRNA and virus resulted in a complete sus-
Our experiments have also verified that the use of ceptibility of the plants to virus infection. This phenomenon
dsRNA molecules is a general strategy to promote inter- may be explained if we assume that during secondary inoc-
ference with the infection of plants by a variety of plant ulation the challenging virus enters the plant tissue through
viruses. N. tabacum plants inoculated with a mixture con- a number of cells that were not originally damaged during
taining infectious TEV genomic RNA transcripts and TEV the first inoculation round and that therefore do not contain
HC-Pro-derived dsRNA were protected against disease dsRNA molecules. This finding gives rise to one remark-
symptoms, and viral RNA did not accumulate in neither able feature of RNAi in our system, i.e. the failure of locally
inoculated nor upper leaves of these plants. Likewise, none introduced dsRNA to trigger a systemic antiviral response
of the N. benthamiana plants inoculated with a mixture of in plants. When dsRNA-treated N. benthamiana plants are
in vitro transcripts of AMV RNAs 1, 2 and 3 plus a dsRNA inoculated with virus in upper, non-treated leaves, they are
covering a 1124-nt region of AMV RNA 3 became infected, found to be susceptible to virus infection.
and AMV RNAs were not detectable in these plants. Experiments performed by others have revealed that
Because entrance of mechanically inoculated-dsRNA into microprojectile bombardment into leaf epidermal cells
the cell occurs upon abrasion of the epidermal tissue on the with beads coated with dsRNA molecules homologous to
leaf surface, accurate quantitation of dsRNA molecules en- endogenous genes triggered local RNAi, although its ef-
tering the cell is not possible, and consequently optimal con- ficiency appeared to decrease along with time (Schweizer
ditions for induction of an RNAi response against viruses et al., 2000). In plants, analysis of PTGS revealed that there
are difficult to establish. Using PMMoV, we could deter- are three separate components in the dynamics of RNA
mine that a 500-fold excess of dsRNA over viral RNA is silencing: initiation, propagation of a systemic silencing
the threshold required for interference with virus multipli- signal, and maintenance (Ruiz et al., 1998). Furthermore,
cation under the experimental conditions used. This appar- it has been reported that a nuclear component, transgene or
ently argues against an amplification step in the interference endogenous gene, homologous to the target RNA is required
process similar to that described in C. elegans, where a few for propagation and amplification of the systemic silencing
molecules of dsRNA are sufficient to trigger RNAi (Fire signal (Palauqui and Balzergue, 1999). In our scenario,
et al., 1998). Amplification of RNA silencing in cases in- dsRNA delivered by inoculation or expressed directly inside
volving transgenes likely reflects a transitive phenomenon plant cells by means of Agrobacterium tumefaciens (see
for which the initial pool of siRNAs generated from dsRNA below) would trigger the initiation step of PTGS targeting
inducer molecules is increased through the formation of sec- viral RNA for degradation. The maintenance step of PTGS
ondary siRNAs. It is suggested that primary siRNAs anneal would require a nuclear component, transgene or DNA se-
with target RNA to prime a PCR-like reaction catalyzed by quences, homologous to the target viral RNA. The absence
a host encoded-RNA dependent RNA polymerase (RdRp) of such a component in our nontransgenic system together
to form dsRNA corresponding to sequences located out- with the failure of input dsRNA to move between cells at
side the primary targeted region of the RNA. In contrast, detectable levels (unpublished data), presumably precludes
RdRp is dispensable in cases involving VIGS since the virus spreading of the sequence-specific signal involved in PTGS
encoded-RNA polymerase would produce dsRNA (Vaistij to other parts of the plant.
et al., 2002). Why amplification of RNA silencing does not
occur in our system remains unclear although it may be due
to the inability of the in vitro synthetized dsRNA to prime the 3. Interference with plant virus by transiently
production of new dsRNA from the target viral RNA. Alter- expressed hairpin RNA
natively, as mechanical inoculation of plant viruses is based
on limited damage to epidermal cells by abrasion, the dose Although the effects mediated by dsRNA in plant virus
effect described above probably reflects a minimum amount infection recapitulate many aspects of RNAi reported in
F. Tenllado et al. / Virus Research 102 (2004) 85–96 89

animals, a direct link between inhibition of plant virus in- infiltrated with hairpin RNA were resistant to a Potato virus
fection by exogenously supplied dsRNA and PTGS-related X (PVX)-based vector carrying sequences homologous to
processes was unequivocally provided by using A. tumefa- the PMMoV hairpin RNA. Second, siRNAs of two size
ciens as a transient expression system. Transient expression classes were found in extracts from hairpin RNA-infiltrated
of genes through infiltration of A. tumefaciens cultures into tissue. These siRNAs are consistently associated with an
leaf tissue (agroinfiltration) has been widely used as a means activated PTGS status (Hamilton and Baulcombe, 1999)
to deliver inducers, targets and suppressors of PTGS into and result from specific cleavage of dsRNA by an RNase
plants (Llave et al., 2000; Johansen and Carrington, 2001). III-like enzyme. Third, the interference with PMMoV infec-
For instance, expression of a given transgene in a plant can tion triggered by a hairpin RNA was partially overcome by
be suppressed by infiltrating the plant with a recombinant bi- transiently expressing a construct encoding the HC-Pro of
nary vector carrying the corresponding transgene sequence. Plum pox virus (PPV). In the last years, it has been demon-
In our laboratory, we have investigated the silencing poten- strated that HC-Pro of some potyviruses act as suppressors
tial of a construct designed to generate a hairpin RNA after of PTGS (Brigneti et al., 1998). Although not formally
agroinfiltration in the plant tissue to interfere with a rapidly demonstrated, we assumed that the HC-Pro of PPV could
replicating plant virus (Tenllado et al., 2003a). behave as a suppressor of PTGS, since it has been previ-
As mentioned in the introduction, virus resistance in ously shown to be involved in viral synergism with PVX in
plants has been reported to be induced at high efficiency N. benthamiana (Sáenz et al., 2002; P. González-Jara et al.,
by stably integrated transgenes expressing inverted-repeat unpublished data). This phenomenon is closely related to the
structures (Smith et al., 2000). We have demonstrated that silencing suppressor activity of HC-Pro in other potyviruses,
transient expression of a hairpin RNA can also effectively which supports its capability to suppress PTGS induced by
block multiplication and spread of a homologous highly transiently expressed hairpin RNA (Pruss et al., 1997).
replicating plant virus in nontransgenic plants. As a result, Although hairpin-dependent interference with viral in-
hairpin-dependent interference with virus infection by tran- fection has been described for PMMoV, it is reasonable
siently expressed constructs provides a potential tool to to expect that many plant RNA viruses will be inhibited
study the onset of PTGS in the context of a viral infection, by Agrobacterium-mediated transient expression of homol-
in contrast to dsRNA-transgenic plants which primarily al- ogous hairpin RNAs. In addition, this approach can give
low the study of PTGS maintenance. N. benthamiana plants valuable information about the step in the PTGS pathway
infiltrated with cultures of Agrobacterium carrying a hair- where viral proteins suppress RNA silencing. In this sense,
pin RNA derived from the 54-kDa region of PMMoV were available evidence indicates that transgene expression ob-
resistant to subsequent infection by PMMoV. In contrast, tained by infiltrating plant tissues with Agrobacterium is pri-
Agrobacterium-mediated expression of constructs contain- marily due to transcription of episomal T-DNA, rather than
ing duplicated (head-to-tail) PMMoV 54-kDa sequences the generation of stably integrated transgenes that are in-
in the sense or antisense orientations were ineffective as volved in PTGS maintenance (Sonti et al., 1995). In previous
initiators of RNAi, and disease symptoms were displayed studies, HC-Pro was shown to interfere with a maintenance
in upper leaves. At an interval of 3 or more days between step of the silenced state (Llave et al., 2000). Our coinfil-
agroinfiltration with PMMoV hairpin RNA and virus in- tration assays, in which expression of HC-Pro occurs at the
oculation plants were protected against virus infection, as same time as initiation of silencing by hairpin RNA, suggest
indicated by the absence of PMMoV RNA in both the that HC-Pro of potyviruses also interferes with the initia-
inoculated and the upper leaf tissue. tion of PTGS. Supporting this, it has been reported that the
The hairpin RNA-dependent interference with virus in- HC-Pro of TEV partially inhibits PTGS induced by a tran-
fection observed with transiently expressed hairpin RNA siently expressed dsRNA from a jellyfish green fluorescent
resembled interference with viral infection by direct deliv- protein at early stages of PTGS (Johansen and Carrington,
ery of dsRNA to leaf cells by mechanical inoculation. We 2001).
found several features associated with transient expression Some limiting aspects of dsRNA-mediated interference
of hairpin RNA that are hallmarks of PTGS. First, the in- with virus infection by mechanical inoculation of dsRNA
terference with virus infection exhibited by hairpin RNA to leaf cells were also described in Agrobacterium-mediated
was dependent on a high level of sequence identity between interference, i.e. the lack of a systemic antiviral response in
the hairpin RNA and the target RNA. We observed that upper parts of the plant triggered by locally induced PTGS.
plants transiently expressing PMMoV-derived hairpin RNA Delivery of virus and hairpin RNA into the same tissues
exhibited systemic infection when inoculated with an unre- seemed necessary to achieve protection against virus in-
lated virus such as AMV or Tobacco mosaic virus (TMV) fection, because Agrobacterium-infiltrated plants challenged
(73% identity). By contrast, agro-infiltrated plants remain with PMMoV in upper, noninfiltrated leaves became in-
uninfected when inoculated with a related strain of PMMoV fected. Similar arguments to those expressed for direct de-
(PMMoV-I), which shares 93% sequence identity with the livery of dsRNA by mechanical inoculation, together with
hairpin RNA. Further evidence for a homology-dependent the failure of Agrobacterium to efficiently move for long
type of resistance came from the observation that plants distances, can be made to explain this failure (see above).
90 F. Tenllado et al. / Virus Research 102 (2004) 85–96

RNA silencing represents an inherent defense mechanism 4. The potential of RNAi in plant virus disease control
whereby a sequence-specific response limits the extent of
virus infection (Voinnet, 2001). Unequivocal evidence exists The findings presented above suggest that exogenously
that RNA silencing can be transmitted throughout plants by supplied dsRNA could form the basis for the development
a mobile systemic signal (Mlotshwa et al., 2002). Viruses of a new biotechnological tool aimed at protecting crops
have moved forward with the development of efficient strate- against virus diseases, provided that some limitations of
gies to counteract or to escape RNA silencing. For instance, the current status of the approach could be overcome. Ob-
several plant viral silencing suppressors are known to af- viously, interference with virus infection mediated by in
fect systemic signaling of silencing, in agreement with the vitro-transcription products or via Agrobacterium-mediated
notion that systemic infection by a virus requires dimin- transient expression of dsRNA is limited to fundamental
ishing the antiviral silencing defense response in the plant studies. Thus, effective means of production and delivery of
(Voinnet et al., 2000). In this sense, the suppression of PTGS adequate interference products onto plant surfaces should
by HC-Pro of potyviruses could be potentially mediated, at be developed before a practical use of RNAi in plant pro-
least in part, by the activation of a pathway that transmits tection could be envisioned. As an attempt to achieve this,
the inhibition of silencing to other parts of the plant by a we established a simple, fast, safe and inexpensive proce-
mechanism similar to the trafficking of endogenous proteins dure to produce large amounts of dsRNA derived from viral
and their transcripts between cells and through the phloem sequences using an inducible expression system based on a
(Jorgensen et al., 1998). The combination of both an in- genetically modified E. coli strain (Tenllado et al., 2003b).
ducer (hairpin RNA) and a suppressor (HC-Pro) of PTGS In C. elegans, the E. coli strain HT115(DE3) has been
transiently expressed by Agrobacterium allowed us to de- used to produce large amounts of dsRNA with the purpose
vise a set of experiments involving the sequential infiltra- of triggering RNAi of endogenous genes in the nematode
tion of plants in different leaves with single Agrobacterium (Timmons and Fire, 1998; Timmons et al., 2001). This strain
cultures carrying either the PPV HC-Pro or the PMMoV is deficient for RNase III, an enzyme that normally de-
hairpin RNA. HC-Pro transiently expressed in lower leaves grades the majority of dsRNAs in the bacterial cell, thereby
failed to suppress the inhibitory effect on PMMoV replica- improving the accumulation of extended dsRNA duplexes
tion triggered by infiltration of hairpin RNA in upper leaves compared to common strains of E. coli. We adopted this
(Fig. 2). Thus, no evidence for a mobile silencing suppres- promising strain, which had also been modified to express
sion signal induced by HC-Pro of PPV was observed in our T7 RNA polymerase from an IPTG-inducible promoter, for
system. However, this result does not completely rule out the preparation of massive amounts of viral dsRNA in vivo.
the involvement of a mobile silencing suppression signal in Upon induction, HT115(DE3) transformed with a construct
natural potyvirus infection. For instance, PTGS suppression encoding a hairpin RNA derived from the 54-kDa region of
signaling to distal parts of the plant may require the accumu- PMMoV accumulated substantial levels of PMMoV-derived
lation of a threshold level of HC-Pro on the onset of a virus dsRNA. More importantly, the 54-kDa dsRNA produced
infection, or perhaps, any other potyviral proteins. The P1 in bacteria was capable of interfering with PMMoV infec-
proteinase of TEV has been shown to enhance suppression tion. In fact, N. benthamiana plants inoculated with mix-
of VIGS by HC-Pro (Anandalakshmi et al., 1998). tures of PMMoV and phenol-extracted nucleic acid extracts

Fig. 2. PPV HC-Pro expressed in lower leaves does not suppress hairpin-mediated interference with PMMoV infection in upper leaves. (A) Schematic
representation of the experimental procedure. Two lower, opposite leaves of N. benthamiana plants were first infiltrated with Agrobacterium containing
either vector alone or HC-Pro. After 7 days, upper leaves of these plants were infiltrated in all the combinations with Agrobacterium containing either
vector alone or hairpin RNA. Four days later, PMMoV was inoculated on the uppermost, infiltrated leaves. (B) Northern blot analysis of total RNA
extracted from the inoculated leaves of plants inoculated with PMMoV at 15 days post-inoculation.
F. Tenllado et al. / Virus Research 102 (2004) 85–96 91

prepared from HT115(DE3) expressing dsRNA were free of Further experiments in our laboratory have proved RNAi
symptoms during the completion of their life cycles. Fur- technology to be suitable to control infection by a variety of
thermore, viral RNA levels were below the limit of detec- plant viruses. PPV is the causal agent of sharka or plum pox,
tion in both the inoculated and the upper leaf tissue. In the most serious disease on woody trees of the genus Prunus.
contrast, all the plants that were inoculated with PMMoV Most cultivated Prunus species are highly susceptible, and
plus extracts derived from HT115(DE3) harboring the empty conventional breeding has not produced highly resistant
cloning vector displayed high levels of viral RNA. In agree- and commercially acceptable varieties (Ravelonandro et al.,
ment with previous results that confirmed the remarkable 2000). This situation encouraged us to apply RNAi against
sequence-specificity of the RNA silencing process, coinoc- PPV in an experimental host, N. benthamiana, with the per-
ulation of nucleic acid extracts containing PMMoV dsRNA spective of providing the basis for the potential control of
together with a nonhomologous virus, TMV, did not affect PPV in its natural host. As predicted, a high proportion of
virus infection in plants. Thus, interference with virus in- plants inoculated with combinations of PPV plus homolo-
fection exhibited by nucleic acid extracts derived from bac- gous dsRNA-expressing preparations showed no symptoms
teria expressing dsRNA was dependent on a high level of of virus disease. Furthermore, no traces of PPV replication
sequence identity with the target viral RNA. were detected by several diagnostic tests (ELISA, RT-PCR)
Large-scale production of dsRNA required for the imple- in the upper leaves of these plants. Thus, our work has
mentation of RNAi in plant protection depends on a sim- demonstrated that easy-to-obtain, crude extracts of bacte-
ple and quick procedure to obtain dsRNA-related interfering rially expressed dsRNAs are highly effective in protecting
products from bacteria, avoiding the use of time-consuming plants against infection by PMMoV and PPV, pathogens
protocols to purify nucleic acids. This prompted us to an- that represent two of the main widespread groups of plant
alyze the interfering activity on virus infection of bacterial viruses in nature.
crude preparations obtained by lysing cell pellets with a From a practical standpoint, we developed a simple
French Press. We found that viral accumulation was com- spray technique for the delivery of dsRNA-containing
pletely inhibited in N. benthamiana leaves that had been crude preparations onto the surface of plant leaves. We
inoculated with a mixture of a bacterial lysate expressing reported that virus infectivity was specifically abolished
PMMoV-derived dsRNA and PMMoV particles. In con- when plants were sprayed with bacterial lysates several
trast, plants coinoculated with virus and bacterial lysate days before mechanical inoculation with either PMMoV
preparations expressing the empty vector or a nonhomol- or PPV in the same leaves. Analysis of the upper leaves
ogous dsRNA (see below) accumulated PMMoV RNA at from dsRNA-treated plants confirmed the absence of either
high levels (Fig. 3). This precluded any effect concerning PMMoV or PPV accumulation. The rationale of this ob-
non-specific toxicity of the bacterial extract on the inter- servation is that dsRNA molecules, which remain on the
ference with virus infection observed by dsRNA-expressing leaf surface after being sprayed, enter the cell along with
preparations. viral particles due to mechanical damage in the epidermal

Fig. 3. Interference with PMMoV infection by bacterial crude preparations expressing PMMoV dsRNA. (A) Northern blot analysis of total RNA
extracted from inoculated leaves of N. benthamiana at 7 days post-inoculation. Plants were inoculated with mixtures of PMMoV plus either a series of
dilutions (1/2–1/20) of the PMMoV dsRNA preparation, or the PPV dsRNA preparation diluted 1/2, as indicated. (B) Response of N. benthamiana to a
combination of PMMoV plus French Press preparations derived from either PMMoV dsRNA (left), PPV dsRNA (middle) or empty vector (right). Plants
displaying disease symptoms (one-half-diluted, PPV dsRNA and vector preparations) or showing protection to virus infection (1/10-diluted PMMoV
dsRNA preparation) were photographed at 30 days post-inoculation.
92 F. Tenllado et al. / Virus Research 102 (2004) 85–96

cells. As a result, the dsRNA is converted into siRNAs that virus, poliovirus, rotavirus, and respiratory syncytial virus
guide sequence-specific cleavage of the homologous viral in human cell lines using siRNAs (Bitko and Barik, 2001;
RNA. The presence of interfering products on leaf surfaces Gitlin et al., 2002; Jacque et al., 2002; Novina et al., 2002).
for several days is not surprising as a previous analysis of Our results summarized here contribute to establish the ba-
the stability of in vitro-synthesized dsRNA molecules after sis of a new application of RNAi for the control of diseases
delivery into leaves, had shown most of the input dsRNA caused by plant viruses. This approach differs from strate-
to be relatively stable and to persist in the leaves for sev- gies based on transgenic plants capable of expressing dsRNA
eral days after inoculation, even after exuberant washing that are immune to viruses (Smith et al., 2000; Kalantidis
(Tenllado and Dı́az-Ruı́z, 2001). These encouraging re- et al., 2002), but still relies on PTGS as a means to achieve
sults lead to the concept of spraying crude preparations of PDR. We established a simple and inexpensive procedure
dsRNA-expressing E. coli HT115(DE3) as a highly con- to produce large amounts of dsRNA derived from viral se-
venient method for delivery of RNAi inducers in plants quences in bacteria, with the view of providing the starting
and further contribute to making RNAi technology widely point for the industrial production of these molecules. When
available and applicable for deployment in the field. applied on plants, the dsRNAs caused specific degradation
of the homologous viral RNA with the resultant protection
against virus infections. The infectivity of virtually any RNA
5. Perspectives of the use of RNAi for crop protection virus in plants can now be inhibited with the correspond-
ing dsRNA, without detrimental effect on plant surfaces. We
Until recently, the only available direct strategy, beyond propose that exogenously supplied dsRNA would mimic the
classical plant breeding, for the potential control of virus dsRNA intermediate involved in viral replication, triggering
diseases in crops was the use of virus-resistant transgenic the initiation step of PTGS that leads to the production of
plants. The demonstration in 1986 that the expression of siRNAs which are incorporated into the nuclease complex
a viral coat protein gene in a transgenic plant could con- (RISC) responsible for the degradation of the target RNA
fer resistance to the virus represented a major breakthrough (Hammond et al., 2000). Thus, the invading virus contain-
(Powell-Abel et al., 1986). It opened the prospect of a virtu- ing sequences homologous to the dsRNA is recognized and
ally unlimited source of virus resistance genes that could be degraded by this defense mechanism.
used in crops in which sources of natural resistance genes As mentioned above, numerous studies have established
were unavailable or inadequate. However, over the past 10 that synthetic siRNAs used to target viral genes can con-
years, questions concerning the potential ecological impact fer resistance to several cytoplasmically replicating animal
of transgenic plants have been raised (Tepfer, 2002). The viruses. To date, siRNAs have only been occasionally used
areas of potential risk include complementation, heterolo- as inducers of RNAi in plants (Klahre et al., 2002), proba-
gous encapsidation, synergy, and genetic flow between or- bly due to the likelihood of triggering silencing using long
ganisms. In view of these concerns, alternative strategies dsRNA molecules. Moreover, delivery of a unique siRNA
aimed at protecting crops against virus diseases reducing or species into a cell may compromise the efficiency of RNAi,
eliminating certain risks possibly associated with transgenic since it may not be sufficient to activate a potent silenc-
expression of viral sequences should be evaluated. ing response (Paddison et al., 2002). At present, little is
In just 5 years, molecular biologists have uncovered a pre- known about the structural and sequence requirements that
viously unsuspected biological mechanism which is used by govern target recognition and subsequent processing by the
nature to silence genes in many different settings. Besides siRNA-containing RISC complex. For instance, it is not pos-
having become a prominent tool in basic research, RNAi sible to anticipate the most optimal viral RNA region for
technology provides a significant new strategy to combat vi- siRNA targeting. Also, the viral genome might be inacces-
ral diseases in many organisms, from plants to mammals. sible to some siRNAs, perhaps due to being enclosed in a
It offers a combination of extreme specificity and the ex- compacted protein-RNA complex or to possessing a partic-
ploitation of an underlying cellular pathway that has seem- ular RNA structure (Bitko and Barik, 2001). Because plant
ingly evolved naturally for this purpose. A common feature cells lack non-specific inhibitory effects activated by dsRNA
of silencing-related processes in different organisms is the in mammals, limitations caused by siRNAs can be circum-
involvement of dsRNA as an initiator molecule. We have vented by the use of long dsRNA. Processing of long dsR-
seen that during cytoplasmic replication of RNA viruses, the NAs by the PTGS machinery present in plant cells should
replicative forms and replicative intermediates may repre- generate a mixture of siRNAs corresponding to different re-
sent the pool of dsRNAs that trigger induction of the PTGS gions of the precursor dsRNA. Sequence-specific degrada-
antiviral defense mechanism in plants, at least under certain tion of viral RNA would occur as a result of multiple siRNAs
circumstances (Dalmay et al., 2000; Voinnet et al., 2000). functioning cooperatively at different positions of the target
At present, it is not clear whether RNAi is part of a vi- RNA. This most likely enhances the robustness of antiviral
ral surveillance system in animal cells. However, antiviral RNAi response in plants.
RNAi technology has been recently applied for the con- We anticipate that inducible expression of dsRNA in bac-
trol of human immunodeficiency virus type 1, hepatitis C teria could easily be scaled-up as a rapid and cost-effective
F. Tenllado et al. / Virus Research 102 (2004) 85–96 93

expression system for a large variety of viral dsRNAs. In in distal tissues does not occur. As a result, protection would
our experience, average yield of dsRNA produced in bacte- be feasible only if adequate coverage of the whole plant
rial cells was 4 ␮g/ml of culture. Large fermenters of up to with interfering dsRNA is guaranteed. Alternatively, the use
100,000 l, such as those used for the commercial production of coadyuvants in the dsRNA-based formulations could fa-
of entomopathogenic nematodes for the control of insect cilitate the spread of interfering products to distant parts of
pests (Georgis and Manwelier, 1994), would potentially the plant triggering a systemic protective effect against virus
meet the enormous demand for the use of these interfering infection. Third, dsRNA retains its activity as an inducer of
products in agriculture. Moreover, bacterial cell cultures RNAi against viruses only for a transitory period of time.
provide a practicable and reproducible approach for mass This fact creates a number of inconveniences since frequent
production of dsRNA. They offer the possibility to be easily protective treatment may be impracticable or not be appli-
pelleted, stored and distributed, without losing their inter- cable at the time required in open-field crops. Alternatively,
fering properties with plant virus infection. Large-scale pro- application of formulations based on viral dsRNA made
duction of dsRNA for its use in agriculture can be achieved at appropriate times during the growing season, coincident
only through simple and inexpensive downstream process- with vector epidemics, should have the potential of activat-
ing steps. Lysing cells with a French Press or any other ing PTGS mechanisms of virus resistance in crops grown
mechanical means is a rapid and efficient procedure that under plastic or glass. Fourth, RNAi mediated by direct
avoids the use of chemicals and time-consuming protocols delivery of dsRNA onto plants is intended to be a practical
to purify nucleic acids from bacteria. The demonstration tool for virus control on a commercial scale. Unfortunately,
under experimental conditions that dsRNA also works in at present, large-scale applications have not been accom-
interfering with virus infection when bacterial crude prepa- plished yet so it remains unknown whether immunization
rations are sprayed onto plant surfaces, further contributes would be successful. In addition, the protective effect has so
to making this technology widely available and applicable far been tested in model plants but not in crop species. Cou-
for deployment in the field. pled with these current problems, costs regarding long-term
Although this approach is very exciting, important prob- applications in crops should be evaluated before directly de-
lems must be solved before this RNAi-based technology can livering dsRNA as a means to initiate RNAi is implemented
be used as a tool for virus control in the field on a commercial in a therapeutic setting. Another determining consideration
scale. First, plants that are protected against mechanically in- to keep in mind is that spraying dsRNA-based formula-
oculated target viruses may not be protected when the virus tions could be more competitive with the costs of chemical
is inoculated by natural vectors. This is especially problem- insecticide programmes currently used to control insect
atic since the vast majority of plant viruses are transmitted vectors.
from plant to plant in nature by invertebrate vectors which, At present, provided that an efficient transformation tech-
in many cases, release the virus particles directly inside the nique is available, RNAi based on stable transformation of
plant cell (López-Moya, 2002). This probably precludes host plants with viral sequences in the form of constructs
the possibility that the interfering dsRNA sprayed onto leaf directing dsRNA formation, is likely the most efficient
surfaces enters the cell together with the virus. Since most method to confer resistance against pathogenic viruses in
ssRNA plant viruses, including those in the economically crops. Biotechnological utilization of RNAi-based engi-
important group of potyviruses, are transmitted by aphids neered resistance is appealing for biosafety reasons as well.
in nature, we are especially interested in using RNAi tech- RNA silencing-based resistance does not involve the trans-
nology to extend the resistance against viral inoculation by genic production of functional viral genes or proteins, nor
aphids. Studies have been initiated trying to facilitate the does it lead to the presence of transgene RNA thus mini-
penetration of bacterially expressed dsRNA into plant tis- mizing potential ecological and environmental risks (Tepfer,
sues other than through epidermal cells. Finding appropriate 2002). Since little or no transgene mRNA is accumulated in
coadyuvants would provide a starting line for the design plant cells, there is essentially no template for events such
of new formulations based on dsRNA aimed at protecting as complementation, heterologous encapsidation, synergy,
plants against this natural means of infection. Successful and recombination. However, despite the enormous benefit
interference with virus infection by RNAi would reduce the for agriculture, questions concerning the potential ecologi-
use of pesticides in agriculture thus minimizing potential cal impact of virus-resistant transgenic plants have signifi-
ecological and environmental risks. At present, the most cantly limited their use. For instance, reversal of PTGS by
widely used strategy to control virus diseases is by chemical virus-encoded suppressors would severely limit the biotech-
control of viral vectors. Alternatives to chemical pesticides nological applications of RNA silencing-based transgenic
are desirable for several reasons, such as an increased aware- resistance. Infection of a PTGS-based virus-resistant trans-
ness of the potentially harmful effects of chemical residues genic plant by a nonhomologous virus with silencing sup-
in food and the environment, and the increasing resistance of pression capabilities, may reverse the silencing status of the
insects to insecticides. Second, dsRNA induces RNA silenc- transgene resulting in high expression of the transgene and
ing in the inoculated tissue but not in the upper parts of the loss of transgene-homologous virus resistance (Savenkov
plants, i.e. intercellular signaling to trigger RNA silencing and Valkonen, 2002). This creates a situation in which
94 F. Tenllado et al. / Virus Research 102 (2004) 85–96

recombination between transgene mRNA and the RNA of ered. The successful exploitation of bacterially expressed,
the infecting virus could potentially occur. exogenously supplied dsRNA for the control of plant virus
RNAi technology based on exogenously supplied dsRNA diseases by RNAi seems to be promising.
has several apparent advantages compared to the require-
ments for regenerating transgenic plants capable to interfere
with plant virus infections. The experimental procedure Acknowledgements
does not require sophisticated equipment and is inexpen-
We wish to thank Natalia Wright for critical reading of
sive, compared to labor- and cost-intensive production of
the manuscript. F.T has been the recipient of a “Ramón y
transgenic plants encoding virus genes. A further advantage
Cajal” contract from MCyT (Spain) and he is currently Se-
of the RNAi technology described here is the facility to de-
nior Investigator of CSIC. C.L. is the recipient of an “I3P”
liver several dsRNAs targeting different viruses in a single
contract from CSIC (Spain). J.R.D.-R. is Research Profes-
formulation, so that multiple resistance traits can be gener-
sor Staff Scientist of CSIC. The research done in the lab-
ated. In stable transgenic plants such manipulation would
oratory has been founded with grants BIO2000-0914 and
require sequential transformations or crosses between trans-
BIO2003-03428 from CICYT-MCyT and 07M/0123/2000
genic plants that could take prolonged periods of time.
and 07G/0043/2003 from Comunidad de Madrid (Spain).
Although PTGS triggered by exogenously supplied dsRNA
could also be broken down by suppressor viruses, dsRNA
molecules can be designed to lack transcriptional regulatory References
regions, such as the CaMV 35S promoter commonly used
in transgenic plants. This would probably impair overaccu- Anandalakshmi, R., Pruss, G.J., Ge, X., Marathe, R., Mallory, A.C.,
mulation of interfering products on dsRNA-treated plants, Smith, T.H., Vance, V.B., 1998. A viral suppressor of gene silencing
reducing the chance of RNA recombination. in plants. Proc. Natl. Acad. Sci. U.S.A. 95, 13079–13084.
Baulcombe, D.C., 1999. Viruses and gene silencing in plants. Arch. Virol.
Suppl. 15, 189–201.
Bernstein, E., Caudy, A.A., Hammond, S.M., Hannon, G.J., 2001. Role
6. Conclusions for a bidentate ribonuclease in the initiation step of RNA interference.
Nature 409, 363–366.
Efficient control of plant viruses affecting economi- Bitko, V., Barik, S., 2001. Phenotypic silencing of cytoplasmic genes
using sequence-specific double-stranded short interfering RNA and its
cally important crop species represents one of the major
application in the reverse genetics of wild type negative-strand RNA
challenges in the development of effective and sustainable viruses. BMC Biotechnol. 1, 34–46.
measures of plant health management. As a necessary com- Brigneti, G., Voinnet, O., Li, W.X., Ji, L.H., Ding, S.W., Baulcombe, D.C.,
plement to traditional methods of virus control, emerging 1998. Viral pathogenicity determinants are suppressors of transgene
technologies should be based on a better understanding of silencing in Nicotiana benthamiana. EMBO J. 17, 6739–6746.
Cayley, P.J., White, R.F., Antoniw, J.F., Walesby, N.J., Kerr, I.M., 1982.
the natural defense mechanisms of the host plants and the
Distribution of the ppp(A2’p)nA-binding protein and interferon-related
counteracting strategies displayed by the viruses. RNA si- enzymes in animals, plants, and lower organisms. Biochem. Biophys.
lencing and other related pathways play an important role Res. Commun. 108, 1243–1250.
in the regulation of basic cellular processes such as gene ex- Dalmay, T., Hamilton, A., Rudd, S., Angell, S., Baulcombe, D.C., 2000.
pression and heterochromatin formation. In addition, RNA An RNA-dependent RNA polymerase gene in Arabidopsis is required
for posttranscriptional gene silencing mediated by a transgene but not
silencing represents an adaptive antiviral defense mecha-
by a virus. Cell 101, 543–553.
nism in plants. RNA silencing is activated in the presence of Denli, A.M., Hannon, G.J., 2003. RNAi: an ever-growing puzzle. Trends
either dsRNA molecules or siRNAs leading to degradation Biochem. Sci. 28, 196–201.
of homologous RNAs in a sequence-specific manner. The Dı́az-Ruı́z, J.R., Kaper, J.M., 1978. Isolation of viral double-stranded
discovery that inducer molecules can trigger inactivation of RNAs using a LiCl fractionation procedure. Prep. Biochem. 8, 1–17.
Djikeng, A., Shi, H., Tschudl, C., Ullu, E., 2001. RNA interference in
target RNAs when directed against virus genomes opens
Trypanosoma brucei: cloning of small interfering RNAs provides evi-
the way to investigate the development of methodologies dence for retroposon-derived 24–26-nucleotide RNAs. RNA 7, 1522–
to deliver RNA silencing inducers into the plant tissue with 1530.
the purpose of interfering with a specific step of virus repli- Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl,
cation and/or movement. In this article, we have discussed T., 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference
in cultured mammalian cells. Nature 411, 494–498.
the concept of RNA silencing as an elegant natural antiviral
Fagard, M., Boutet, S., Morel, J.-B., Bellini, C., Vaucheret, H., 2000.
mechanism in plants and dissected the prospective of RNAi AGO-1, QDE-2, and RDE-1 are related proteins required for
technology as a tool for control of virus diseases in crop post-transcriptional gene silencing in plants, quelling in fungi, and
plants. RNA interference in animals. Proc. Natl. Acad. Sci. U.S.A. 97, 11650–
Our long-term goal is the development of RNAi as a 11654.
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello,
biotechnological approach to control plant virus diseases
C.C., 1998. Potent and specific genetic interference by double-stranded
in crops in an effective and environmentally safety manner. RNA in Caenorhabditis elegans. Nature 391, 806–811.
Although much progress has been made recently in our Georgis, R., Manwelier, S.A., 1994. Entomopathogenic nematodes: a
understanding of PTGS, much still remains to be discov- developing biological control technology. Agric. Zool. Rev. 6, 63–94.
F. Tenllado et al. / Virus Research 102 (2004) 85–96 95

Gil, J., Esteban, M., 2000. Induction of apoptosis by the dsRNA-dependent Palauqui, J.C., Balzergue, S., 1999. Activation of systemic acquired si-
protein kinase (PKR): mechanism of action. Apoptosis 5, 107–114. lencing by localised introduction of DNA. Curr. Biol. 9, 59–66.
Gitlin, L., Karelsky, S., Andino, R., 2002. Short interfering RNA confers Pasquinelli, A.E., Ruvkun, G., 2002. Control of developmental timing by
intracellular antiviral immunity in human cells. Nature 418, 430–434. microRNAs and their targets. Annu. Rev. Cell Dev. Biol. 18, 495–513.
Goldbach, R., Bucher, E., Prins, M., 2003. Resistance mechanisms to Peña, L., Trad, J., Dı́az-Ruı́z, J.R., McGarvey, P.B., Kaper, J.M., 1994.
plant viruses: an overview. Virus Res. 92, 207–212. Cucumber mosaic virus protection in transgenic tobacco plants ex-
Hamilton, A., Voinnet, O., Chappell, L., Baulcombe, D.C., 2002. Two pressing monomeric, dimeric or partial sequences of a bening satellite
classes of short interfering RNA in RNA silencing. EMBO J. 21, RNA. Plant Sci. 100, 71–81.
4671–4679. Powell-Abel, P., Nelson, R.S., De, B., Hoffmann, N., Rogers, S.G., Fraley,
Hamilton, A.J., Baulcombe, D.C., 1999. A species of small antisense RNA R.T., Beachy, R.N., 1986. Delay of disease development in transgenic
in posttranscriptional gene silencing in plants. Science 286, 950–952. plants that express the tobacco mosaic virus coat protein gene. Science
Hamilton, A.J., Brown, S., Yuanhai, H., Ishizuka, M., Lowe, A., Solis, 232, 738–743.
A.G.A., Grierson, D., 1998. A transgene with repeat DNA causes Pruss, G., Ge, X., Shi, X.M., Carrington, G.C., Vance, V.B., 1997.
high frequency, post-transcriptional suppression of ACC-oxidase gene Plant viral synergism: the potyviral genome encodes a broad-range
expression in tomato. Plant J. 15, 737–746. pathogenicity enhancer that transactivates replication of heterologous
Hammond, S., Bernstein, E., Beach, D., Hannon, G., 2000. An viruses. Plant Cell 9, 1–11.
RNA-directed nuclease mediates post-transcriptional gene silencing in Ratcliff, F., MacFarlane, S., Baulcombe, D.C., 1999. Gene silencing with-
Drosophila cells. Nature 404, 293–296. out DNA: RNA-mediated cross protection between viruses. Plant Cell
Hammond, S.M., Caudy, A.A., Hannon, G.J., 2001. Post-transcriptional 11, 1207–1215.
gene silencing by double-stranded RNA. Nat. Rev. Genet. 2, 110–119. Ravelonandro, M., Scorza, R., Callahan, A., Levy, L., Jacquet, C., Mon-
Hannon, G.J., 2002. RNA interference. Nature 418, 244–251. sion, M., Damsteegt, V., 2000. The use of transgenic fruit trees as a
Hutvágner, G., Zamore, P.D., 2002. A microRNA in a multiple-turnover resistance strategy for virus epidemics: the plum pox (sharka) model.
RNAi enzyme comple. Science 297, 2056–2060. Virus Res. 71, 63–69.
Jacque, J.M., Triques, K., Stevenson, M., 2002. Modulation of HIV-1 Romano, N., Macino, G., 1992. Quelling: transient inactivation of gene
replication by RNA interference. Nature 418, 435–438. expression in Neurospora crassa by transformation with homologous
Johansen, L.K., Carrington, J.C., 2001. Silencing on the spot. Induction sequences. Mol. Microbiol. 22, 3343–3353.
and suppression of RNA silencing in the Agrobacterium-mediated Ruiz, M.T., Voinnet, O., Baulcombe, D.C., 1998. Initiation and mainte-
transient expression system. Plant Physiol. 126, 930–938. nance of virus-induced gene silencing. Plant Cell 10, 937–946.
Jorgensen, R.A., Atkinson, R.G., Forster, R.L., Lucas, W.J., 1998. An Sáenz, P., Salvador, B., Simón-Mateo, C., Kasschau, K.D., Carrington,
RNA-based information superhighway in plants. Science 279, 1486– J.C., Garcı́a, J.A., 2002. Host-specific involvement of the HC protein
1487. in the long-distance movement of potyviruses. J. Virol. 76, 1922–1930.
Kalantidis, K., Psaradakis, S., Tabler, M., Tsagris, M., 2002. The occur- Sanford, J.C., Johnston, S.A., 1985. The concept of parasite-derived re-
rence of CMV-specific short RNAs in transgenic tobacco expressing sistance. Deriving resistance genes from the parasite’s own genome.
virus-derived double-stranded RNA is indicative of resistance to the J. Theor. Biol. 113, 395–405.
virus. Mol. Plant-Microbe Interact. 15, 826–833. Savenkov, E.I., Valkonen, J.P., 2002. Silencing of a viral RNA silencing
Ketting, R.F., Haverkamp, T.H., van Luenen, H.G., Plasterk, R.H., 1999. suppressor in transgenic plants. J. Gen. Virol. 83, 2325–2335.
Mut-9 of C. elegans, required for transposon silencing and RNA Schweizer, P., Pokorny, J., Schulze-Lefert, P., Dudler, R., 2000.
interference, is a homolog of Werner syndrome helicase and RNaseD. Double-stranded RNA interferes with gene function at the single-cell
Cell 99, 133–141. level in cereals. Plant J. 24, 895–903.
Klahre, U., Crete, P., Leuenberger, S.A., Iglesias, V.A., Meins, F., 2002. Smith, N., Singh, S., Wang, M.B., Stoutjesdijk, P., Green, A., Waterhouse,
High molecular weight RNAs and small interfering RNAs induce P.M., 2000. Total silencing by intron-spliced hairpin RNAs. Nature
systemic posttranscriptional gene silencing in plants. Proc. Natl. Acad. 407, 319–320.
Sci. U.S.A. 99, 11981–11986. Sonti, R.V., Chiurazzi, M., Wong, D., Davies, C.S., Harlow, G.R., Mount,
Li, H., Li, W.X., Ding, S.W., 2002. Induction of suppression of RNA D.W., Signer, E.R., 1995. Arabidopsis mutants deficient in T-DNA
silencing by an animal virus. Science 296, 1319–1321. integration. Proc. Natl. Acad. Sci. U.S.A. 92, 11786–11790.
López-Moya, J.J., 2002. Genes involved in insect-mediated transmission Tabara, H., Grishok, A., Mello, C.C., 1998. RNAi in C. elegans: soaking
of plant viruses. In: Khan, J.A., Dijkstra, J. (Eds.), Plant Viruses as in the genome sequence. Science 282, 430–431.
Molecular Pathogens. The Haworth Press Inc., London, pp. 31–61. Tang, G., Reinhart, B.J., Bartel, D.P., Zamore, P.D., 2003. A biochemical
Llave, C., Kasschau, K.D., Carrington, J.C., 2000. Virus-encoded suppres- framework for RNA silencing in plants. Genes Dev. 17, 49–63.
sor of posttranscriptional gene silencing targets a maintenance step in Tavernarakis, N., Wang, S.L., Dorovkov, M., Ryazanov, A., Driscoll, M.,
the silencing pathway. Proc. Natl. Acad. Sci. U.S.A. 97, 13401–13406. 2000. Heritable and inducible genetic interference by double-stranded
Llave, C., Xie, Z., Kasschau, K.D., Carrington, J.C., 2002. Cleavage RNA encoded by transgenes. Nat. Genet. 24, 180–183.
of Scarecrow-like mRNA targets directed by a class of Arabidopsis Tenllado, F., Barajas, D., Vargas, M., Atencio, F.A., González-Jara, P.,
miRNA. Science 297, 2053–2056. Dı́az-Ruı́z, J.R., 2003a. Transient expression of homologous hairpin
Mlotshwa, S., Voinnet, O., Mette, M.F., Matzke, M., Vaucheret, H., Ding, RNA causes interference with plant virus infection and is overcome
S.W., Pruss, G., Vance, V.B., 2002. RNA silencing and the mobile by a virus encoded suppressor of gene silencing. Mol. Plant-Microbe
silencing signal. Plant Cell 14 (Suppl.), S289–S301. Interact. 16, 149–158.
Napoli, C., Lemieux, C., Jorgensen, R., 1990. Introduction of a Tenllado, F., Dı́az-Ruı́z, J.R., 1999. Complete resistance to pepper mild
chimeric chalcone synthase gene into Petunia results in reversible mottle tobamovirus mediated by viral replicase sequences partially
co-suppression of homologous genes in trans. Plant Cell 2, 279–289. depends on transgene homozygosity and is based on a gene silencing
Novina, C.D., Murray, M.F., Dykxhoorn, D.M., Beresford, P.J., Riess, J., mechanism. Transgenic Res. 8, 83–93.
Lee, S.K., Collman, R.G., Lieberman, J., Shankar, P., Sharp, P.A., Tenllado, F., Dı́az-Ruı́z, J.R., 2001. Double-stranded RNA-mediated in-
2002. siRNA-directed inhibition of HIV-1 infection. Nat. Med. 8, terference with plant virus infection. J. Virol. 75, 12288–12297.
681–686. Tenllado, F., Garcı́a-Luque, I., Serra, M.T., Dı́az-Ruı́z, J.R., 1995. Nico-
Paddison, P.J., Caudy, A.A., Bernstein, E., Hannon, G.J., Conklin, D.S., tiana benthamiana plants transformed with the 54-kDa region of the
2002. Short hairpin RNAs (shRNAs) induce sequence-specific silenc- pepper mild mottle tobamovirus replicase gene exhibit two types of
ing in mammalian cells. Genes Dev. 16, 948–958. resistance responses against viral infection. Virology 211, 170–183.
96 F. Tenllado et al. / Virus Research 102 (2004) 85–96

Tenllado, F., Garcı́a-Luque, I., Serra, M.T., Dı́az-Ruı́z, J.R., 1996. Re- of the target gene and a putative RNA-dependent RNA polymerase.
sistance to pepper mild mottle tobamovirus conferred by the 54-kDa Plant Cell 14, 857–867.
gene sequence in transgenic plants does not require expression of the Vazquez Rovere, C., del Vas, M., Hopp, H.E., 2002. RNA-mediated virus
wild-type 54-kDa protein. Virology 219, 330–335. resistance. Curr. Opin. Biotechnol. 13, 167–172.
Tenllado, F., Martinez-Garcia, B., Vargas, M., Dı́az-Ruı́z, J.R., 2003b. Voinnet, O., 2001. RNA silencing as a plant immune system against
Crude extracts of bacterially expressed dsRNA can be used to protect viruses. Trends Genet. 17, 449–459.
plants against virus infections. BMC Biotechnol. 3, 3. Voinnet, O., Lederer, C., Baulcombe, D.C., 2000. A viral movement
Tepfer, M., 2002. Risk assessment of virus-resistant transgenic plants. protein prevents spread of the gene silencing signal in Nicotiana
Annu. Rev. Phytopathol. 40, 467–491. benthamiana. Cell 103, 157–167.
Thomas, C.L., Jones, L., Baulcombe, D.C., Maule, A.J., 2001. Size Volpe, T.A., Kidner, C., Hall, I.M., Teng, G., Grewal, S.L., Martienssen,
constraints for targeting post-transcriptional gene silencing and for R.A., 2002. Regulation of heterochromatic silencing and histone H3
RNA-directed methylation in Nicotiana benthamiana using a potato lysine-9 methylation by RNAi. Science 297, 1833–1837.
virus X vector. Plant J. 25, 417–425. Wang, M.B., Abbott, D.C., Waterhouse, P.M., 2000. A single copy
Tijsterman, M., Ketting, R.F., Plasterk, R.H., 2002. The genetic of RNA of a virus-derived transgene encoding hairpin RNA gives immu-
silencing. Annu. Rev. Genet. 36, 489–519. nity to barley yellow dwarf virus. Mol. Plant Pathol. 1, 347–
Timmons, L., Court, D.L., Fire, A., 2001. Ingestion of bacterially ex- 356.
pressed dsRNAs can produce specific and potent genetic interference Waterhouse, P.M., Gramham, M., Wang, M.B., 1998. Virus resistance and
in Caenorhabditis elegans. Gene 263, 103–112. gene silencing in plants can be induced by simultaneous expression of
Timmons, L., Fire, A., 1998. Specific interference by ingested dsRNA. sense and antisense RNA. Proc. Natl. Acad. Sci. U.S.A. 95, 13959–
Nature 395, 854. 13964.
Vaistij, F.E., Jones, L., Baulcombe, D.C., 2002. Spreading of RNA tar- Waterhouse, P.M., Wang, M.B., Lough, T., 2001. Gene silencing as an
geting and DNA methylation in RNA silencing requires transcription adaptive defense against viruses. Nature 411, 834–842.
Virus Research 102 (2004) 97–108

Plant viral suppressors of RNA silencing


Braden M. Roth, Gail J. Pruss, Vicki B. Vance∗
Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA

Abstract

RNA silencing is an ancient eukaryotic pathway in which double stranded RNA (dsRNA) triggers destruction of related RNAs in the cell.
Early studies in plants pointed to a role for this pathway as a defense against viruses. Most known plant viruses have RNA genomes and
replicate via dsRNA intermediates, thereby serving as potent inducers of RNA silencing early in replication and as silencing targets later in
infection. Because RNA silencing is an antiviral mechanism, it is not surprising that many plant viruses encode suppressors of RNA silencing.
This review focuses on the currently known plant virus encoded suppressors of silencing and the functional assays used to identify these
proteins. Because they interfere with silencing at different points in the pathway, these viral suppressors are powerful tools to help unravel
the mechanism of RNA silencing in plants.
© 2004 Elsevier B.V. All rights reserved.

Keywords: RNA silencing; RNAi; Viral suppressor protein; HC-Pro; CMV 2b

1. Introduction Vance et al., 1995). Expression of HC-Pro in transgenic


plants allowed a broad range of heterologous viruses to ac-
RNA silencing is an adaptive defense mechanism that is cumulate beyond the normal level, suggesting that HC-Pro
triggered by double stranded RNA (dsRNA). Three initially blocked a general plant defense mechanism (Pruss et al.,
unrelated lines of research led to the recognition of RNA si- 1997). Remarkably, the mechanism blocked by HC-Pro was
lencing as an important means of defense against viruses and found to be RNA silencing (Anandalakshmi et al., 1998;
other nucleic acid invaders. The discovery that plant viruses Brigneti et al., 1998; Kasschau and Carrington, 1998). Since
encode suppressors of RNA silencing is a key element in the initial demonstration that HC-Pro blocks RNA silencing,
the story. The first line of research led to the discovery of many other plant viral suppressors of silencing have been
transgene-induced RNA silencing. Attempts to over-express identified (see Table 1).
endogenous genes by introducing additional copies resulted
instead in turning off the endogenous gene as well as the
transgene (Napoli et al., 1990; Smith et al., 1990; van der 2. RNA silencing
Krol et al., 1990). The next piece of the puzzle came from
studies of pathogen-derived resistance in which RNA silenc- The term RNA silencing refers to a set of related pathways
ing directed against a viral transgene provided resistance to found in a broad range of eukaryotic organisms. Genetic and
any virus carrying the targeted sequence (Baulcombe, 1996; biochemical experiments have established a general mecha-
Dougherty and Parks, 1995; English et al., 1996; Goodwin nistic model for these related pathways and identified factors
et al., 1996; Lindbo et al., 1993; Smith et al., 1994). Thus, that are required for RNA silencing in a variety of organ-
viruses could be targets of RNA silencing. The third clue isms (Fig. 1). The process is initially triggered by dsRNA,
came from studies of synergistic viral diseases caused by which can be introduced experimentally or arise from en-
certain pairs of co-infecting viruses. A viral protein called dogenous transposons, replicating RNA viruses, or the tran-
helper component proteinase (HC-Pro) was shown to medi- scription of transgenes. The dsRNA trigger is cleaved by a
ate one class of viral synergistic disease (Shi et al., 1997; ribonuclease III (RNAse III)-like enzyme termed Dicer into
21–24 nucleotide duplexes termed short-interfering RNAs
(siRNAs) (Bernstein et al., 2001; Hamilton and Baulcombe,
∗ Corresponding author. 1999; Zamore et al., 2000). The production of siRNAs by
E-mail address: vance@biol.sc.edu (V.B. Vance). Dicer is an ATP-dependent step (Bernstein et al., 2001;

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.020
98
Table 1
Plant viral suppressors of RNA silencing
Genus Virus Suppressor Evidence Reference

Carmovirus Turnip crinkle virus (TCV) CP TCV infection does not reverse silencing. In agro-coinfiltration Qu et al. (2003) and Thomas et al. (2003)
assay, CP blocks sense and antisense induced local silencing and
prevents systemic silencing.
Closterovirus Beet yellows virus (BYV) p21 Suppresses inverted repeat (IR) induced local silencing in Reed et al. (2003)
Beet yellow stunt virus (BYSV) p22 agro-coinfiltration assay. BYV p21 corresponds to BYSV p22.
Cucumovirus Cucumber mosaic virus (CMV) 2b Infection with CMV or with PVX-2b vector blocks silencing. Li et al. (2002), see text
Tomato aspermy virus (TAV) Interferes with systemic signal (see text).
Furovirus Beet necrotic yellow vein virus (BNYVV) P14 Agro-coinfiltration assay with sense induced silencing. BNYVV Dunoyer et al. (2002)

B.M. Roth et al. / Virus Research 102 (2004) 97–108


P14 corresponds to PCV P15.
Geminivirus African cassava mosaic virus (ACMV) AC2 Infection with ACMV, PVX-AC2, or PVX-C2 reverses silencing. Dong et al. (2003), Voinnet et al. (1999)
Tomato yellow leaf curl virus-China (TYLCV-C) C2 Blocks sense induced silencing in agro-coinfiltration assay. AC2 and van Wezel et al. (2002)
and C2 are homologs.
Hordeivirus Barley stripe mosaic virus (BSMV) ␥b RNA mediated cross protection between PVX-GFP and TMV-GFP Yelina et al. (2002)
Poa semilatent virus (PSLV) vectors is eliminated when ␥b is expressed from the PVX vector.
Pecluvirus Peanut clump virus (PCV) P15 PCV infection blocks silencing. p15 blocks local and delays Dunoyer et al. (2002)
systemic sense-induced silencing in agro-coinfiltration assay.
Polerovirus Beet western yellows virus (BWYV) PO BWYV PO suppresses local but not systemic sense-induced Pfeffer et al. (2002)
Cucurbit aphid-borne yellows virus (CABYV) silencing in agro-coinfiltration assay. CABYV PO tested only on
local silencing.
Potexvirus Potato virus X (PVX) p25 PVX infection does not suppress silencing. In agro-coinfiltration, See text
p25 blocks systemic but not always local silencing (see text).
Potyvirus Potato virus Y (PVY) HC-Pro Evidence from multiple types of assay. Does not block systemic See text
Tobacco etch virus (TEV) silencing in stable expression grafting assay, but does in
agro-coinfiltration assay (see text).
Sobemovirus Rice yellow mottle virus (RYMV) P1 Infection with PVX-P1 viral vector reverses silencing. Voinnet et al. (1999)
Tenuivirusa Rice hoja blanca virus (RHBV) NS3 Agro-coinfiltration assay of sense induced local silencing. Bucher et al. (2003)
Tombusvirus Tomato bushy stunt virus (TBSV) P19 Limited activity in reversal of silencing; strong activity in Voinnet et al. (2003), Qu and Morris
Cymbidium ringspot virus (CymRSV) agro-coinfiltration (see text). AMCV (artichoke mottled crinkle (2002) and Takeda et al. (2002), see text
virus) P19 also works as a suppressor.

Tospovirusa Tomato spotted wilt virus (TSWV) NSs TSWV infection reverses silencing. In agro-coinfiltration, NSs Bucher et al. (2003) and Takeda et al. (2002)
suppressed sense, but not IR, induced local and systemic silencing.
a Tospoviruses and tenuiviruses replicate in their insect vectors and in plants.
B.M. Roth et al. / Virus Research 102 (2004) 97–108 99

One fascinating aspect of RNA silencing is that it is


non-cell-autonomous, and this feature may reflect the antivi-
ral nature of the process (for a recent review, see Mlotshwa
et al., 2002b). Virus infection usually starts with entry via
a small wound. The virus replicates in the initially infected
cell and then moves into adjacent cells, spreading from cell
to cell until it enters the vascular system, which allows rapid
movement to distant parts of the plant. In response, the host
plant initiates RNA silencing against the viral RNA and pro-
duces the mobile silencing signal. The mobile signal moves
along the same route the virus takes. Thus, the plant and the
virus enter a race. If the virus moves ahead of the signal, it
can establish infection when it enters the distant cells. How-
ever, if the mobile silencing signal gets there first, the virus
will enter the distant cell only to find itself targeted by RNA
silencing, and the infection will fail to become systemic.
One major class of viral suppressors comprises proteins that
block systemic silencing, suggesting the co-evolution of de-
fense and counter-defense between the host plant and the
invading virus at the level of systemic spread.

3. Functional assays used to identify suppressors of


RNA silencing

Three major approaches have been widely used to iden-


tify plant viral suppressors of RNA silencing: (1) transient
expression assays, (2) the reversal of silencing assay, and (3)
stable expression assays. These assays are described below
and shown in cartoon form in Figs. 2 through 4.

3.1. Transient expression assays—Agrobacterium


co-infiltration

Fig. 1. RNA silencing pathway. This approach provides a rapid and easy test of sup-
pressor activity and is currently the technique most com-
monly used to identify viral suppressors (Llave et al., 2000;
Zamore et al., 2000) and probably involves interactions Voinnet et al., 2000). The method makes use of a commonly
with other proteins, including an argonaute-like protein, used bacterial pathogen of plants, Agrobacterium tumefa-
a dsRNA binding protein, and an RNA helicase (Tabara ciens. The Agrobacterium serves two purposes: one strain
et al., 2002). There are four Dicer-like (DCL) homologs is used to induce RNA silencing of a reporter gene (usually
in Arabidopsis; however, the specific enzyme responsible green fluorescent protein (GFP)), and another strain is used
for siRNA production in plants has not yet been identified. to express the candidate suppressor. The overall strategy is
The siRNAs produced from a fully double-stranded RNA to co-infiltrate mixtures of the two bacterial strains (one act-
substrate by Dicer have distinctive characteristics: they ing to induce silencing, the other to suppress it) into a plant
represent both polarities and have two nucleotide 3 over- leaf and then examine the infiltrated patch over time for si-
hangs with 5 phosphate and 3 hydroxyl groups (Elbashir lencing of the reporter (Fig. 2A). Nicotiana benthamiana is
et al., 2001a,b). In another ATP-dependent step (Nykanen well suited to these assays because the leaves are easily in-
et al., 2001), the siRNAs are denatured and incorporated filtrated and produce high quantities of protein in response
into a multi-subunit endonuclease silencing complex called to agro-infiltration. Non-transgenic N. benthamiana as well
RNA-induced silencing complex (RISC; Hammond et al., as N. benthamiana expressing the reporter gene can be used.
2000). Within the activated RISC, single-stranded siRNAs In a typical experiment with Agrobacterium expressing GFP
act as guides to bring the complex into contact with com- as the inducer, the infiltrated patch initially expresses high
plementary mRNAs and thereby cause their degradation levels of GFP and glows bright green under ultraviolet (UV)
(Bernstein et al., 2001; Elbashir et al., 2001a; Hammond light (Fig. 3A, first panel). However, in three to five days,
et al., 2000, 2001; Zamore et al., 2000). the procedure triggers local RNA silencing, and the patch
100 B.M. Roth et al. / Virus Research 102 (2004) 97–108

Fig. 2. Cartoon guide to transient expression assays. (A) Assay for suppressors of local silencing. (B) Assay for suppressors of systemic silencing.

Fig. 3. Agrobacterium-induced systemic silencing (A) and cartoon guide to reversal of silencing assay (B).
B.M. Roth et al. / Virus Research 102 (2004) 97–108 101

becomes a dirty red color under UV light due to a mixture Thus, candidate suppressors from heterologous viruses can
of green from residual GFP and red from chlorophyll in the be tested in the reversal of silencing assay by expressing
leaves (Fig. 3A, second panel). If the candidate suppressor them from a PVX vector (Fig. 3B). Many viral suppressors
expressed from the co-infiltrated Agrobacterium interferes have been identified by expression from PVX in this manner
with RNA silencing, the patch will remain bright green: if it (Table 1; Voinnet et al., 1999).
does not, the patch will turn red (Fig. 2A). Variations of this
technique include using Agrobacterium expressing inverted 3.3. Stable expression assays
repeat (IR) or viral cDNA constructs to induce silencing
in combination with or in place of Agrobacterium express- In this approach, a stable transgenic line expressing a can-
ing a sense gene construct (Johansen and Carrington, 2001; didate suppressor of silencing (usually initially identified in
Voinnet et al., 2000). one of the previous two assays) is crossed to a series of
Another variation of the transient expression approach has well-characterized transgenic lines silenced for a reporter
been widely used to investigate the effect of silencing sup- gene (Fig. 4A; Anandalakshmi et al., 1998; Kasschau and
pressors on the mobile silencing signal (Fig. 2B). For this Carrington, 1998). An advantage of this approach is that
purpose, infiltration is performed on a transgenic N. ben- it offers the opportunity to examine the effect of the sup-
thamiana line that expresses GFP (line 16C, Ruiz et al., pressor on different well-defined types of transgene-induced
1998). Early experiments using an Agrobacterium strain ex- RNA silencing, thus, providing information about the mech-
pressing a sense GFP construct as the inducer of silencing anism of suppression. The stable expression assays are also
demonstrated that local induction of GFP silencing produced well suited to investigate the role of suppressors in sys-
a mobile silencing signal that moved out of the infiltrated temic silencing using grafting (Fig. 4B; Guo and Ding, 2002;
spot, along veins, and into surrounding tissues, leaving a trail Mallory et al., 2001, 2003). In these assays, the ability of a
of red marking its path (Fig. 3A, third panel) and eventually plant to send a mobile silencing signal is assayed by graft-
silencing GFP throughout the whole plant (Fig. 3A, fourth ing an expressing line onto the top of it (the bottom plant
panel) (Voinnet and Baulcombe, 1997). Thus, the ability is called the rootstock and the expressing plant grafted onto
of suppressors to interfere with systemic silencing can be the top is called the scion) (Palauqui et al., 1997). If the sup-
tested in this system by co-infiltrating the inducer together pressor of silencing in the rootstock blocks either the pro-
with a putative suppressor of silencing and observing long duction or movement of the systemic silencing signal, then
enough to determine whether the plant becomes systemi- the transgene in the scion will continue to be expressed. If
cally silenced (Fig. 2B). a suppressor does not block the systemic silencing signal
in either of these ways, then the transgene in the scion will
3.2. Reversal of silencing assay become silenced (Fig. 4B).

This versatile assay can be used to first identify candi-


date viruses that may suppress silencing as a major line of 4. Mechanism of suppression
counter-defense (Fig. 3B; Brigneti et al., 1998). A modifi-
cation of the same technique can then be used to identify The assays discussed above have proven extremely use-
the specific viral gene product that suppresses silencing. The ful as rapid and sensitive methods to identify suppressors
overall strategy is to infect a silenced plant with the candi- of silencing. They have also enabled rudimentary charac-
date virus and determine whether the silenced phenotype is terization of suppressor mechanism, but conflicting results
reversed (Fig. 3B). The most common version of this tech- from different assays have made it difficult to draw firm con-
nique uses the GFP-expressing transgenic N. benthamiana clusions for many suppressors. Interestingly, the currently
line 16C described above. Soon after germination (at about known suppressors share no obvious similarities at either
the four-leaf stage), the plant is infiltrated with Agrobac- the nucleic acid or the protein level, perhaps reflecting dif-
terium expressing GFP, which triggers local silencing and ferences at the mechanistic level as well. At present, two
then systemic silencing as described above. Ultimately, the major classes of suppressor action have been identified.
plant becomes completely silenced for GFP (red in UV light)
(Fig. 3A, fourth panel). At this point, the plant is inocu- 4.1. Suppressors that affect small RNA metabolism
lated with the virus being tested for suppressor activity. If
virus infection allows the plant to express GFP, the infect- Many suppressors reduce the accumulation of siRNAs,
ing virus likely encodes a suppressor of silencing (Voinnet raising the possibility that silencing is blocked at the step
et al., 1999). Individual genes can be assayed for suppres- at which Dicer processes the dsRNA that triggers silencing.
sor activity using a modification of the reversal of silencing Thus, it may be that many suppressors prevent silencing by
technique that employs potato virus X (PVX). PVX is an ef- blocking production of the siRNAs that provide the sequence
ficient vector for systemic expression of heterologous genes specificity of the process. A second mechanism involving
in N. benthamiana and does not, itself, encode a suppres- siRNAs is exemplified by the suppressor P19, which has
sor of silencing that works in the reversal of silencing assay. been shown to bind siRNAs, perhaps sequestering them and
102 B.M. Roth et al. / Virus Research 102 (2004) 97–108

Fig. 4. Cartoon guide to stable expression assays. (A) Genetic crosses with silenced transgenic lines. (B) Grafting assay for suppression of systemic
silencing.

thereby blocking their function (Silhavy et al., 2002). Inter- (Mallory et al., 2001, 2003), it interfered with systemic si-
estingly, the effect of suppressors on small RNA metabolism lencing in the transient expression assay (Hamilton et al.,
may extend to other types of small regulatory RNAs. For 2002). Because HC-Pro is a powerful suppressor of local
example the silencing suppressor HC-Pro affects the accu- silencing in all assays, these results suggest that HC-Pro pri-
mulation not only of the siRNAs that mediate silencing but marily affects local silencing, but also has a smaller effect
also of endogenous microRNAs (miRNAs), which have been on systemic silencing. In contrast, CMV 2b primarily targets
implicated in development (Kasschau et al., 2003; Mallory systemic silencing because it blocks movement of the sig-
et al., 2002b). Surprisingly, although HC-Pro blocks the ac- nal in many assays, but has a lesser effect on local silencing
cumulation of siRNAs, it enhances the accumulation of miR- (Brigneti et al., 1998; Bucher et al., 2003; Guo and Ding,
NAs (Mallory et al., 2002b). Even more interesting, there 2002). It may not be surprising that a primary effect of a
is evidence that HC-Pro might block the function of miR- particular suppressor on one aspect of silencing could lead,
NAs (Kasschau et al., 2003). It remains to be seen if other perhaps through feedback mechanisms, to secondary effects
viral suppressors also affect the miRNA pathway, perhaps on other parts of the pathway, thereby making it appear that
using that pathway to turn off expression of genes required the suppressor works at multiple points.
for silencing.
4.3. Conflicting results using different assays
4.2. Suppressors that affect systemic silencing
Why do different assays give different results when look-
Systemic silencing can be assayed in either transient ex- ing at effects of viral suppressors on systemic silencing?
pression experiments or in experiments with stably trans- The major conflicts are seen with stable expression assays
formed transgenic lines (Figs. 2B and 4B). Many suppressors versus transient ones and probably reflect a number of in-
have been demonstrated to block systemic silencing in at trinsic differences between the systems. Transient expres-
least one of these assays (Table 1). Because of the conflicting sion assays and the reversal of silencing assay use Agrobac-
results sometimes obtained with different assays, the most terium to induce silencing. Because Agrobacterium is a plant
successful attempts to determine whether a suppressor pri- pathogen, infiltration likely induces plant defensive and bac-
marily affects systemic silencing have used a multi-pronged terial counter-defensive interactions, which might modify
approach rather than relying on just one type of assay. the activity of some viral suppressors. Thus, attempts to
For example, although HC-Pro did not block systemic si- compare suppressor activities or to understand the mecha-
lencing in grafting experiments using stable transgenic lines nism of action of the different suppressors are complicated
B.M. Roth et al. / Virus Research 102 (2004) 97–108 103

by the largely unknown effects of Agrobacterium on the although reduced, was not eliminated. However, grafting
system. Similarly, the reversal of silencing assay involves experiments definitively demonstrated that CMV 2b blocks
virus infection and likely induces accompanying defense the movement of the systemic silencing signal. In the first
and counter-defensive responses that complicate the inter- protocol, 6b5 rootstocks suppressed for silencing by CMV
pretation of results. Further sources of variation include dif- 2b did not silence grafted GUS-expressing scions, showing
ferences in the level and time course of expression of the that expression of CMV 2b in the rootstocks prevented
silencing suppressor and silencing inducer in the different production or transmission of the systemic silencing signal
systems. (Fig. 5A, first panel). In the second protocol, a small spacer
Of the three widely used types of assay (Section 3), stable of transgenic tobacco expressing CMV 2b, grafted between
expression assays are the ones most free of complications a GUS-silenced 6b5 rootstock and a GUS-expressing scion,
due to extraneous pathogens. In addition, use of the same blocked systemic silencing (Fig. 5A, second panel). Thus,
well-characterized silenced transgenic line to test the effects CMV 2b prevents transmission of the systemic silencing
of different viral suppressors affords a degree of standard- signal in a stable expression assay.
ization from lab to lab not yet found with transient expres- The experiments described above provide compelling ev-
sion assays. It is important to use well-characterized lines idence that the mechanism of action of CMV 2b, at least in
to compare suppressor activities because different types of part, is to block systemic silencing. Guo and Ding (2002)
transgenes trigger silencing in different ways (Vance and also report similar conclusions based on Agrobacterium
Vaucheret, 2001). co-infiltration experiments. Paradoxically, the same type of
Understanding the basis of the conflicting results given co-infiltration experiments in another laboratory produced a
by the currently widely used assays will likely offer con- different result: CMV 2b delayed but did not block systemic
siderable insight into the mechanisms of suppressor action. silencing (Hamilton et al., 2002). A possible cause of this
Alternative approaches such as yeast two-hybrid, biochemi- discrepancy is that the two labs were using different ratios
cal, and localization studies (see Section 5) are increasingly of silencing inducer to suppressor in the co-infiltrations
being used to investigate the mechanisms of action of the (S.W. Ding, personal communication), suggesting that a
different viral suppressors of silencing and will undoubtedly standardized methodology for co-infiltration assays would
help clear up the confusion. help resolve some of the current inconsistencies.
How does CMV 2b protein block the movement of the
mobile silencing signal? The ability of 2b protein to pre-
5. Better studied suppressors of silencing vent the transmission of the signal suggests that it either se-
questers or inactivates the signal in the phloem stream. One
5.1. Cucumber mosaic virus (CMV) 2b possibility is that 2b acts directly by binding to the signal.
However, the finding that 2b localizes to the nucleus (Lucy
The CMV 2b protein was one of the first identified sup- et al., 2000) suggests that the suppressor acts indirectly, per-
pressors of RNA silencing and also one of the best studied haps by activating one or more processes that subsequently
from a mechanistic standpoint. The initial indication that affect the signal.
CMV 2b suppressed silencing came from the reversal of
silencing assay in which 2b expressed from PVX could 5.2. Potyviral helper-component protease (HC-Pro)
prevent the initiation of silencing but could not reverse si-
lencing that was already established (Brigneti et al., 1998). HC-Pro was the first identified suppressor of RNA silenc-
That early result raised the possibility that 2b might block ing. The original reports demonstrated that it suppresses both
systemic silencing. Subsequently, stable expression assays transgene- and virus-induced silencing (Anandalakshmi
and grafting experiments provided an elegant demonstration et al., 1998; Kasschau and Carrington, 1998). In contrast to
that 2b blocks the movement of the systemic silencing signal CMV 2b protein, it is able to reverse established silencing
(Guo and Ding, 2002). The stable expression assays made in the reversal of silencing assay (Brigneti et al., 1998), sug-
use of a well-characterized silenced tobacco line called gesting that the two suppressors work at different steps in the
6b5 (Elmayan and Vaucheret, 1996), which is silenced for silencing pathway. Although HC-Pro alone has suppressor
the reporter gene GUS and is a model for sense-transgene activity (Anandalakshmi et al., 1998; Brigneti et al., 1998),
induced RNA silencing. The 6b5-GUS locus triggers si- it is frequently expressed as the proteinase 1 (P1)/HC-Pro
lencing even when it is hemizygous, produces high levels polyprotein in suppression of silencing studies. The
of GUS siRNAs, and is highly effective in the production P1/HC-Pro construct is the N-terminal portion of the natural
and transmission of a systemic silencing signal as demon- viral polyprotein and allows HC-Pro to be proteolytically
strated by grafting experiments. A genetic cross between processed just as when expressed from the viral genome.
line 6b5 and a stable transgenic tobacco line expressing Some evidence suggests that P1 might enhance HC-Pro
CMV 2b established that 2b could partially suppress local activity as a suppressor of silencing (Pruss et al., 1997).
sense-transgene induced silencing: offspring of the cross A variety of approaches, including both transient and sta-
accumulated GUS mRNA, but GUS siRNA accumulation, ble expression assays, have been used to investigate how
104 B.M. Roth et al. / Virus Research 102 (2004) 97–108

Fig. 5. Cartoon guide to (A) CMV 2b and (B) HC-Pro grafting experiments.

HC-Pro suppresses RNA silencing. Despite some conflicting correlate with systemic silencing in transient expression
results, at least one common finding has emerged: HC-Pro assays (Hamilton et al., 2002) but not in stable expression
affects small RNA metabolism. Exactly how HC-Pro exerts assays (Mallory et al., 2002b). Finally and surprisingly,
its effect on small RNAs and how this leads to suppression the accumulation of endogenous miRNAs is increased in
of silencing are questions that remain to be resolved. The both tobacco and Arabidopsis transgenic lines that express
interaction(s) of HC-Pro and its cellular effector(s) probably HC-Pro (Kasschau et al., 2003; Mallory et al., 2002b),
occur in the cytoplasm because HC-Pro is found primarily suggesting a more general role in the biogenesis of small
in cytoplasm (Mlotshwa et al., 2002a). regulatory RNAs. A recent report that HC-Pro enhances
the stability of several miRNA target messages raises the
5.2.1. HC-Pro and small RNAs possibility that HC-Pro affects the function as well as the
HC-Pro has been reported to alter the accumulation of biogenesis of small RNAs (Kasschau et al., 2003).
several classes of small RNAs: the siRNAs that direct RNA
degradation during silencing, a novel class of slightly-larger 5.2.2. HC-Pro and systemic silencing
small RNAs of unknown function, and the endogenous mi- In stable expression experiments utilizing grafting,
croRNAs (miRNAs) that have been implicated in regulation HC-Pro failed to block systemic silencing (Fig. 5B, first
of development. In stable expression assays in tobacco, panel; Mallory et al., 2001, 2003). Three different trans-
HC-Pro has been reported to suppress three classes of genic tobacco lines were used as rootstocks, representing
transgene-induced RNA silencing, in each case interfering three different means of inducing silencing: sense trans-
with the accumulation of siRNAs (Mallory et al., 2001, gene, inverted repeat transgene, and amplicon transgene.
2002b). Similarly, siRNA accumulation is dramatically In the case of the amplicon-transgene induced silencing,
reduced during HC-Pro suppression of silencing in tran- in which the transgene is the cDNA of a replication com-
sient expression assays (Johansen and Carrington, 2001; petent virus, HC-Pro actually promoted systemic silencing
Llave et al., 2000). In stable expression assays in tobacco, (Mallory et al., 2003). These results suggest that production
HC-Pro suppression of IR transgene-induced silencing and transmission of the systemic silencing signal are largely
or amplicon-transgene-induced silencing—but not sense unaffected by HC-Pro, implying that HC-Pro suppression
transgene-induced silencing—resulted in the accumulation of silencing occurs downstream of the signal. To test that
of a novel class of slightly-larger small RNAs (Mallory hypothesis, the effect of HC-Pro on perception of and/or
et al., 2002b). Similar slightly-larger small RNAs have also response to the silencing signal was tested in additional
been observed in transient expression assays (Hamilton grafting experiments (Fig. 5B, second panel; Mallory et al.,
et al., 2002). The function of this class of small RNAs is 2001). A silenced GUS line previously shown to silence
not clear; however, they do not appear to act as siRNAs GUS systemically across a graft junction was used as root-
because their presence is not correlated with RNA degra- stock, and the scion was a line that expressed both GUS
dation (Hamilton et al., 2002; Mallory et al., 2002b). They and HC-Pro. The scion failed to become silenced for GUS,
B.M. Roth et al. / Virus Research 102 (2004) 97–108 105

showing definitively that HC-Pro works downstream of the virus-induced gene silencing and did not suppress silencing
systemic silencing signal: The rootstock is known to send induced by a defective interfering RNA that accumulates to
a signal; therefore, in the presence of HC-Pro, the scion high levels (Qiu et al., 2002). In transient expression as-
either fails to perceive the signal or fails to respond to it. says, however, P19 is a star, blocking both local and sys-
Although the grafting experiments described above make temic silencing and apparently eliminating all small RNAs
a convincing case that HC-Pro does not block the systemic (Hamilton et al., 2002; Silhavy et al., 2002; Takeda et al.,
silencing signal, results from Agrobacterium co-infiltration 2002; Voinnet et al., 2003). Interestingly, biochemical stud-
assays suggest that this suppressor does interfere with sys- ies have shown that P19 binds siRNAs and that binding de-
temic silencing (Hamilton et al., 2002). There are a num- pends on characteristics of RNase III products (dsRNAs with
ber of possible reasons for obtaining different results with two nucleotide 3 overhangs) (Silhavy et al., 2002). This re-
the two assays. First, the stable transgenic lines express the sult raises the possibility that P19 suppresses silencing by
P1/HC-Pro polyprotein, whereas the transient assay exper- sequestering siRNAs, thereby preventing their incorporation
iments used a construct that encodes only HC-Pro and has into the RISC complex to serve as guides. This is a novel
a methionine substituted for the natural N-terminal amino mechanism among suppressors, and because it theoretically
acid of the protein. The protein produced in the transient as- stems from an intrinsic property of the protein to bind func-
say, therefore, is not identical to HC-Pro produced from the tional siRNAs, it is possible that P19 could interfere with
virus. Thus, it is possible that the difference between the two silencing in a broad range of different plants (and even other
assays reflects this small difference in the proteins. Indeed, organisms).
earlier experiments expressing either P1/HC-Pro or HC-Pro
(with an N-terminal methionine) from a PVX vector reported 5.4. Potato virus X (PVX) p25
a dramatic difference in the effect on PVX replication (Pruss
et al., 1997). A second possibility is that the result in the Three of five potexviruses tested in the reversal of silenc-
transient assay depends on the ratio of inducer to suppres- ing assay suppressed RNA silencing, although PVX was
sor, as suggested for CMV 2b protein (see Section 5.1), and one that did not show suppressor activity (Voinnet et al.,
this possibility could be tested by using a range of ratios. 1999). Paradoxically, however, PVX is the only potexvirus
A more exciting possibility is that the discrepancy reflects for which a suppressor of RNA silencing has subsequently
an intrinsic difference between the two assays, as discussed been characterized. In grafting experiments designed to sep-
in Section 4.3. Thus, HC-Pro might block systemic silenc- arate movement of the systemic silencing signal from that
ing in some circumstances, but not in others, and identify- of the virus in virus induced silencing (VIGS), Voinnet et al.
ing the important variables will help in our understanding (2000) unexpectedly found that PVX infection failed to pro-
of HC-Pro suppression of silencing. duce systemic silencing independent of the presence of virus,
suggesting that a PVX-encoded protein interferes with pro-
5.2.3. HC-Pro interacting proteins duction or transmission of the systemic signal. Protein p25,
If HC-Pro works by interacting with components or regu- which is required for cell-to-cell movement of potexviruses,
lators of the silencing machinery, identification of plant pro- was identified as the culprit in a set of experiments in which
teins that interact with HC-Pro should provide clues about deletion derivatives of replication competent PVX-GFP viral
its mechanism of action. Using the yeast two-hybrid sys- vector constructs were agro-infiltrated into plants express-
tem, an HC-Pro-interacting protein called regulator of gene ing GFP. The PVX-GFP constructs were restricted to ini-
silencing calmodulin-like protein (rgsCaM) was identified tially infiltrated cells because they all lacked coat protein,
(Anandalakshmi et al., 2000). Like HC-Pro, rgsCaM could which is also required for potexviral movement. Induction
reverse silencing when expressed from PVX in the rever- of systemic silencing in these experiments, therefore, de-
sal of silencing assay. In addition, when over-expressed in pends entirely on the systemic signal. Only viral constructs
stable expression experiments, rgsCaM caused a develop- from which p25 expression had been eliminated induced
mental phenotype typical of HC-Pro-expressing transgenic widespread systemic silencing in all plants.
lines and interfered with virus induced gene silencing. Be- Agro-coinfiltration experiments showed that p25, with-
cause calmodulins are classic signaling molecules, these re- out any other PVX protein, was sufficient to block sys-
sults raise the possibility that HC-Pro suppresses silencing temic silencing. Moreover, p25 blocked systemic silencing
indirectly via a signal cascade involving rgsCaM. whether silencing was induced by a simple transgene con-
struct (35S-GFP) or by a replication competent PVX-GFP
5.3. Tombusvirus P19 construct. The effect of p25 on local silencing in these ex-
periments, however, depended on the nature of the construct
This protein has been an exciting addition to the reper- used to induce silencing. Although p25 suppressed local si-
toire of plant viral suppressors. Initially described in the re- lencing in agro-coinfiltration assays with 35S-GFP, it did
versal of silencing assay (Voinnet et al., 1999), it appeared not suppress local silencing induced by infiltration of repli-
to be a weak suppressor, only reversing silencing in the re- cation competent PVX-GFP constructs. p25 suppression of
gion of veins. Similarly, P19 delayed but did not prevent systemic silencing in agro-coinfiltration assays is correlated
106 B.M. Roth et al. / Virus Research 102 (2004) 97–108

with the absence of a slightly-larger class of small RNAs, RNA silencing. Much of what is currently known about
possibly indicating the step in the pathway affected by p25 the RNA silencing pathway comes from elegant in vitro
(Hamilton et al., 2002). and genetic studies in organisms other than plants (for a
Although these transient expression experiments make a recent review see Tijsterman et al., 2002). In plants, tra-
convincing case that PVX p25 blocks systemic silencing, ditional genetic approaches have led to the identification
a different result was obtained in experiments using trans- of a number of cellular genes required for RNA silencing
genic plants constitutively expressing white clover mosaic (Dalmay et al., 2000, 2001; Fagard et al., 2000; Mourrain
potexvirus (WClMV) p25. Agro-infiltration of a 35S-IR con- et al., 2000). Surprisingly, however, all of these genes are
struct systemically silenced these plants (Foster et al., 2002). required for sense-, but not IR-transgene induced silencing
Moreover, the systemic silencing reverted a severe develop- (Boutet et al., 2003). The plant viral suppressors, many of
mental phenotype, indicating that the systemic signal was which appear to work downstream of dsRNA, provide a
able to enter the shoot apical meristem. Thus, WClMV p25 novel means of entry into parts of the silencing pathway
did not prevent systemic silencing in this experimental sys- that are not easily accessible by genetic means. The cur-
tem. Whether the difference in the results obtained with the rently known suppressors appear to work at different steps
PVX and WClMV p25 proteins reflects differences in the in silencing, thereby providing access to a number of points
assays or in the activities of the proteins has not yet been in the pathway where silencing can be controlled.
determined. Identifying host proteins that interact with a viral sup-
pressor of RNA silencing is one very promising approach
that is being used to take advantage of viral suppressors
6. Do all plant viruses have suppressors of silencing? to elucidate the silencing pathway. The yeast two-hybrid
system has been used to find tobacco proteins that in-
The discovery that plants have a generalized antiviral de- teract with HC-Pro, identifying a calmodulin-related pro-
fense mechanism triggered by dsRNA has revolutionized tein called rgsCaM that suppresses RNA silencing when
our thinking about plant–virus interactions. In the euphoric over-expressed (Anandalakshmi et al., 2000). This result
aftermath of the initial identification of plant viral suppres- suggests that a calcium controlled signal transduction path-
sors of silencing, a popular expectation was that most or all way involving rgsCaM is one of the mechanisms regulating
plant viruses would encode a suppressor of RNA silencing. RNA silencing. Intriguingly, in the case of geminiviruses,
Although many different viral suppressors have been iden- yeast two-hybrid studies have identified SNF1 kinase as a
tified, the fact that HC-Pro helps so many different viruses cellular interactor of the tomato golden mosaic virus AL2
suggests that a lot of viruses do not effectively suppress si- protein (Hao et al., 2003). AL2 is a homologue of the
lencing. Such viruses may have evolved other ways to try ACMV and TYLCV-C suppressors of silencing. Whether
to avoid silencing, such as by replicating within spherules the interaction of AL2 with SNF1, which is a regulator of
in the ER (Schwartz et al., 2002), where the dsRNA is hid- metabolism in response to stress, plays any role in suppres-
den, or by replicating and moving rapidly enough to outrun sion of silencing is unknown as yet.
the mobile silencing signal. Furthermore, plants have other The potential of using viral suppressors to help understand
defense mechanisms, and silencing might not be the major the mechanism of RNA silencing in plants is largely un-
threat for all viruses. Some viruses, therefore, may well have tapped, and these studies promise to be an exciting and fer-
suppressors of other defense pathways. tile area of research. The recent identification of a viral sup-
pressor that works in animal cells (Li et al., 2002) offers the
possibility that such proteins may provide a similar tool to
7. Suppressors of silencing as tools understand the silencing pathway in other organisms as well.

The finding that certain viral proteins suppress RNA si- Note added in proof
lencing has provided a new tool for technologies utilizing
genetically modified plants and is, therefore, of practical sig- The tomato mosaic virus replication protein has recently
nificance. Many biotechnological applications are impaired been reported to suppress RNA silencing (Kubota, K., Tsuda,
by RNA silencing, and suppressors of silencing can be used S., Tamai, A., Meshi, T., 2003. Tomato mosaic virus repli-
to attain consistent, high-level expression of transgenes in cation protein suppresses virus-targeted posttranscriptional
plants (Mallory et al., 2002a; Voinnet et al., 2003). With si- gene silencing. J. Virol. 77, 11016–11026).
lencing under control, transgenic plants can be engineered to
produce a range of transgene expression: moderate levels of
expression to produce desired plant traits or very high-level Acknowledgements
expression to use the plant as a factory producing pharma-
ceuticals, vaccines or other high-value gene products. VBV gratefully acknowledges support from the USDA
Perhaps more importantly, viral suppressors of silencing Competitive Grants Program, NIH, and Dow AgroSciences
also provide unique tools to understand the mechanism of LLC.
B.M. Roth et al. / Virus Research 102 (2004) 97–108 107

References Guo, H.S., Ding, S.W., 2002. A viral protein inhibits the long range
signaling activity of the gene silencing signal. EMBO J. 21, 398–407.
Hamilton, A., Voinnet, O., Chappell, L., Baulcombe, D., 2002. Two classes
Anandalakshmi, R., Marathe, R., Ge, X., Herr Jr., J.M., Mau, C., Mallory,
of short interfering RNA in RNA silencing. EMBO J. 21, 4671–4679.
A., Pruss, G., Bowman, L., Vance, V.B., 2000. A calmodulin-related
Hamilton, A.J., Baulcombe, D.C., 1999. A species of small antisense RNA
protein that suppresses posttranscriptional gene silencing in plants.
in posttranscriptional gene silencing in plants. Science 286, 950–952.
Science 290, 142–144.
Hammond, S.M., Bernstein, E., Beach, D., Hannon, G.J., 2000. An
Anandalakshmi, R., Pruss, G.J., Ge, X., Marathe, R., Mallory, A.C.,
RNA-directed nuclease mediates post-transcriptional gene silencing in
Smith, T.H., Vance, V.B., 1998. A viral suppressor of gene silencing
Drosophila cells. Nature 404, 293–296.
in plants. Proc. Natl. Acad. Sci. USA 95, 13079–13084.
Hammond, S.M., Caudy, A.A., Hannon, G.J., 2001. Post-transcriptional
Baulcombe, D.C., 1996. RNA as a target and an initiator of
gene silencing by double-stranded RNA. Nat. Rev. Genet. 2, 110–119.
post-transcriptional gene silencing in transgenic plants. Plant Mol.
Hao, L., Wang, H., Sunter, G., Bisaro, D.M., 2003. Geminivirus AL2 and
Biol. 32, 79–88.
L2 proteins interact with and inactivate SNF1 kinase. Plant Cell 15,
Bernstein, E., Caudy, A.A., Hammond, S.M., Hannon, G.J., 2001. Role
1034–1048.
for a bidentate ribonuclease in the initiation step of RNA interference.
Johansen, L.K., Carrington, J.C., 2001. Silencing on the spot. Induction
Nature 409, 363–366.
and suppression of RNA silencing in the Agrobacterium-mediated
Boutet, S., Vazquez, F., Liu, J., Beclin, C., Fagard, M., Gratias, A., Morel,
transient expression system. Plant Physiol. 126, 930–938.
J.B., Crete, P., Chen, X., Vaucheret, H., 2003. Arabidopsis HEN1.
Kasschau, K.D., Carrington, J.C., 1998. A counterdefensive strategy of
A genetic link between endogenous miRNA controlling development
plant viruses: suppression of posttranscriptional gene silencing. Cell
and siRNA controlling transgene silencing and virus resistance. Curr.
95, 461–470.
Biol. 13, 843–848.
Kasschau, K.D., Xie, Z., Allen, E., Llave, C., Chapman, E.J., Krizan,
Brigneti, G., Voinnet, O., Li, W.X., Ji, L.H., Ding, S.W., Baulcombe, D.C.,
K.A., Carrington, J.C., 2003. P1/HC-Pro, a viral suppressor of RNA
1998. Viral pathogenicity determinants are suppressors of transgene
silencing, interferes with Arabidopsis development and miRNA unc-
silencing in Nicotiana benthamiana. EMBO J. 17, 6739–6746.
tion. Dev. Cell 4, 205–217.
Bucher, E., Sijen, T., De Haan, P., Goldbach, R., Prins, M., 2003.
Li, H., Li, W.X., Ding, S.W., 2002. Induction and suppression of RNA
Negative-strand tospoviruses and tenuiviruses carry a gene for a sup-
silencing by an animal virus. Science 296, 1319–1321.
pressor of gene silencing at analogous genomic positions. J. Virol.
Lindbo, J.A., Silva-Rosales, L., Proebsting, W.M., Dougherty, W.G., 1993.
77, 1329–1336.
Induction of a highly specific antiviral state in transgenic plants:
Dalmay, T., Hamilton, A., Rudd, S., Angell, S., Baulcombe, D.C., 2000.
implications for regulation of gene expression and virus resistance.
An RNA-dependent RNA polymerase gene in Arabidopsis is required
Plant Cell 5, 1749–1759.
for posttranscriptional gene silencing mediated by a transgene but not
Llave, C., Kasschau, K.D., Carrington, J.C., 2000. Virus-encoded suppres-
by a virus. Cell 101, 543–553.
sor of posttranscriptional gene silencing targets a maintenance step in
Dalmay, T., Horsefield, R., Braunstein, T.H., Baulcombe, D.C., 2001.
the silencing pathway. Proc. Natl. Acad. Sci. USA 97, 13401–13406.
SDE3 encodes an RNA helicase required for post-transcriptional gene
Lucy, A.P., Guo, H.S., Li, W.X., Ding, S.W., 2000. Suppression of
silencing in Arabidopsis. EMBO J. 20, 2069–2078.
post-transcriptional gene silencing by a plant viral protein localized
Dong, X., van Wezel, R., Stanley, J., Hong, Y., 2003. Functional charac- in the nucleus. EMBO J. 19, 1672–1680.
terization of the nuclear localization signal for a suppressor of post-
Mallory, A.C., Ely, L., Smith, T.H., Marathe, R., Anandalakshmi, R.,
transcriptional gene silencing. J. Virol. 77, 7026–7033.
Fagard, M., Vaucheret, H., Pruss, G., Bowman, L., Vance, V.B., 2001.
Dougherty, W.G., Parks, T.D., 1995. Transgenes and gene suppression: HC-Pro suppression of transgene silencing eliminates the small RNAs
telling us something new? Curr. Opin. Cell Biol. 7, 399–405. but not transgene methylation or the mobile signal. Plant Cell 13,
Dunoyer, P., Pfeffer, S., Fritsch, C., Hemmer, O., Voinnet, O., Richards, 571–583.
K.E., 2002. Identification, subcellular localization and some properties Mallory, A.C., Mlotshwa, S., Bowman, L.H., Vance, V.B., 2003. The
of a cysteine-rich suppressor of gene silencing encoded by peanut capacity of transgenic tobacco to send a systemic RNA silencing
clump virus. Plant J. 29, 555–567. signal depends on the nature of the inducing transgene locus. Plant J.
Elbashir, S.M., Lendeckel, W., Tuschl, T., 2001a. RNA interference is 35, 82–92.
mediated by 21- and 22-nucleotide RNAs. Genes Dev. 15, 188–200. Mallory, A.C., Parks, G., Endres, M.W., Baulcombe, D., Bowman,
Elbashir, S.M., Martinez, J., Patkaniowska, A., Lendeckel, W., Tuschl, T., L.H., Pruss, G.J., Vance, V.B., 2002a. The amplicon-plus system for
2001b. Functional anatomy of siRNAs for mediating efficient RNAi high-level expression of transgenes in plants. Nat. Biotechnol. 20,
in Drosophila melanogaster embryo lysate. EMBO J. 20, 6877–6888. 622–625.
Elmayan, T., Vaucheret, H., 1996. Expression of single copies of a strongly Mallory, A.C., Reinhart, B.J., Bartel, D., Vance, V.B., Bowman, L.H.,
expressed 35S transgene can be silenced post-transcriptionally. Plant 2002b. From the cover: a viral suppressor of RNA silencing dif-
J. 9, 787–797. ferentially regulates the accumulation of short interfering RNAs and
English, J.J., Mueller, E., Baulcombe, D.C., 1996. Suppression of virus micro-RNAs in tobacco. Proc. Natl. Acad. Sci. USA 99, 15228–
accumulation in transgenic plants exhibiting silencing of nuclear genes. 15233.
Plant Cell 8, 179–188. Mlotshwa, S., Verver, J., Sithole-Niang, I., Prins, M., Van Kammen,
Fagard, M., Boutet, S., Morel, J.B., Bellini, C., Vaucheret, H., 2000. A.B., Wellink, J., 2002a. Transgenic plants expressing HC-Pro show
AGO1, QDE-2, and RDE-1 are related proteins required for enhanced virus sensitivity while silencing of the transgene results in
post-transcriptional gene silencing in plants, quelling in fungi, and resistance. Virus Genes 25, 45–57.
RNA interference in animals. Proc. Natl. Acad. Sci. USA 97, 11650– Mlotshwa, S., Voinnet, O., Mette, M.F., Matzke, M., Vaucheret, H., Ding,
11654. S.W., Pruss, G., Vance, V.B., 2002b. RNA silencing and the mobile
Foster, T.M., Lough, T.J., Emerson, S.J., Lee, R.H., Bowman, J.L., Forster, silencing signal. Plant Cell 14, S289–301.
R.L., Lucas, W.J., 2002. A surveillance system regulates selective Mourrain, P., Beclin, C., Elmayan, T., Feuerbach, F., Godon, C., Morel,
entry of RNA into the shoot apex. Plant Cell 14, 1497–1508. J.B., Jouette, D., Lacombe, A.M., Nikic, S., Picault, N., Remoue, K.,
Goodwin, J., Chapman, K., Swaney, S., Parks, T.D., Wernsman, E.A., Sanial, M., Vo, T.A., Vaucheret, H., 2000. Arabidopsis SGS2 and
Dougherty, W.G., 1996. Genetic and biochemical dissection of trans- SGS3 genes are required for posttranscriptional gene silencing and
genic RNA-mediated virus resistance. Plant Cell 8, 95–105. natural virus resistance. Cell 101, 533–542.
108 B.M. Roth et al. / Virus Research 102 (2004) 97–108

Napoli, C., Lemieux, C., Jorgensen, R., 1990. Introduction of a chimeric Smith, H.A., Swaney, S.L., Parks, T.D., Wernsman, E.A., Dougherty, W.G.,
chalcone sythase gene into Petunia results in reversible co-suppression 1994. Transgenic plant virus resistance mediated by untranslatable
of homologous gene in trans. Plant Cell 2, 279–289. sense RNAs: expression, regulation, and fate of nonessential RNAs.
Nykanen, A., Haley, B., Zamore, P.D., 2001. ATP requirements and small Plant Cell 6, 1441–1453.
interfering RNA structure in the RNA interference pathway. Cell 107, Tabara, H., Yigit, E., Siomi, H., Mello, C.C., 2002. The dsRNA bind-
309–321. ing protein RDE-4 interacts with RDE-1, DCR-1, and a DExH-box
Palauqui, J.C., Elmayan, T., Pollien, J.M., Vaucheret, H., 1997. Systemic helicase to direct RNAi in C. elegans. Cell 109, 861–871.
acquired silencing: transgene-specific post-transcriptional silencing is Takeda, A., Sugiyama, K., Nagano, H., Mori, M., Kaido, M., Mise, K.,
transmitted by grafting from silenced stocks to non-silenced scions. Tsuda, S., Okuno, T., 2002. Identification of a novel RNA silencing
EMBO J. 16, 4738–4745. suppressor, NSs protein of Tomato spotted wilt virus. FEBS Lett. 532,
Pfeffer, S., Dunoyer, P., Heim, F., Richards, K.E., Jonard, G., Ziegler-Graff, 75–79.
V., 2002. P0 of beet Western yellows virus is a suppressor of post- Thomas, C.L., Leh, V., Lederer, C., Maule, A.J., 2003. Turnip crinkle
transcriptional gene silencing. J. Virol. 76, 6815–6824. virus coat protein mediates suppression of RNA silencing in Nicotiana
Pruss, G., Ge, X., Shi, X.M., Carrington, J.C., Bowman Vance, V., 1997. benthamiana. Virology 306, 33–41.
Plant viral synergism: the potyviral genome encodes a broad-range Tijsterman, M., Ketting, R.F., Plasterk, R.H., 2002. The genetics of RNA
pathogenicity enhancer that transactivates replication of heterologous silencing. Annu. Rev. Genet. 36, 489–519.
viruses. Plant Cell 9, 859–868. van der Krol, A.R., Mur, L.A., Beld, M., Mol, J.N., Stuitje, A.R., 1990.
Qiu, W., Park, J.W., Scholthof, H.B., 2002. Tombusvirus P19-mediated Flavonoid genes in petunia: addition of a limited number of gene
suppression of virus-induced gene silencing is controlled by genetic copies may lead to a suppression of gene expression. Plant Cell 2,
and dosage features that influence pathogenicity. Mol. Plant Microbe 291–299.
Interact. 15, 269–280. van Wezel, R., Dong, X., Liu, H., Tien, P., Stanley, J., Hong, Y.,
Qu, F., Morris, T.J., 2002. Efficient infection of Nicotiana benthamiana 2002. Mutation of three cysteine residues in Tomato yellow leaf curl
by Tomato bushy stunt virus is facilitated by the coat protein and virus—China C2 protein causes dysfunction in pathogenesis and post-
maintained by p19 through suppression of gene silencing. Mol. Plant transcriptional gene silencing suppression. Mol. Plant Microbe Inter-
Microbe Interact. 15, 193–202. act. 15, 203–208.
Qu, F., Ren, T., Morris, T.J., 2003. The coat protein of turnip crinkle virus Vance, V., Vaucheret, H., 2001. RNA silencing in plants—defense and
suppresses posttranscriptional gene silencing at an early initiation step. counterdefense. Science 292, 2277–2280.
J. Virol. 77, 511–522. Vance, V.B., Berger, P.H., Carrington, J.C., Hunt, A.G., Shi, X.M., 1995.
Reed, J.C., Kasschau, K.D., Prokhnevsky, A.I., Gopinath, K., Pogue, G.P., 5 proximal potyviral sequences mediate potato virus X/potyviral syn-
Carrington, J.C., Dolja, V.V., 2003. Suppressor of RNA silencing ergistic disease in transgenic tobacco. Virology 206, 583–590.
encoded by Beet yellows virus. Virology 306, 203–209. Voinnet, O., Baulcombe, D.C., 1997. Systemic signalling in gene silencing.
Ruiz, M.T., Voinnet, O., Baulcombe, D.C., 1998. Initiation and main- Nature 389, 553.
tenance of virus-induced gene silencing. Plant Cell 10, 937– Voinnet, O., Lederer, C., Baulcombe, D.C., 2000. A viral movement
946. protein prevents spread of the gene silencing signal in Nicotiana
Schwartz, M., Chen, J., Janda, M., Sullivan, M., den Boon, J., Ahlquist, benthamiana. Cell 103, 157–167.
P., 2002. A positive-strand RNA virus replication complex paral- Voinnet, O., Pinto, Y.M., Baulcombe, D.C., 1999. Suppression of gene
lels form and function of retrovirus capsids. Mol. Cell 9, 505– silencing: a general strategy used by diverse DNA and RNA viruses
514. of plants. Proc. Natl. Acad. Sci. USA 96, 14147–14152.
Shi, X.M., Miller, H., Verchot, J., Carrington, J.C., Vance, V.B., 1997. Voinnet, O., Rivas, S., Mestre, P., Baulcombe, D., 2003. An enhanced
Mutations in the region encoding the central domain of helper transient expression system in plants based on suppression of gene
component-proteinase (HC-Pro) eliminate potato virus X/potyviral silencing by the p19 protein of tomato bushy stunt virus. Plant J. 33,
synergism. Virology 231, 35–42. 949–956.
Silhavy, D., Molnar, A., Lucioli, A., Szittya, G., Hornyik, C., Tavazza, Yelina, N.E., Savenkov, E.I., Solovyev, A.G., Morozov, S.Y., Valkonen,
M., Burgyan, J., 2002. A viral protein suppresses RNA silencing J.P., 2002. Long-distance movement, virulence, and RNA silencing
and binds silencing-generated, 21- to 25-nucleotide double-stranded suppression controlled by a single protein in hordei- and potyviruses:
RNAs. EMBO J. 21, 3070–3080. complementary functions between virus families. J. Virol. 76, 12981–
Smith, C.J., Watson, C.F., Bird, C.R., Ray, J., Schuch, W., Grierson, 12991.
D., 1990. Expression of a truncated tomato polygalacturonase gene Zamore, P.D., Tuschl, T., Sharp, P.A., Bartel, D.P., 2000. RNAi:
inhibits expression of the endogenous gene in transgenic plants. Mol. double-stranded RNA directs the ATP-dependent cleavage of mRNA
Gen. Genet. 224, 477–481. at 21 to 23 nucleotide intervals. Cell 101, 25–33.
Virus Research 102 (2004) 109–115

RNA silencing: a conserved antiviral immunity of plants and animals


Shou-Wei Ding a,b,∗ , Hongwei Li a , Rui Lu a , Feng Li b , Wan-Xiang Li a
a Center for Plant Cell Biology, Department of Plant Pathology, Riverside, CA 92521, USA
b Microbiology Program, University of California, Riverside, CA 92521, USA

Abstract

RNA silencing is a novel RNA-guided gene regulatory mechanism operational in a wide range of eukaryotic organisms from fission yeast,
plants, to mammals. This article reviews the recent progress on aspects of RNA silencing that are related to its biological function as a conserved
antiviral immunity of plants and animals, and highlights features of this novel antiviral response in invertebrate animals as compared to the
known innate and adaptive immunities. Finally, we discuss evidence that suggests a natural antiviral role for RNA silencing in vertebrates as
well as experimental approaches that may facilitate the identification of first mammalian viral suppressors of RNA silencing.
© 2004 Elsevier B.V. All rights reserved.

Keywords: RNA silencing; RNAi; Antiviral immunity; Drosophila

1. Introduction Martinez et al., 2002), siRNAs act as guides to select mRNA


targets for degradation by siRNA/mRNA base-pairing. A
RNA silencing is a generic term describing related RNA- fascinating feature of RNA silencing is its ability to spread
guided gene regulatory mechanisms that include post- both cell-to-cell and long distantly to cause systemic RNA
transcriptional gene silencing (PTGS) in plants, quelling silencing in whole organisms via a sequence-specific si-
in fungi, and RNA interference (RNAi) in animals lencing signal produced following the induction of RNA
(Baulcombe, 1999; Ding, 2000; Vance and Vaucheret, silencing in single cells (Mlotshwa et al., 2002; Palauqui
2001). The first examples of RNA silencing were reported et al., 1997; Voinnet and Baulcombe, 1997).
in transgenic plants carrying artificially introduced trans- RNA silencing and related pathways are involved not only
genes (Matzke et al., 1989; Napoli et al., 1990; Van der in the response to viruses and transposable elements, but also
Krol et al., 1990). Similar RNA silencing phenomena have in gene regulation and heterochromatin formation (Denli and
subsequently been described in a wide range of eukaryotic Hannon, 2003). This short review covers recent progress on
organisms such as fungi, worms, flies, and mammals (Denli RNA silencing as a natural antiviral immunity in plants, in
and Hannon, 2003; Fire et al., 1998). invertebrate and vertebrate animals.
Small interfering RNAs (siRNAs) are considered as the
central component of RNA silencing in all of the silencing
systems characterized to date (Denli and Hannon, 2003; 2. RNA silencing is a natural antiviral response
Hamilton and Baulcombe, 1999). siRNAs are processed in plants
from double-stranded RNA (dsRNA) precursors by the
type III endoribonuclease Dicer (Hammond et al., 2000; Three main lines of evidence support a naturally antiviral
Zamore et al., 2000), which also recognizes fold-back, role for RNA silencing in plants. Firstly, virus infection trig-
imperfectly base-paired single-stranded RNA (ssRNA) sub- gers RNA silencing in infected plants that specifically targets
strates (Hutvagner et al., 2001). After incorporation into the the viral and homologous RNAs for degradation. This is best
RNA-induced silencing complex (RISC), which contains at illustrated by the detection of virus-specific siRNAs of both
least one Argonaute (AGO) protein (Hammond et al., 2001; sense and antisense polarities in wild type plants infected
with plus-strand RNA viruses (Hamilton and Baulcombe,
1999). Presumably, silencing of viral RNAs is initiated by
∗ Corresponding author. Dicer cleavage of the viral dsRNA replicative intermediates,
E-mail address: shou-wei.ding@ucr.edu (S.-W. Ding). which predicts that the host has the potential to recognize

0168-1702/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.virusres.2004.01.021
110 S.-W. Ding et al. / Virus Research 102 (2004) 109–115

and target the entire length of a replicating viral RNA. How- prevents the spread of RNA silencing into tissues that do not
ever, the viral siRNA population may also contain siRNAs contain the initial silencing trigger, but it does not inhibit the
derived from Dicer cleavage of either highly structured ss- initiation of intracellular silencing (Guo and Ding, 2002).
RNAs (Szittya et al., 2002) or dsRNAs generated by the ac- Thus, while a portion of CMV RNAs is constantly destroyed
tion of a cellular RNA-dependent RNA polymerase (RdRP) in infected cells, uninfected tissues will remain suscepti-
(Sijen et al., 2001; Vaistij et al., 2002). The specific cleav- ble due to lack of import of the antiviral silencing signal.
age of ssRNA targets guided by viral siRNAs is consistent In contrast, the potyviral HC-Pro is a potent suppressor of
with a number of key observations that provided the first intracellular silencing, but silencing signal is continuously
clues for the antiviral role of RNA silencing. For example, being produced and exported out of the HC-Pro-expressing
the observed induction of RNA silencing of a tobacco etch cells (Mallory et al., 2001) and as a result, may reduce the
virus (TEV) transgene in tobacco plants by TEV infection rate of virus spread in infected plants. Thus, mixed viral in-
(Lindbo et al., 1993) could be initiated by antisense TEV fections will induce synergistic diseases when suppressors
siRNAs produced in the infected plants in response to the carried by unrelated viruses target distinct steps in the si-
replicating TEV RNA. Similarly, this would also explain lencing pathway or when a related virus provides a more
why transgenes encoding a replication-competent viral RNA efficient suppression of RNA silencing at the same step. No-
genome are the more consistent inducers of RNA silenc- tably, most of the plant viruses contain a compact genome
ing than simple sense transgenes (Angell and Baulcombe, shorter than 10 kilobases encoding on the average four–five
1997) and why non-virus-derived transgenes and endoge- proteins. Therefore, the fact that silencing suppression re-
nous genes can be specifically silenced following infection mains as a conserved genomic function of plant viruses is
with a recombinant RNA or DNA virus carrying a cDNA also indicative of RNA silencing as being a major antiviral
fragment of the corresponding gene (Kjemtrup et al., 1998; response of plants.
Kumagai et al., 1995; Ruiz et al., 1998). The latter tech- The third support came from the observation that host
nique, known as virus-induced gene silencing (VIGS), has plants compromised in RNA silencing exhibit enhanced
been developed into a high throughput platform technology susceptibility to virus infection. In this respect, SGS2/SDE1,
for functional genomics studies in several host species (Lu SGS3, SDE3 and AGO1 have been identified in Arabidop-
et al., 2003). sis thaliana and are essential for transgene-induced RNA
The second and perhaps the strongest support for a silencing, possibly by contributing to production of dsRNA
naturally antiviral role of RNA silencing in plants is the (Vance and Vaucheret, 2001). Intriguingly, A. thaliana mu-
demonstration that plant viruses encode proteins capable of tants defective in either of these genes were all hypersensi-
suppressing RNA silencing (Li and Ding, 2001; Roth et al., tive to CMV, but were as susceptible as the wild type plants
this issue). The first viral suppressors of RNA silencing to the infection of at least five other viruses tested (Boutet
reported were HC-Pro and the 2b protein encoded by po- et al., 2003; Dalmay et al., 2000, 2001; Morel et al., 2002;
tyviruses and cucumoviruses, respectively (Anandalakshmi Mourrain et al., 2000). In contrast, tobacco and A. thaliana
et al., 1998; Brigneti et al., 1998; Kasschau and Carrington, plants deficient in a gene related to SGS2/SDE1, which,
1998; Li et al., 1999), both of which were previously identi- unlike SGS2/SDE1, is induced by salicylic acid and virus
fied as determinants of synergistic viral diseases (Ding et al., infection, were more susceptible to both tobacco mosaic
1996; Pruss et al., 1997). Many plant viruses have since virus and PVX (Xie et al., 2001; Yu et al., 2003). It will
been found to encode silencing suppression activity, includ- be informative to determine if infection with virus mutants
ing a DNA virus and potato virus X (PVX) that did not that lack a functional suppressor of RNA silencing could
exhibit any suppressor activity in a ‘reversal of silencing’ be complemented specifically in any of those host mutants
assay, even though suppressors encoded by viruses from compromised in RNA silencing.
distinct taxa may share little sequence homology (Li et al.,
1999; Li and Ding, 2001; Voinnet et al., 1999, 2000). It
would not be unexpected if all plant viruses are found to 3. Animal viral proteins that suppress RNA
encode at least one suppressor of RNA silencing, provided silencing in plants
each of the viral proteins is assayed for possible suppression
of both intra- and inter-cellular RNA silencing. Silencing Detection of silencing suppressor activity for the flock
suppression clearly plays a role in establishing virus infec- house virus (FHV) B2 protein, using a silencing assay es-
tions as most of these suppressors were previously shown tablished in plants, provided the first indication that RNA
essential for systemic virus spread in their hosts. silencing might be antiviral in animals (Li et al., 2002). The
In contrast to the specificity involved in the selection animal nodaviral B2 gene shares several features with the
of silencing targets, viral suppression of RNA silencing is plant cucumoviral 2b gene (Ding et al., 1995), known to en-
broad-spectrum, suggesting that they target a core RNA si- code a suppressor of RNA silencing (Brigneti et al., 1998;
lencing pathway. It is important to note that silencing sup- Li et al., 1999). Both genes overlap the C-terminus, and
pression by all known viral proteins is partial or incomplete. occupy the +1 reading frame, of the viral RdRP gene and
For example, the cucumber mosaic virus (CMV) 2b protein are translated in vivo from a subgenomic RNA, though their
S.-W. Ding et al. / Virus Research 102 (2004) 109–115 111

Fig. 1. Open reading frames (ORFs) encoded by CMV RNA 2 and FHV RNA 1. The maps show the location of both start (arrow) and stop (triangle)
codons in each reading frame represented by, respectively, short and long vertical lines. The ORFs encoding the viral RdRP are called 2a in CMV and
A in FHV, respectively. The start and stop codons for proteins B2 and 2b are indicated. The location of the conserved GDD codons of the RdRP of
CMV (nt 1908–1916) and FHV (nt 2110–2118) is also shown.

encoded proteins do not exhibit any detectable sequence pathway was made partially defective by depleting AGO2,
similarities (Fig. 1). Knockout of either B2 in FHV or 2b in an essential component of RISC (Hammond et al., 2001).
CMV resulted in a defect in virus spreading, but neither is These findings indicate that recognition and targeting of the
required for viral RNA replication (Ball, 1995; Ding et al., replicating FHV RNAs for degradation by the RNAi path-
1995). In Nicotiana benthamiana plants, B2 suppression of way represents a natural response of the wild type fly cells
transgene-induced RNA silencing was as potent as the 2b to virus infection. Further mutagenesis analysis showed an
protein encoded by tomato aspermy cucumovirus, which is essential role of B2 in FHV infection since a B2-deletion
among the strongest plant viral suppressors known (Li et al., mutant of FHV did not accumulate to a detectable level in
2002). By comparison, the 2b of CMV was a much weaker wild type fly cells. However, an abundant accumulation of
suppressor that delayed, but did not prevent the initiation the B2-knockout mutant was detected in the fly cells that
of RNA silencing (Guo and Ding, 2002). Significantly, B2 either carried a B2-expressing plasmid or were depleted for
was able to functionally substitute for 2b of CMV in whole AGO2. Thus, the observed essential role of B2 is to sup-
plant infections (Li et al., 2002). press the RNA silencing antiviral response, rather than being
A recent work found that the mammalian reovirus ␴3 pro- required for viral replication, which is in agreement with a
tein also suppresses RNA silencing in the same Agrobac- previous work carried out in mammalian cells (Ball, 1995).
terium co-infiltration assay established in N. benthamiana Recent work showed that B2 is also a suppressor of RNAi
plants carrying a highly expressed green fluorescent protein since specific degradation of a GFP mRNA targeted by an
(GFP) transgene (Lichner et al., 2003). ␴3 is a virion outer introduced GFP dsRNA was inhibited in the B2-expressing
shell protein that binds dsRNA (Miller and Samuel, 1992), fly cells (Li et al., 2004).
suggesting a possible link between dsRNA binding and si- Similar to fruit flies, it was found recently that infection of
lencing suppression (Lichner et al., 2003). mosquito cells with a HIV-related nodavirus also triggered
an AGO2-dependent antiviral immunity that was sensitive
to suppression of either the homologous or heterologous
4. RNA silencing as an adaptive antiviral nodaviral B2 (Li et al., 2004).
immunity in animals Therefore, nodavirus infection of invertebrate animal
cells triggers RNA silencing that specifically targets the
Direct experimental evidence supporting an antiviral role invading viral RNA, and nodaviruses encode a B2 protein
of RNA silencing in the animal kingdom has recently been that is able to suppress PTGS in plants and RNAi in fruit
documented in Drosophila (Li et al., 2002). In cultured flies. Significantly, B2 is essential for nodavirus infection of
Drosophila S2 cells, infection with FHV led to the accu- wild type animal cells, but is dispensable in cells depleted
mulation of the 22-nt, FHV-specific siRNAs in both the for a RISC component, AGO2, indicating that nodavirus
sense and antisense polarities. Virus-specific siRNAs were RNAs are targeted for degradation by the RISC-mediated
also detected in the silkmoth (Bombyx mori) larvae infected RNA silencing antiviral response. These data establish RNA
with Sindbis virus (Uhlirova et al., 2003). FHV RNAs ac- silencing as a natural antiviral mechanism in invertebrate
cumulated to higher levels in the fly cells after the RNAi animals.
112 S.-W. Ding et al. / Virus Research 102 (2004) 109–115

5. Features of the RNA silencing antiviral response in marily infected cells, whereas it may be essential for animal
invertebrate animal cells viral suppressors to inhibit the intracellular silencing.

The RNA silencing antiviral response of invertebrates


is adaptive, potent and rapid. In contrast to the broad- 6. Is RNA silencing naturally antiviral in vertebrates?
spectrum innate immunity in Drosophila and mosquitoes
(Christophides et al., 2002), the RNA silencing immunity The RNA silencing pathway is also operational in mam-
has features similar to the peptide-based adaptive immunity malian cells since specific mRNA degradation is achieved
(Whitton and Oldstone, 2001). The specificity determinants by introducing exogenous synthetic siRNAs (Caplen et al.,
of the RNA silencing response are siRNAs, which are 2001; Elbashir et al., 2001). Remarkably, the siRNAs intro-
derived and processed from the invading virus. After up- duced into mammalian cells are found in a silencing com-
loaded into RISC, these virus-specific siRNAs selectively plex essentially identical to the RISC initially characterized
recruit target ssRNAs by base-pairing for RISC-mediated in Drosophila (Hammond et al., 2001; Martinez et al., 2002).
cleavage. At whole organism level, RNA silencing may As in fruit flies, the cellular RdRP involved in transitive RNA
also have the capacity to provide a long-term memory, silencing in A. thaliana and C. elegans is not found in either
as has been detected in plants, which, after being re- mouse or human (Denli and Hannon, 2003; Schwarz et al.,
covered from a virulent primary infection, maintain an 2002). Furthermore, the human genome encodes a Dicer that
RNA silencing-mediated resistance to secondary infections recognizes both dsRNA and hairpin ssRNA substrates as the
(Covey et al., 1997; Ratcliff et al., 1997, 1999; Xin and Drosophila Dicer does (Bernstein et al., 2001; Hutvagner
Ding, 2003). However, while the peptide-based immunity et al., 2001; Provost et al., 2002; Zhang et al., 2002).
in vertebrates usually takes more than a week to respond Thus, it is conceivable that the mammalian Dicer could
(Whitton and Oldstone, 2001), the RNA silencing response also detect viral dsRNA replicative intermediates and/or
in fruit flies and mosquitoes is rapid and effective in primar- stem-loop structures to initiate RNA silencing in response
ily infected cells, which is similar to the innate immunity. to virus infection of mammalian cells, as has been demon-
In fact, the genome of the B2-knockout mutant of FHV strated in fruit flies (Li et al., 2002). Recent reports have
never accumulated to detectable levels after transfection, elegantly demonstrated potent inhibition of production of
indicating that in the absence of viral suppression, RNA several types of mammalian viruses by synthetic siRNAs
silencing is sufficient to rapidly clear viral RNAs within (Gitlin and Andino, 2003), indicating that viral RNAs are
primarily infected cells (Li et al., 2002). In this regard, it is accessible to an activated RISC in mammalian cells. The
possible that RNA silencing may contribute to, or be part mammalian Dicer could act in an antagonist pathway to the
of, the robust innate immunity occurred in early stages of dsRNA-specific protein kinase (PKR). Alternatively, initi-
viral infections in vertebrates (Parham, 2003). ation of the RNAi antiviral immunity by the mammalian
It is known that common components, such as siRNA, Dicer could occur in specific cell types or at certain de-
Dicer and RISC, are involved in PTGS in plants and RNAi velopmental stages in which the non-specific translational
in animals (Tang et al., 2003; Vance and Vaucheret, 2001). shutdown response mediated by PKR is inactive (Wianny
Suppression of the two processes in plants and Drosophila and Zernicka-Goetz, 2000; Svoboda et al., 2000; Billy et al.,
by both the animal viral B2 protein and the plant viral p19 2001).
provides direct experimental evidence for a conserved RNA There is evidence suggesting that the RNAi machinery
silencing pathway in the plant and animal kingdoms (Voinnet in mammalian cells is capable of detecting the replicating
et al., 1999; Li et al., 2002; Lindenbach and Rice, 2002; Li viral RNAs. For example, FHV also replicates and accu-
et al., 2004). However, the cellular RdRP is found only in mulates in mammalian cells. Although B2 was not essen-
plants, fungi and worms, but not in fruit flies, mouse and tial for FHV replication, B2-deficient FHV production in
human. Furthermore, it appears that at the single cell level, mammalian cells impaired infectivity of viral progeny (Ball,
the RNA silencing response targeting a replicating virus in 1995), suggesting that the newly identified activity of B2 in
plants is not as potent as in fruit flies and mosquitoes. For silencing suppression may play a role in FHV infection of
example, plant virus mutants that lack a functional suppres- mammalian cells. Similarly, in reovirus-infected mammalian
sor have defects in spreading cell-to-cell or systemically in cells, the expression of ␴3 might lead to inactivation of the
whole plant infections, but they exhibit no obvious defect RNA silencing antiviral response in addition to inhibition of
in isolated single cells (protoplasts) (Li and Ding, 2001; PKR-mediated responses (Lichner et al., 2003; Miller and
Silhavy et al., 2002). This is in contrast to an essential role Samuel, 1992).
for B2 suppression in FHV infection of cultured fly cells (Li Future work will be necessary to determine if virus
et al., 2002), suggesting that RISC as a multiple turnover infection indeed elicits the RNA silencing antiviral re-
enzymatic complex may be more efficient in organisms that sponse in mammalian cells. This may include detection of
do not carry a cellular RdRP. These results also suggest that virus-specific siRNAs, a homology-dependent RNA degra-
plant viral suppressors may mainly interfere with the spread dation, or viral suppression of RNA silencing in infected
of the RNA silencing antiviral response away from the pri- mammalian cells. The in planta silencing assay will be
S.-W. Ding et al. / Virus Research 102 (2004) 109–115 113

useful in identifying novel animal viral suppressors of Boutet, S., Vazquez, F., Liu, J., Beclin, C., Fagard, M., Gratias, A., Morel,
RNA silencing. However, it will be limited to those sup- J.B., Crete, P., Chen, X., Vaucheret, H., 2003. Arabidopsis HEN1:
a genetic link between endogenous miRNA controlling development
pressors that target the elements conserved between the and siRNA controlling transgene silencing and virus resistance. Curr.
RdRP-dependent silencing pathway in plants and the Biol. 13, 843–848.
RdRP-independent silencing pathway in animals. As a re- Brigneti, G., Voinnet, O., Li, W.X., Ji, L.H., Ding, S.W., Baulcombe, D.C.,
sult, a suppressor of RNA silencing identified in plant assays 1998. Viral pathogenicity determinants are suppressors of transgene
may not necessarily suppress RNA silencing in animal cells silencing in Nicotiana benthamiana. EMBO J. 17, 6739–6746.
Caplen, N.J., Parrish, S., Imani, F., Fire, A., Morgan, R.A., 2001. Spe-
and vice versa. Therefore, it will be important to establish cific inhibition of gene expression by small double-stranded RNAs in
an RNA silencing system in mammalian cells suitable for invertebrate and vertebrate systems. Proc. Natl. Acad. Sci. U.S.A. 98,
assaying viral suppression of RNA silencing. It is of interest 9742–9747.
to consider that most, if not all, of the characterized plant Christophides, G.K., Zdobnov, E., Barillas-Mury, C., Birney, E., Blandin,
viral suppressors target steps upstream of siRNAs or RISC S., Blass, C., Brey, P.T., Collins, F.H., Danielli, A., Dimopoulos, G.,
Hetru, C., Hoa, N.T., Hoffmann, J.A., Kanzok, S.M., Letunic, I.,
(Li and Ding, 2001; Roth et al., this issue). Identification of Levashina, E.A., Loukeris, T.G., Lycett, G., Meister, S., Michel, K.,
similar animal viral suppressors will likely require a silenc- Moita, L.F., Muller, H.M., Osta, M.A., Paskewitz, S.M., Reichhart,
ing system that initiates early in the silencing pathway and J.M., Rzhetsky, A., Troxler, L., Vernick, K.D., Vlachou, D., Volz,
is not initiated by siRNAs. Thus, clearance of virus RNAs J., von Mering, C., Xu, J., Zheng, L., Bork, P., Kafatos, F.C., 2002.
in infected cells by introduced siRNAs (Dector et al., 2002; Immunity-related genes and gene families in Anopheles gambiae.
Science 298, 159–165.
Gitlin et al., 2002; Randall et al., 2003) may not necessarily Covey, S.N.A.-K., N.S., Langara, A., Turner, D.S., 1997. Plants combat
argue against the possibility that these mammalian viruses infection by gene silencing. Nature 385, 781–782.
carry a suppressor of the RNA silencing antiviral immunity. Dalmay, T., Hamilton, A., Rudd, S., Angell, S., Baulcombe, D.C., 2000.
Given the closer similarity of the RNA silencing pathway An RNA-dependent RNA polymerase gene in Arabidopsis is required
between Drosophila and humans, the nodavirus RNA si- for posttranscriptional gene silencing mediated by a transgene but not
by a virus. Cell 101, 543–553.
lencing system established in cultured fruit fly cells (Li
Dalmay, T., Horsefield, R., Braunstein, T.H., Baulcombe, D.C., 2001.
et al., 2002) may provide a valuable assay for the identifica- SDE3 encodes an RNA helicase required for post-transcriptional gene
tion of mammalian viral suppressors. Indeed, NS1 encoded silencing in Arabidopsis. EMBO J. 20, 2069–2078.
by influenza A/B/C viruses and E3L by vaccinia virus were Dector, M.A., Romero, P., Lopez, S., Arias, C.F., 2002. Rotavirus gene
recently identified as suppressors of the nodavirus-induced silencing by small interfering RNAs. EMBO Rep. 3, 1175–1180.
Denli, A.M., Hannon, G.J., 2003. RNAi: an ever-growing puzzle. Trends
RNA silencing in Drosophila cells, implicating RNA si-
Biochem. Sci. 28, 196–201.
lencing as an RNA-based antiviral immunity in mammalian Ding, S.W., 2000. RNA silencing. Curr. Opin. Biotechnol. 11, 152–156.
cells (Li et al., 2004). Ding, S.W., Li, W.X., Symons, R.H., 1995. A novel naturally-occurring
hybrid gene encoded by a plant RNA virus facilitates long-distance
virus movement. EMBO J. 14, 5762–5772.
Ding, S.W., Shi, B.J., Li, W.X., Symons, R.H., 1996. An interspecies
Acknowledgements
hybrid RNA virus is significantly more virulent than either parental
virus. Proc. Natl. Acad. Sci. U.S.A. 93, 7470–7474.
The research projects on the RNA silencing antiviral re- Elbashir, S.M., Lendeckel, W., Tuschl, T., 2001. RNA interference is
sponse in authors’ lab are supported by grants from NIH, mediated by 21-and 22-nucleotide RNAs. Genes. Dev. 15, 188–
USDA, CRB, CDFA and UC-BIOSTAR. The authors also 200.
Fire, A., Xu, S.Q., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello,
wish to thank Karla Kirkegaard for discussion.
C.C., 1998. Potent and specific genetic interference by double-stranded
RNA in Caenorhabditis elegans. Nature 391, 806–811.
Gitlin, L., Andino, R., 2003. Nucleic acid-based immune system: the
References antiviral potential of mammalian RNA silencing. J. Virol. 77, 7159–
7165.
Anandalakshmi, R., Pruss, G.J., Ge, X., Marathe, R., Mallory, A.C., Gitlin, L., Karelsky, S., Andino, R., 2002. Short interfering RNA confers
Smith, T.H., Vance, V.B., 1998. A viral suppressor of gene silencing intracellular antiviral immunity in human cells. Nature 418, 430–
in plants. Proc. Natl. Acad. Sci. U.S.A. 95, 13079–13084. 434.
Angell, S.M., Baulcombe, D.C., 1997. Consistent gene silencing in trans- Guo, H.S., Ding, S.W., 2002. A viral protein inhibits the long range
genic plants expressing a replicating potato virus X RNA. EMBO J. signaling activity of the gene silencing signal. EMBO J. 21, 398–407.
16, 3675–3684. Hamilton, A.J., Baulcombe, D.C., 1999. A species of small antisense RNA
Ball, L.A., 1995. Requirements for the self-directed replication of flock in posttranscriptional gene silencing in plants. Science 286, 950–952.
house virus RNA 1. J. Virol. 69, 2722. Hammond, S.M., Bernstein, E., Beach, D., Hannon, G.J., 2000. An
Baulcombe, D.C., 1999. Gene silencing: RNA makes RNA makes no RNA-directed nuclease mediates post-transcriptional gene silencing in
protein. Curr. Biol. 9, R599–R601. Drosophila cells. Nature 404, 293–296.
Bernstein, E., Caudy, A.A., Hammond, S.M., Hannon, G.J., 2001. Role Hammond, S.M., Boettcher, S., Caudy, A.A., Kobayashi, R., Hannon, G.J.,
for a bidentate ribonuclease in the initiation step of RNA interference. 2001. Argonaute 2, a link between genetic and biochemical analyses
Nature 409, 363–366. of RNAi. Science 293, 1146–1150.
Billy, E., Brondani, V., Zhang, H., Muller, U., Filipowicz, W., Hutvagner, G., McLachlan, J., Pasquinelli, A.E., Balint, E., Tuschl, T.,
2001. Specific interference with gene expression induced by long, Zamore, P.D., 2001. A cellular function for the RNA-interference
double-stranded RNA in mouse embryonal teratocarcinoma cell lines. enzyme Dicer in the maturation of the let-7 small temporal RNA.
Proc. Natl. Acad. Sci. U.S.A. 98, 14428–14433. Science 293, 834–838.
114 S.-W. Ding et al. / Virus Research 102 (2004) 109–115

Kasschau, K.D., Carrington, J.C., 1998. A counterdefensive strategy of Parham, P., 2003. Innate immunity: the unsung heroes. Nature 423, 20.
plant viruses: Suppression of posttranscriptional gene silencing. Cell Provost, P., Dishart, D., Doucet, J., Frendewey, D., Samuelsson, B., Rad-
95, 461–470. mark, O., 2002. Ribonuclease activity and RNA binding of recombi-
Kjemtrup, S., Sampson, K.S., Peele, C.G., Nguyen, L.V., Conkling, M.A., nant human Dicer. EMBO J. 21, 5864–5874.
Thompsosn, W.F., Robertson, D., 1998. Gene silencing from plant Pruss, G., Ge, X., Shi, X.M., Carrington, J.C., Vance, V.B., 1997. Plant vi-
DNA carried by a geminivirus. Plant J. 14, 91–100. ral synergism: the potyviral genome encodes a broad-range pathogenic-
Kumagai, M.H., Donson, J., della-Cioppa, G., Harvey, D., Hanley, K., ity enhancer that transactivates replication of heterologous viruses.
Grill, L.K., 1995. Cytoplasmic inhibition of carotenoid biosynthesis Plant Cell 9, 859–868.
with virus-derived RNA. Proc. Natl. Acad. Sci. U.S.A. 92, 1679–1683. Randall, G., Grakoui, A., Rice, C.M., 2003. Clearance of replicating
Li, H.W., Li, W.X., Ding, S.W., 2002. Induction and suppression of RNA hepatitis C virus replicon RNAs in cell culture by small interfering
silencing by an animal virus. Science 296, 1319–1321. RNAs. Proc. Natl. Acad. Sci. U.S.A. 100, 235–240.
Li, H.W., Lucy, A.P., Guo, H.S., Li, W.X., Ji, L.H., Wong, S.M., Ding, Ratcliff, F., Harrison, B.D., Baulcombe, D.C., 1997. A similarity between
S.W., 1999. Strong host resistance targeted against a viral suppressor viral defense and gene silencing in plants. Science 276, 1558–1560.
of the plant gene silencing defence mechanism. EMBO J. 18, 2683– Ratcliff, F.G., MacFarlane, S.A., Baulcombe, D.C., 1999. Gene silencing
2691. without DNA: RNA-mediated cross-protection between viruses. Plant
Li, W.X., Ding, S.W., 2001. Viral suppressors of RNA silencing. Curr. Cell 11, 1207–1215.
Opin. Biotechnol. 12, 150–154. Roth, B.M., Pruss, G.J., Vance, V.B., 2004. Plant viral suppressors of
Li, W.X., Li, H.W., Lu, R., Li, F., Dus, M., Atkinson, P.W., Brydon, E., RNA silencing. Virus Res., this issue.
Johnson, K.L., Garcia-Sastre, A., Ball, L.A., Palese, P., Ding, S.W., Ruiz, M.T., Voinnet, O., Baulcombe, D.C., 1998. Initiation and mainte-
2004. Interferon antagonist proteins of influenza and vaccinia viruses nance of virus-induced gene silencing. Plant Cell 10, 937–946.
are suppressors of RNA silencing. Proc. Natl. Acad. Sci. USA 101, Schwarz, D.S., Hutvagner, G., Haley, B., Zamore, P.D., 2002. Evidence
1350–1355. that siRNAs function as guides, not primers, in the Drosophila and
Lichner, Z., Silhavy, D., Burgyan, J., 2003. Double-stranded RNA-binding human RNAi pathways. Mol. Cell 10, 537–548.
proteins could suppress RNA interference-mediated antiviral defences. Sijen, T., Fleenor, J., Simmer, F., Thijssen, K.L., Parrish, S., Timmons,
J. Gen. Virol. 84, 975–980. L., Plasterk, R.H.A., Fire, A., 2001. On the role of RNA amplification
Lindbo, J.A., Silva-Rosales, L., Proebsting, W.M., Dougherty, W.G., 1993. in dsRNA-triggered gene silencing. Cell 107, 465–476.
Induction of a highly specific antiviral state in transgenic plants: Silhavy, D., Molnar, A., Lucioli, A., Szittya, G., Hornyik, C., Tavazza,
implications for regulation of gene expression and virus resistance. M., Burgyan, J., 2002. A viral protein suppresses RNA silencing
Plant Cell 5, 1749–1759. and binds silencing-generated, 21- to 25-nucleotide double-stranded
Lindenbach, B.D., Rice, C.M., 2002. RNAi targeting an animal virus: RNAs. EMBO J. 21, 3070–3080.
news from the front. Mol. Cell 9, 925–927. Svoboda, P., Stein, P., Hayashi, H., Schultz, R.M., 2000. Selective reduc-
Lu, R., Martin-Hernandez, A.M., Peart, J.R., Malcuit, I., Baulcombe, D.C., tion of dormant maternal mRNAs in mouse oocytes by RNA interfer-
2003. Virus-induced gene silencing in plants. Methods 30, 296–303. ence. Development 127, 4147–4156.
Mallory, A.C., Ely, L., Smith, T.H., Marathe, R., Anandalakshmi, R., Szittya, G., Molnar, A., Silhavy, D., Hornyik, C., Burgyan, J., 2002.
Fagard, M., Vaucheret, H., Pruss, G., Bowman, L., Vance, V.B., 2001. Short defective interfering RNAs of tombusviruses are not targeted but
HC-Pro suppression of transgene silencing eliminates the small RNAs trigger post-transcriptional gene silencing against their helper virus.
but not transgene methylation or the mobile signal. Plant Cell 13, Plant Cell 14, 359–372.
571–583. Tang, G., Reinhart, B.J., Bartel, D.P., Zamore, P.D., 2003. A biochemical
Martinez, J., Patkaniowska, A., Urlaub, H., Luhrmann, R., Tuschl, T., framework for RNA silencing in plants. Genes Dev. 17, 49–63.
2002. Single-stranded antisense siRNAs guide target RNA cleavage Uhlirova, M., Foy, B.D., Beaty, B.J., Olson, K.E., Riddiford, L.M., Jindra,
in RNAi. Cell 110, 563–574. M., 2003. Use of Sindbis virus-mediated RNA interference to demon-
Matzke, M.A., Primig, M., Trnovsky, J., Matzke, A.J.M., 1989. Reversible strate a conserved role of Broad-Complex in insect metamorphosis.
methylation and inactivation of marker genes in sequentially trans- Proc. Natl. Acad. Sci. USA 100, 15607–15612.
formed tobacco plants. EMBO J. 8, 643–649. Vaistij, F.E., Jones, L., Baulcombe, D.C., 2002. Spreading of RNA tar-
Miller, J.E., Samuel, T., 1992. Proteolytic cleavage of the reovirus sigma geting and DNA methylation in RNA silencing requires transcription
3 protein results in enhanced double-stranded RNA-binding activ- of the target gene and a putative RNA-dependent RNA polymerase.
ity: identification of a repeated basic amino acid motif within the Plant Cell 14, 857–867.
C-terminal binding region. J. Virol. 66, 5347–5356. Van der Krol, A.R., Mur, L.A., Beld, M., Mol, J.N., Stuitje, A.R., 1990.
Mlotshwa, S., Voinnet, O., Mette, M.F., Matzke, M., Vaucheret, H., Ding, Flavonoid genes in petunia: addition of a limited number of gene
S.W., Pruss, G., Vance, V.B., 2002. RNA silencing and the mobile copies may lead to a suppression of gene expression. Plant Cell 2,
silencing signal. Plant Cell 14, S289–S301. 291–299.
Morel, J.B., Godon, C., Mourrain, P., Beclin, C., Boutet, S., Feuerbach, F., Vance, V., Vaucheret, H., 2001. RNA silencing in plants—defense and
Proux, F., Vaucheret, H., 2002. Fertile hypomorphic ARGONAUTE counterdefense. Science 292, 2277–2280.
(ago1) mutants impaired in post-transcriptional gene silencing and Voinnet, O., Baulcombe, D.C., 1997. Systemic signalling in gene silencing.
virus resistance. Plant Cell 14, 629–639. Nature 389, 553.
Mourrain, P., Beclin, C., Elmayan, T., Feuerbach, F., Godon, C., Morel, Voinnet, O., Lederer, C., Baulcombe, D.C., 2000. A viral movement
J.B., Jouette, D., Lacombe, A.M., Nikic, S., Picault, N., Remoue, protein prevents spread of the gene silencing signal in Nicotiana
K., Sanial, M., Vo, T.A., Vaucheret, H., 2000. Arabidopsis SGS2 and benthamiana. Cell 103, 157–167.
SGS3 genes are required for posttranscriptional gene silencing and Voinnet, O., Pinto, Y.M., Baulcombe, D.C., 1999. Suppression of gene
natural virus resistance. Cell 101, 533–542. silencing: a general strategy used by diverse DNA and RNA viruses
Napoli, C., Lemieux, C., Jorgensen, R., 1990. Introduction of a of plants. Proc. Natl. Acad. Sci. U.S.A. 96, 14147–14152.
chimeric chalcone synthase gene into Petunia results in reversible Whitton, J.L., Oldstone, B.A., 2001. The immune responses to viruses. In:
co-suppression of homologous genes in trans. Plant Cell 2, 279–289. Knipe, D.M., Howley, P.M. (Eds.), Fields Virology, 4th ed. Lippincott
Palauqui, J.C., Elmayan, T., Pollien, J.M., Vaucheret, H., 1997. Systemic Williams & Wilkins, Philadephia, pp. 285–320.
acquired silencing: transgene-specific post-transcriptional silencing is Wianny, F., Zernicka-Goetz, M., 2000. Specific interference with gene
transmitted by grafting from silenced stocks to non-silenced scions. function by double-stranded RNA in early mouse development. Nat.
EMBO J. 16, 4738–4745. Cell Biol. 2, 70–75.
S.-W. Ding et al. / Virus Research 102 (2004) 109–115 115

Xie, Z., Fan, B., Chen, C., Chen, Z., 2001. An important role of an merase in antiviral defense. Mol. Plant Microb. Interact. 16, 206–
inducible RNA-dependent RNA polymerase in plant antiviral defense. 216.
Proc. Natl. Acad. Sci. U.S.A. 98, 6516–6521. Zamore, P.D., Tuschl, T., Sharp, P.A., Bartel, D.P., 2000. RNAi:
Xin, H.W., Ding, S.W., 2003. Identification and molecular characterization Double-stranded RNA directs the ATP-dependent cleavage of mRNA
of a naturally occurring RNA virus mutant defective in the initiation at 21–23 nucleotide intervals. Cell 101, 25–33.
of host recovery. Virology 317, 253–262. Zhang, H., Kolb, F.A., Brondani, V., Billy, E., Filipowicz, W., 2002.
Yu, D., Fan, B., MacFarlane, S.A., Chen, Z., 2003. Analysis of the in- Human Dicer preferentially cleaves dsRNAs at their termini without
volvement of an inducible Arabidopsis RNA-dependent RNA poly- a requirement for ATP. EMBO J. 21, 5875–5885.
Virus Research 102 (2004) 117–124

Generation of an shRNAi expression library against


the whole human transcripts
Makoto Miyagishi a,b , Sahohime Matsumoto a,b,c , Kazunari Taira a,b,∗
aDepartment of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Hongo, Tokyo 113-8656, Japan
b Gene Function Research Center, National Institute of Advanced Industrial Science and Technology (AIST),

Central 4, 1-1-1 Higashi, Tsukuba Science City 305-8562, Japan


c Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo 113-0033, Japan

Abstract

RNA interference is a phenomenon in which expression of an individual gene can be specifically silenced by introducing a double-stranded
RNA, homologous to the gene, and is receiving attention as a powerful tool for reverse genetics in the post-genome era. Throughout our
current research to generate an siRNA expression library for the whole human genome, we face many technical difficulties. We present here the
strategies for overcoming some of the difficulties, including the development of genetically stable and highly active siRNA expression vectors,
the selection procedure of the favorable target sites, and the efficient and low cost procedure for constructing an siRNA expression library.
Furthermore, we demonstrate that the screening using the constructed siRNA-expression library indeed works, by evaluating siRNA-expression
library against apoptosis-related genes.
© 2004 Published by Elsevier B.V.

Keywords: RNA interference; U6 promoter; Algorithm; RNAi library

1. Introduction co-workers demonstrated that 21- or 22-nt RNAs with two


nt 3 -overhangs (siRNA) can induce gene silencing without
Recent advances in the Human Genome Project and avail- the nonspecific inhibition of the gene expression in cultured
ability of the complete genome sequence have clarified the mammalian cells (Caplen et al., 2001; Elbashir et al., 2001).
existence of numerous genes whose functions are unknown. Furthermore, in the past year, various groups, including our
Together with the hybrid ribozyme technology (Warashina own, have developed systems for vector-mediated specific
et al., 2001; Kawasaki and Taira, 2002; Kawasaki et al., RNAi in mammalian cells (Brummelkamp et al., 2002;
2002; Suyama et al., 2003a,b), RNA interference (RNAi) is Kawasaki and Taira, 2003; Lee et al., 2002; Miyagishi and
receiving attention as the technology that could unveil their Taira, 2002; Paddison et al., 2002; Sui et al., 2002; Paul
functions (McManus and Sharp, 2002). et al., 2002; Yu et al., 2002; reviewed in McManus and
RNAi is an evolutionarily conserved process in plants and Sharp, 2002; Tuschl, 2002). These vector systems use a pol
animals by which double-stranded RNA (dsRNA) induces III promoter, such as the U6, H1, and tRNA promoters, and
sequence specific degradation of homologous RNA (Fire the systems have been classified into two groups depend-
et al., 1998; reviewed in Zamore, 2001). Although RNAi ing on whether the expressed RNAs are tandem-type or
works well in various organisms, the silencing of specific hairpin-type.
genes by RNAi has proven difficult to detect in mammalian Since gene silencing by siRNA is highly specific and has
systems, because of the dsRNA-dependent nonspecific in- extremely high activity, genome-wide and comprehensive
hibition of protein synthesis by the PKR pathway and the gene analysis in various organism should be expected. Re-
nonspecific RNA degradation by activation of RNase L cently, in Caenorhabditis elegans, a systematic functional
(Elbashir et al., 2001). However, recent works by Tuschl and analysis of a number of genes by RNAi has been per-
formed, and many of these genes were identified as genes
∗ Corresponding author. Tel.: +81-3-5841-8828/298-61-3015; that show mutant phenotype (Fraser et al., 2000; Gonczy
fax: +81-3-5841-7340/8828. et al., 2000). In the near future, it is predicted that a similar
E-mail address: taira@chembio.t.u-tokyo.ac.jp (K. Taira). genome-wide RNAi library approach could be extended

0168-1702/$ – see front matter © 2004 Published by Elsevier B.V.


doi:10.1016/j.virusres.2004.01.022
118 M. Miyagishi et al. / Virus Research 102 (2004) 117–124

to mammalian cells by an siRNA oligonucleotide library Tandem type


or an siRNA expression library, with the success that ran-
+1 +1
domized ribozyme libraries have had in the identification U6 or H1 promoter Sense TTTTT U6 or H1 promoter
Antisense
TTTTT
of novel genes (Beger et al., 2001; Kawasaki and Taira,
2002; Kawasaki et al., 2002; Kruger et al., 2000; Li et al.,
2000; Onuki et al., 2002; Rhoades and Wong-Staal, 2003; Transcription,
hybridization
Suyama et al., 2003a,b; Welch et al., 2000). In fact, several
Sense
groups, including ours, have already begun the generation
of an siRNA library against the whole human genome. In Antisense

this report, we present the practical problems and meth- siRNA duplex
ods for solving some of the difficulties that were faced in
generating the siRNA expression library. Hairpin type
+1 Loop
Antisense
U6 or H1 promoter TTTTT
2. Materials and methods

2.1. Constructs used in transient transfections


Transcription

We prepared tandem-type and hairpin-type siRNA expres-


Sense
sion vectors using the piGENE hU6 vector (Miyagishi and
Taira, 2003; iGENE Therapeutics, Inc.; http//www.iGENE- Antisense

therapeutics.co.jp), which contains a human U6 promoter Hairpin RNA


and two BspMI sites. For the construction of tandem-type
Processing
siRNA expression plasmids, we amplified DNA fragments
that included the sequences of the sense and antisense
Sense
regions and the U6 promoter by PCR with the piGENE
hU6 vector as a template, and primers that included the Antisense

sequences of the sense or antisense sequence plus the ter- siRNA duplex
minator (Fig. 1). After digestion of products of PCR with
Fig. 1. Schematic representation of tandem-type and hairpin-type
BspMI, each fragment was ligated into the BspMI sites of siRNA-expression vectors.
the piGENE hU6 vector to yield a series of siRNA expres-
sion vectors. For the construction of hairpin-type siRNA
expression vectors, we synthesized oligonucleotides with ciferase expression plasmid (pGL3; Promega, Madison,
the hairpin sequence, the terminator sequence, and over- WI, USA), and 10 ng of Renilla luciferase expression plas-
hanging sequences. Then we annealed the fragments and mid (pRL-RSV; Miyagishi et al., 2000), as described by
inserted them into the BspMI sites of the piGENE hU6 Promega. Luciferase activities were analyzed after 24 h
vector. H1 promoter-driven siRNA expression vectors were with a Dual Luciferase System (Promega). To ensure equal
generated as described by Brummelkamp et al. (2002). DNA amounts, empty plasmids were added at appropriate
Sequences inserted immediately downstream of the U6 pro- levels in each transfection.
moters or the H1 promoters were as follows (only the sense
sequences are shown): in pU6tandem19, pU6hairpin19,
and pH1hairpin19, GTG CGC TGC TGG TGC CAA C; in 2.3. Algorithm for searching favorable target sites
pU6tandem26, GTG CGC TGC TGG TGC CAA CCC TAT
TC; in pU6hairpin21, GTG CGC TGC TGG TGC CAA Prediction modeling of the RNAi effect was performed
CCC; in pU6hairpin23, GTG CGC TGC TGG TGC CAA by partial least squares (PLS) method. One hundred and
CCC TA; in pU6hairpin25, GTG CGC TGC TGG TGC fourteen data sets of siRNA sequences and their suppression
CAA CCC TAT T. The sequences of the various mutated values, obtained from published results and our own data,
constructs are indicated in Fig. 2. were analyzed and significantly correlated factors, including
GC contents, were extracted. A PLS calibration model was
2.2. Culture and transfection of cells and assays of the made with the target site sequences, their suppression values
expression of reporter genes and the extracted factors using the PLS1 algorithm of our
own making (Lindberg et al., 1983). The optimum number
HeLa S3 cells were cultured in Dulbecco’s modified of PLS factors was determined by cross-validation, that is,
Eagle’s medium supplemented with 10% fetal bovine serum. the best calibration as judged by the lowest standard error
Transfections were performed with LipofectamineTM 2000 of cross-validation. After equations were established, the
reagent (Life Technologies, Rockville, MD, USA) using suppression values at the various sites of the firefly luciferase
30 ng of siRNA expression plasmid, 30 ng of firefly lu- were predicted using their target sequences.
M. Miyagishi et al. / Virus Research 102 (2004) 117–124 119

Fig. 2. (A) Comparative analysis of the effects of the U6 promoter-driven 21-nt tandem-type and 21-nt hairpin-type siRNA expression vectors, the H1
promoter-driven 21-nt hairpin-type siRNA-expression vector and the U6-promoter-driven 21-nt hairpin-type with various mutations in hairpin region.
HeLa S3 cells were cotransfected with 10 ng of pRL-RSV, 30 ng of pGL3, and 30 ng of each siRNA expression vector targeted to the firefly gene
for luciferase. The activities of firefly luciferase were normalized by reference to those of Renilla luciferase. Each bar indicates an average value and
horizontal bars indicate standard errors of triplicate assays. (B) Schematic representation of the proposed siRNA expression system.

3. Results and discussion siRNA expression vectors, targeted against the same site of
firefly luciferase gene, and compared their suppressive ac-
3.1. Optimization of siRNA expression system: tandem-type tivities. Although both had high levels of suppressive activ-
or hairpin-type? ities at a high concentration of siRNA expression vectors,
it was found that the hairpin-type siRNA expression vec-
To construct the siRNA expression library, an optimiza- tor (21-nt stem length) had significantly higher suppressive
tion of the siRNA expression system, that has high suppres- activity than the tandem-type siRNA expression vector at a
sive activity and high genetic stability, should be initially low concentration of the siRNA expression vector (Fig. 2A).
addressed.
The major class of siRNA expression systems uses pol 3.2. Genetically stable hairpin-type siRNA expression
III promoter, such as U6 or H1 promoter. The systems have vector
been classified into two groups, tandem-type or hairpin-type,
depending on their expressing RNA (Fig. 1). In the case Although the hairpin-type siRNA expression vector has
of tandem-type siRNA expression vector, both sense and higher suppressive activity than the tandem-type siRNA ex-
antisense strands are driven separately by a respective pol pression vector, the palindromic sequence of the hairpin-type
III promoter, anneal in the cells and form siRNA duplexes siRNA expression vector frequently causes two serious tech-
with about four-nt overhang at each 3 -end (Lee et al., 2002; nical problems. Firstly, it is difficult to sequence constructs
Miyagishi and Taira, 2002). In the hairpin-type of siRNA ex- that contained a hairpin region, probably because of the tight
pression, sense and antisense nucleotides are connected by a palindromic structure. Secondly, approximately 20–40% of
loop and expressed as a single chain (Brummelkamp et al., constructs are mutated within the hairpin region of the con-
2002; Kawasaki and Taira, 2003; Paddison et al., 2002; Paul structs upon introduction into Escherichia coli, which is the
et al., 2002; Sui et al., 2002; Taira and Miyagishi, 2001; Yu most serious problem for the library construction. We were
et al., 2002). The transcribed RNA will rapidly form a hair- aware that constructs with two or more point mutations or
pin structure with a stem and loop and seems to be processed insertions/deletions in the sense or antisense region could be
to siRNA by Dicer, like micro RNAs (miRNAs) (Ketting sequenced without any problems and some of them retained
et al., 2001). We tried to examine which vector system had the suppressive activity. Therefore, we examined in detail
higher suppressive activity. We constructed both types of the activities of constructs with various mutations and/or
120 M. Miyagishi et al. / Virus Research 102 (2004) 117–124

insertions/deletions in the sense or antisense region (detailed promoter-driven siRNA construct targeted against firefly
analysis will be published elsewhere). As shown in Fig. 2A, luciferase has an activity comparable, but slightly lower,
as many as five-point mutations from C to T, which gen- than that of the U6-driven siRNA construct (comparing
erated G:U base-pairing, in the sense strand did not affect pH1hairpin21C > T5 with pU6hairpin21C > T5 in Fig 2A,
the silencing effect (pU6hairpin21C > T5 in Fig. 2A). In and other unpublished results). Therefore, we selected
contrast, insertion of a single C–T mutation in the antisense U6-driven siRNA expression vector as the promoter for
strand (pU6hairpinG > A1(AS) in Fig. 2A), which recog- the siRNA expression library. The tRNA-linked siRNA ex-
nizes the target transcript, resulted in reduced suppression. pression vector is a recently developed expression system,
Therefore, it appears that, in practice, it is not possible to which would enable us to forcibly transport the hairpin RNA
introduce mutations into the antisense strand and retain ac- to the cytoplasm (Kawasaki and Taira, 2003). However, it
tivity. requires a longer stem length (30-nt) and it would be more
While generating these constructs with mutated hairpin costly; hence we adopted the U6-driven siRNA expression
structures, we found that the rates of mutation in E. coli of vector for the construction of the siRNA expression library.
the hairpin region of the constructs were markedly reduced
when we inserted more than three mutations in the sense or 3.4. Optimization of the loop sequence
antisense strand (Taira and Miyagishi, 2001). This property
of such constructs is obviously important for the mainte- Different loop sequences of a hairpin-type siRNA ex-
nance of corresponding plasmids in their prokaryotic host. pression were used according to respective researchers.
Collectively, our data suggest that the way to generate a use- For example, Agami’s group uses a 9-nt loop sequence,
ful hairpin-type siRNA expression vector that enables us to 5 -UUCAAGAGA-3 , (Brummelkamp et al., 2002) and
construct an siRNA expression library is to introduce multi- Paul et al. (2002) use a 4-nt, 5 -UUCG-3 , tetra nucleotide
ple C–T (or A–G) mutations only into the sense strand part sequence. Our experiments using hairpin-type siRNA ex-
of the hairpin (Fig. 2B). pression vector with randomized 6-nt loop sequences re-
vealed that the hairpin RNAs with different loop sequences
3.3. Comparative analysis of various pol III promoters for had different suppression activities (Kawasaki and Taira,
silencing activity 2003; Miyagishi et al., 2004), therefore, the loop sequence
appears to somewhat influence the RNAi effect.
We also compared the RNAi activity of the U6 promoter We wondered whether the natural loop sequence of miR-
and the H1 promoter, both of which belong to the type II NAs might be a preferable sequence for the loop of the
Polymerase III promoters, and contain the same consensus hairpin RNA produced from hairpin-type siRNA expres-
motifs, distal sequence element (DSE), proximal sequence sion vectors. Examination of several miRNA-derived loop
element (PSE) and TATA box. As shown in Fig. 2A, H1 sequences revealed that loop 7, which is 11-nt in length and

1.0
Observed suppressive activity (relative)

0.8

0.6

0.4

0.2

0.0
0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1.0

Predicted suppressive activity (relative)


Fig. 3. Relationship between predicted and actual values of suppression activity. The actual values were measured by co-transfection experiments with
10 ng of pRL-RSV, 30 ng of pGL3, and 10 nM of each siRNA oligonucleotides targeted to the firefly gene for luciferase into HeLa cells. The activities
of firefly luciferase were normalized by reference to those of Renilla luciferase and the suppression values were calculated as the ratio to the negative
control. The predicted values were calculated from the target sequence of the siRNA with the PLS algorithm.
M. Miyagishi et al. / Virus Research 102 (2004) 117–124 121

derived from the human miRNA miR-26b (the sequence is and our own experimental data, extracted several correlated
5 -GUG UGC UGU CC-3 ), is highly effective (the loop factors, and generated an algorithm by nonlinear regression
is shown in Fig. 2B). Therefore, we adopted this loop as methods.
the optimized loop sequence of the hairpin-type siRNA Fig. 3 shows predicted versus measured results for siR-
expression vector for the siRNA expression library. NAs targeted against firefly luciferase, which were not used
in the generation of the algorithm. The correlation coeffi-
3.5. Method for selection of favorable target site cient was not high (0.58) but, notably, all of the siRNAs that
were calculated as scoring more than 0.73 showed high sup-
One of the most important and critical problems of the pressive activities (more than 90%), therefore, especially at
siRNA expression library would be selection of the tar- high prediction values, the predicted scores seem to be ac-
get sites. It is well-known that the effectiveness of siRNA curate. We use target sites that have a predicted suppressive
is very dependent on the target site of a message and the activity of more than 0.75 for the construction of the siRNA
possibility that siRNAs at an arbitrarily selected target site expression library.
have high suppressive activity, seems to be only at about
20–40% or less. To generate an siRNA expression library of 3.6. Confirmation of specificity of the target sequence
high-quality, an algorithm that can accurately predict an ef-
fective site must be developed. We analyzed siRNA data sets When one selects a certain target site in a specific gene,
of several hundreds of target sites, including published work there is a need to check the specificity of the sequence of the

(A) piGENE hU6


EcoRI BspMI BspMI
TTTTT
U6 promoter

Transcription start site


U6 promoter BspMI
+1
linker
CTTGTGGAAAGGACGAAA CACCGTGAGCAGGT---------------ACCTGCAGGC ATGCAAGCTT
GAACACCTTTCCTGCTTTGTGG CACTCGTCCA---------------TGGACGTCCGTACG TTCGAA
Cleavage site Cleavage site
BspMI

BspMI Loop BspMI


5'-CACC sense gtgtgctgtcc antisense ttttt-3
3'- sense cacacgacacc antisense aaaaaTACG-5'

(B)

Annealing individual
oligos (96 oligos) Ligated into
the U6 plasmid

Pick up 384 colonies

Sequencing
About 90% clones could be recovered

Fig. 4. (A) Cloning site and the insert sequence of hairpin-type siRNA-expression vector. (B) Our strategy for the construction of siRNA-expression library.
122
M. Miyagishi et al. / Virus Research 102 (2004) 117–124
Fig. 5. Identification of genes involved in apoptosis by using shRNAi library. (A) The siRNA-expression vectors targeted against PKR can block dsRNA-induced apoptosis. HeLaS3 cells were transfected
with siRNA-expression vectors targeted against two sites of PKR transcript (Site A; 5’TAA TGA ATC AAT CAA TTC ATA TC-3’ and Site B; 5’AAG ACT AAC TGT AAA TTA TGA AC-3’). After
the selection with puromycin, the cells were induced to undergo apoptosis by dsRNA transfection. The survived cells were fixed, and stained with crystal violet (0.2%). To check the reproducibility, each
independent plasmid was transfected into cells in three different wells (triplicate experiments). NC: negative control (unrelated siRNA-expression vector). (B) Light microscopic images of the cells in the
experiment of (B). (C) An example of the screening using siRNA-expression library. PC: positive control; NC1, NC2: negative controls (unrelated siRNA-expression vectors); C1–C11: siRNA-expression
vectors targeted against specific genes, for example kinases, transcription factors, or apoptosis-related genes. The shRNA expression vector in C7 inhibits the dsRNA-dependent apoptosis.
M. Miyagishi et al. / Virus Research 102 (2004) 117–124 123

target site, that is, to examine that there are no highly ho- siRNA expression vectors per month (Taira and Miyagishi,
mologous sequences in other genes. Under what conditions 2001).
should one check the specificity? If very stringent condi-
tions are applied, for example, more than three base mis- 3.8. Advantages and disadvantages of siRNA expression
matches are needed to determine the discrimination between library compared to siRNA oligonucleotide library
the correct and homologous targets, significant portions of
favorable target sites that were predicted as highly active For the plasmid-based siRNA expression library and the
sites could be eliminated by the BLAST search program. On alternative oligonucleotide-based siRNA library, there are
the other hand, if less stringent conditions are applied, for advantages and disadvantages.
example, only one base mismatch is allowed for the dis- Transfection efficiency of siRNA into cells using lipofec-
crimination, the designed siRNA might have the potential to tion depends on cell types and the RNAi effect only seems
disrupt not only the correct target but also other homologous to be sustained for a period of time. The advantage of the
genes as well, although the cleavage efficiency is reduced plasmid-based siRNA is the capability of removing those
for the latter. Under these circumstances, we have decided cells that were not transfected with the plasmids by se-
to use the relatively low stringent condition for examining lecting the transfected cells with antibiotic resistant genes.
target site specificity, because multiple positive candidates Moreover, the RNAi effect lasts for much longer periods
can be further analyzed by creating new siRNAs targeted at when plasmid-based siRNAs are used. Additionally, virus
different sites within the candidate gene. vectors enable us to deliver siRNA expression cassettes into
Alternative splicing and highly homologous gene fami- cells with higher transfection efficiency, and in the case of
lies are also major problems for target site selection. Re- lentivirus and retrovirus, it is easy to make stable knock-
cent genome-wide analysis of transcripts reveals that many down cells by the integration of the virus vector into the
genes have alternative splicing forms. For example, in case genome. If one wants to use siRNA expression vectors as
where one tries to disrupt a specific alternative variant, an a bulk library, these virus vectors, which can generate sta-
siRNA expression vector targeted against the specific vari- ble knockdown cells, would be most suitable for the bulk
ant can be created, without damaging other alternative vari- library screen.
ants originating from the same pre-spliced form. However, The advantage of oligonucleotide-based siRNA is that, in
for the generation of the first draft library, in the targeting some favorable cells, transfection efficiency is greater than
of genes which have multiple alternative splicing forms, the 90%, and thus higher than that of the plasmid. However, for
common sequence of all alternative transcripts (similarly, the library, once plasmid-based siRNAs are generated, they
the common sequence for the homologous gene family) can can be amplified unlimitedly, especially with our construct
be aimed as a target site. This is more economical for the (Fig. 2B) which does not undergo unwanted mutation (un-
creation of the first library and additional siRNAs can eas- like conventional ones) during amplification in E. coli cells,
ily be made once the targeted gene is identified as being an resulting in a more economical construction of a library
attractive candidate. set.

3.7. Improved method for library construction


4. Conclusions
In constructing an siRNA expression library against all
genes in the genome, one would become focused on practical During the present work, we have developed a genet-
problems, such as how to construct the library efficiently, ically stable and highly active siRNA expression vector,
rapidly, and at a reduced price. and have established a system for constructing high-quality
We have established a large-scale method for construct- and genome-wide scale siRNA expression libraries very
ing a library, as shown in Fig. 4. Specifically, 96 sets of efficiently. Recently, we have started to screen functional
oligonucleotides that include hairpin sequences correspond- genes by using the constructed siRNA-expression library.
ing to 96 target sequences were synthesized and annealed Fig. 5 shows an example of screening of functional genes in
separately. Then the annealed oligonucleotides were mixed, the apoptosis pathway. Positive controls (siRNA-expression
and were ligated into BspMI site of the U6-driven siRNA vectors targeted against PKR) could block dsRNA-induced
expression vector. After transformation into E. coli, 384 apoptosis (Fig. 5A), and in the experiments using siRNA ex-
clones were picked up and were sequenced to identify each pression vectors targeted against unknown genes (Fig. 5B;
clone. In our data, about 88.5% of the clones were recov- C1–C11), one positive clone could be identified fron 11 can-
ered in a single procedure. The unrecovered clones were didates (Fig. 5B; C7). Within the next few years, several
followed by the next round of the construction. This bulk groups, including our own would expect to identify a num-
procedure allowed the production of an siRNA expression ber of genes through the use of various siRNA libraries.
vector dramatically cheaper and faster than the traditional In the past, we were able to identify miRNA sequences by
procedure in which each clone is made separately. Us- our ribozyme library (Kuwabara et al., 2004). Since miRNA
ing this procedure, we can practically make 3000–5000 plays an important role in development, and possibly in other
124 M. Miyagishi et al. / Virus Research 102 (2004) 117–124

biological phenomena of mammalian cells (Kawasaki et al., against HIV-1 rev transcripts in human cells. Nat. Biotechnol. 20,
2004), the siRNA library should also cover miRNAs and un- 500–505.
Li, Q.X., Robbins, J.M., Welch, P.J., Wong-Staal, F., Barber, J.R., 2000. A
defined noncoding RNAs as targets. Scientists are making novel functional genomics approach identifies mTERT as a suppressor
the first great step in functional genome analysis, and human of fibroblast transformation. Nucleic Acids Res. 28, 2605–2612.
beings will soon enjoy deeper knowledge of the unraveled Lindberg, W., Persson, J.A., Wold, S., 1983. Partial least-squares method
life of cells. for spectrofluorimetric analysis of mixtures of humic acid and lignin
sulfonate. Anal. Chem. 55, 643–648.
McManus, M.T., Sharp, P.A., 2002. Gene silencing in mammals by small
interfering RNAs. Nat. Rev. Genet. 3, 737–747.
References Miyagishi, M., Taira, K., 2002. U6 promoter-driven siRNAs with four
uridine 3 overhangs efficiently suppress targeted gene expression in
Beger, C., Pierce, L.N., Kruger, M., Marcusson, E.G., Robbins, J.M., mammalian cells. Nat. Biotechnol. 20, 497–500.
Welcsh, P., Welch, P.J., Welte, K., King, M.C., Barber, J.R., Miyagishi, M., Taira, K., 2003. Expression of siRNA from a pol III
Wong-Staal, F., 2001. Identification of Id4 as a regulator of BRCA1 promoter in mammalian cells. In: Appasani, K. (Ed.), Perspectives
expression by using a ribozyme-library-based inverse genomics ap- in Gene Expression, The Eaton Publishers, Westboro, MA. pp. 361–
proach. Proc. Natl. Acad. Sci. U.S.A. 98, 130–135. 376.
Brummelkamp, T.R., Bernards, R., Agami, R., 2002. A system for stable Miyagishi, M., Fujii, R., Hatta, M., Yoshida, E., Araya, N., Nagafuchi,
expression of short interfering RNAs in mammalian cells. Science A., Ishihara, S., Nakajima, T., Fukamizu, A., 2000. Regulation of
296, 550–553. Lef-mediated transcription and p53-dependent pathway by associating
Caplen, N.J., Parrish, S., Imani, F., Fire, A., Morgan, R.A., 2001. Spe- ␤-catenin with CBP/p300. J. Biol. Chem. 275, 35170–35175.
cific inhibition of gene expression by small double-stranded RNAs in Miyagishi, M., Suminoto, H., Miyoshi, H., Kawakami, Y., Taira, K., 2004.
invertebrate and vertebrate systems. Proc. Natl. Acad. Sci. U.S.A. 98, Optimization of an siRNA-expression system with a mutated hair-
9742–9747. pin and its significant suppressive effects upon HIV vector-mediated
Elbashir, S.M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., Tuschl, transfer into mammalian cells. J. Gene Med., in press.
T., 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference Onuki, R., Nagasaki, A., Kawasaki, H., Baba, T., Uyeda, T.Q., Taira, K.,
in cultured mammalian cells. Nature 411, 494–498. 2002. Confirmation by FRET in individual living cells of the absence
Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E., Mello, of significant amyloid beta-mediated caspase 8 activation. Proc. Natl.
C.C., 1998. Potent and specific genetic interference by double-stranded Acad. Sci. U.S.A. 99, 14716–14721.
RNA in Caenorhabditis elegans. Nature 391, 806–811. Paddison, P.J., Caudy, A.A., Bernstein, E., Hannon, G.J., Conklin, D.S.,
Fraser, A.G., Kamath, R.S., Zipperlen, P., Martinez-Campos, M., 2002. Short hairpin RNAs (shRNAs) induce sequence-specific silenc-
Sohrmann, M., Ahringer, J., 2000. Functional genomic analysis of C. ing in mammalian cells. Genes Dev. 16, 948–958.
elegans chromosome I by systematic RNA interference. Nature 408, Paul, C.P., Good, P.D., Winer, I., Engelke, D.R., 2002. Effective expression
325–330. of small interfering RNA in human cells. Nat. Biotechnol. 20, 505–
Gonczy, P., Echeverri, C., Oegema, K., Coulson, A., Jones, S.J., Copley, 508.
R.R., Duperon, J., Oegema, J., Brehm, M., Cassin, E., Hannak, E., Rhoades, K., Wong-Staal, F., 2003. Inverse Genomics as a powerful tool to
Kirkham, M., Pichler, S., Flohrs, K., Goessen, A., Leidel, S., Alleaume, identify novel targets for the treatment of neurodegenerative diseases.
A.M., Martin, C., Ozlu, N., Bork, P., Hyman, A.A., 2000. Functional Mech. Ageing Dev. 124, 125–132.
genomic analysis of cell division in C. elegans using RNAi of genes Sui, G., Soohoo, C., Affarel, B., Gay, F., Shi, Y., Forrester, W.C., Shi,
on chromosome III. Nature 408, 331–336. Y., 2002. A DNA vector-based RNAi technology to suppress gene
Kawasaki, H., Taira, K., 2002. Identification of genes by hybrid ribozymes expression in mammalian cells. Proc. Natl. Acad. Sci. U.S.A. 99,
that couple cleavage activity with the unwinding activity of an en- 5515–5520.
dogenous RNA helicase. EMBO Rep. 3, 443–450. Suyama, E., Kawasaki, H., Kasaoka, T., Taira, K., 2003a. Identification
Kawasaki, H., Taira, K., 2003. Short hairpin type of dsRNAs that are of genes responsible for cell migration by a library of randomized
controlled by tRNAVal promoter significantly induce RNAi-mediated ribozymes. Cancer Res. 63, 119–124.
gene silencing in the cytoplasm of human cells. Nucleic Acids Res. Suyama, E., Kawasaki, H., Nakajima, M., Taira, K., 2003b. Identification
31, 700–707. of genes involved in cell invasion by using a library of randomized
Kawasaki, H., Onuki, R., Suyama, E., Taira, K., 2002. Identification of hybrid ribozymes. Proc. Natl. Acad. Sci. U.S.A. 100, 5616–5621.
genes that function in the TNF-alpha-mediated apoptotic pathway Taira, K., Miyagishi, M., 2001. siRNA Expression System and Method
using randomized hybrid ribozyme libraries. Nat. Biotechnol. 20, 376– for Producing Functional Gene Knock-down Cell Using the System.
380. Japanese Patent Application, Heiser–13–363385 (PCT/JP02/12447).
Kawasaki, H., Wadhwa, R., Taira, K., 2004. World of small RNAs: from Tuschl, T., 2002. Expanding small RNA interference. Nat. Biotechnol.
ribozymes to siRNA and miRNA. Differentiation, in press. 20, 446–448.
Ketting, R.F., Fischer, S.E., Bernstein, E., Sijen, T., Hannon, G.J., Plasterk, Warashina, M., Kuwabara, T., Kato, Y., Sano, M., Taira, K., 2001.
R.H., 2001. Dicer functions in RNA interference and in synthesis RNA-protein hybrid ribozymes that efficiently cleave any mRNA in-
of small RNA involved in developmental timing in Caenorhabditis dependently of the structure of the target RNA. Proc. Natl. Acad. Sci.
elegans. Genes Dev. 15, 2654–2659. U.S.A. 98, 5572–5577.
Kruger, M., Beger, C., Li, Q.X., Welch, P.J., Tritz, R., Leavitt, M., Welch, P.J., Marcusson, E.G., Li, Q.X., Beger, C., Kruger, M., Zhou,
Barber, J.R., Wong-Staal, F., 2000. Identification of eIF2Bgamma and C., Leavitt, M., Wong-Staal, F., Barber, J.R., 2000. Identification and
eIF2gamma as cofactors of hepatitis C virus internal ribosome entry validation of a gene involved in anchorage-independent cell growth
site-mediated translation using a functional genomics approach. Proc. control using a library of randomized hairpin ribozymes. Genomics
Natl. Acad. Sci. U.S.A. 97, 8566–8571. 66, 74–83.
Kuwabara, T., Hsieh, J., Nakashima, K., Taira, K., Gage, F.H., 2004. Yu, J.Y., DeRuiter, S.L., Turner, D.L., 2002. RNA interference by ex-
Small Modulatory dsRNA specifies the Fate of Adult Neural Stem pression of short-interfering RNAs and hairpin RNAs in mammalian
Cells. Cell, in press. cells. Proc. Natl. Acad. Sci. U.S.A. 99, 6047–6052.
Lee, N.S., Dohjima, T., Bauer, G., Li, H., Li, M.J., Ehsani, A., Salvaterra, Zamore, P.D., 2001. RNA interference: listening to the sound of silence.
P., Rossi, J., 2002. Expression of small interfering RNAs targeted Nat. Struct. Biol. 8, 746–750.
| |

An International Journal of Molecular and Cellular Virology


Instructions to Authors
EDITOR-IN-CHIEF
These can be found in the prelims of the first issue of each volume, and accessed on the
Dr. B.W.J. Mahy, Virus Research, Mailstop C12, National Center for Infectious Diseases, Centers for Disease Control and Prevention, World Wide Web under http://www.elsevier.com
Atlanta, GA 30333, USA. Tel. No.: þ1 404 728-0564; Fax No.: þ1 404 728-0032. E-mail: virology@bellsouth.net
This Journal is edited by Brian WJ Mahy in his private capacity. No official support or endorsement by CDC is intended or should be
inferred.

EDITORS Author enquiries


Dr. L. Enjuanes (European Editor), Virus Research, Department of Molecular and Cell Biology, National Center of Biotechnology For enquiries relating to the submission of articles (including electronic submission
(CNB), CSIC, Campus Universidad Autonoma, 28049 Madrid, Spain, Tel. No.: þ34 91 585 4555; Fax No.: þ34 91 585 4915. where available) please visit Elsevier’s Author Gateway at http://authors.elsevier.com.
E-mail: L.Enjuanes@cnb.uam.es The Author Gateway also provides the facility to track accepted articles and set up
Dr. Y. Kawaoka (Address for manuscript submission) Virus Research, Department of Pathobiological Sciences, School of Veterinary
Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA, Tel. No.: þ1 608 265 4925; Fax No.:
e-mail alerts to inform you of when an article’s status has changed, as well as detailed
þ1 608 265 5622. E-mail: VirusResearch@svm.vetmed.wisc.edu artwork guidelines, copyright information, frequently asked questions and more.
Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1,
Shirokanedai, Minato-Ku, Tokyo 108-8639, Japan Contact details for questions arising after acceptance of an article, especially those
relating to proofs, are provided after registration of an article for publication.
EDITORIAL BOARD
A. Barrett, Galveston, TX, USA R. Goldbach, Wageningen, The S. Nichol, Atlanta, GA, USA
T. Barrett, Pirbright, UK Netherlands A. Nomoto, Tokyo, Japan
Elsevier will do everything possible to get your article corrected and published as
W.J. Bellini, Atlanta, GA, USA F. Gonzales-Scarano, Philadelphia, F. Novembre, Atlanta, GA, USA quickly and accurately as possible. Therefore, it is important to ensure that all of your
W. Britt, Birmingham, AL, USA PA, USA D. O’Callaghan, Shreveport, LA, USA corrections are sent back to us in one communication. Subsequent corrections will not
M.S. Campo, Glasgow, UK D. Griffin, Baltimore, MD, USA N. Raab-Traub, Chapel Hill, NC, USA be possible, so please ensure that your first sending is complete.
D. Cavanagh, Compton, UK L. Guarino, College Station, TX, USA R. Ray, St Louis, MO, USA
P. Collins, Bethesda, MD, USA I. Jones, Reading, UK A.B. Rickinson, Birmingham, UK
R.W. Compans, Atlanta, GA, USA J. Katz, Atlanta, GA, USA J. Ridpath, Ames, IA, USA
K. Conzelmann, Muenchen, Germany H.-D. Klenk, Marburg, Germany B. Robertson, Atlanta, GA, USA
T. Dermody, Nashville, TN, USA M. Koopmans, Bilthoven, L. Roux, Geneva, Switzerland
V. Deubel, Lyon, France The Netherlands D. Rowlands, Leeds, UK
W. Doerfler, Cologne, Germany M. Lai, Los Angeles, CA, USA P. Roy, London, UK
E. Domingo, Madrid, Spain I. Lipkin, New York, NY, USA C. Schmaljohn, Frederick, MD, USA USA mailing notice: Virus Research (ISSN 0168-1702) is published monthly January to
R. Elliott, Glasgow, UK R.B. Luftig, New Orleans, LA, USA M.J. Studdert, Parkville, Victoria, December by Elsevier B.V. (P.O. Box 211, 1000 AE Amsterdam, The Netherlands).
H. Feldmann, Winnipeg, Canada W.S. Mason, Philadelphia, PA, USA Australia
B. Fleckenstein, Erlangen, Germany J.W. McCauley, Compton, UK G.T.W. Wertz, Birmingham, AL, USA Annual subscription price in the USA US$3245 (valid in North, Central and South
T. Folks, Atlanta, GA, USA P. Moore, Pittsburgh, PA, USA America), including air speed delivery. Periodical postage rate paid at Jamaica, NY
A. Garcia-Sastre, New York, NY, USA M. Mulligan, Birmingham, AL, USA 11431.
USA POSTMASTER: Send address changes to Virus Research, Publications Expedit-
AIMS AND SCOPE
ing Inc., 200 Meacham Avenue, Elmont, NY 11003.
Virus Research provides a means of rapid publication for original papers on fundamental research concerning virus structure, replication, AIRFREIGHT AND MAILING in the USA by Publications Expediting Inc., 200
and pathogenesis. These include reports describing virus morphology, the function and antigenic analysis of virus structural components,
virus genome structure and expression, analysis of virus replication processes, effects of viruses on their host cells, including oncogene Meacham Avenue, Elmont, NY 11003.
activation and transduction, neoplastic transformation, and the pathogenesis of virus infections including tumour induction. The journal
also publishes review articles on topics of current interest and occasional book reviews and meeting reports.

Publication information: Virus Research (ISSN 0168-1702). For 2004, volumes 99–106 are scheduled for publication.
Subscription prices are available upon request from the Publisher or from the Regional Sales Office nearest you or from Advertising information: Advertising orders and enquiries can be sent to: USA,
this journal’s website (http://www.elsevier.com/locate/virusres). Further information is available on this journal and other Canada and South America: Mr Tino DeCarlo, The Advertising Department,
Elsevier products through Elsevier’s website: (http://www.elsevier.com). Elsevier Inc., 360 Park Avenue South, New York, NY 10010-1710, USA; phone:
Subscriptions are accepted on a prepaid basis only and are entered on a calendar year basis. Issues are sent by standard
mail (surface within Europe, air delivery outside Europe). Priority rates are available upon request. Claims for missing (þ1) (212) 633 3815; fax: (þ1) (212) 633 3820; e-mail: t.decarlo@elsevier.com.
issues should be made within six months of the date of dispatch. Japan: The Advertising Department, Elsevier K.K., 4F Higashi-Azabu, 1-Chome
Bldg, 1-9-15 Higashi-Azabu, Minato-ku, Tokyo 106-0044, Japan; phone: (þ81) (3)
Orders, claims, and journal enquiries: please contact the Customer Service Department at the Regional Sales Office nearest you:
Orlando: Elsevier, Customer Service Department, 6277 Sea Harbor Drive, Orlando, FL 32887-4800, USA; phone: (þ1) (877) 8397126
5561 5037; fax: (þ81) (3) 5561 5047; e-mail: jp.info@elsevier.com. Europe and
[toll free number for US customers], or (þ1) (407) 3454020 [customers outside US]; fax: (þ1) (407) 3631354; e-mail: usjcs@ ROW: Katrina Barton, Print Operations Co-ordinator, Elsevier, 84 Theobald’s Road,
elsevier.com London WC1X 8RR, UK. Tel.: þ44 (0) 20 7611 4117; Fax: þ44 (0) 20 7611 4463;
Amsterdam: Elsevier, Customer Service Department, PO Box 211, 1000 AE Amsterdam, The Netherlands; phone: (þ31) (20) 4853757;
fax: (þ31) (20) 4853432; e-mail: nlinfo-f@elsevier.com
E-mail: k.barton@elsevier.com
Tokyo: Elsevier, Customer Service Department, 4F Higashi-Azabu, 1-Chome Bldg, 1-9-15 Higashi-Azabu, Minato-ku, Tokyo 106-0044,
Japan; phone: (þ81) (3) 5561 5037; fax: (þ81) (3) 5561 5047; e-mail: jp.info@elsevier.com
Singapore: Elsevier, Customer Service Department, 3 Killiney Road, #08-01 Winsland House I, Singapore 239519; phone: (þ65)
63490222; fax: (þ65) 67331510; e-mail: asiainfo@elsevier.com

doi:10.1016/S0168-1702(04)00146-7

| |
Viral Gene Silencing by RNA
Interference

Guest Editors
Susana Lopez
&
Carlos F. Arias
(Department of Developmental Genetics and Molecular Physiology,
Instituto de Biotechnologia, Universidad Nacional Autonoma de Mexico,
Avenida Universidad 2001,Colonia Chamilpa,
Cuernavaca, Morelos, 62210, Mexico)

Cover Image: Cryoelectron micrograph of spikeless rotavirus particles obtained by silencing


the spike protein VP4. The red asterisks indicate where the spikes should have
been.
[Courtesy of Dr. B.V.V. Prasad. Baylor College of Medicine, Houston, Texas].

doi:10.1016/S0168-1702(04)00155-8

You might also like