Skip to main content
Advertisement
  • Loading metrics

Sequence-Specific Inhibition of Small RNA Function

  • György Hutvágner ,

    Contributed equally to this work with: György Hutvágner, Martin J Simard

    Affiliation Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America

  • Martin J Simard ,

    Contributed equally to this work with: György Hutvágner, Martin J Simard

    Affiliation Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America

  • Craig C Mello,

    Affiliations Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America

  • Phillip D Zamore

    Affiliation Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America

Abstract

Hundreds of microRNAs (miRNAs) and endogenous small interfering RNAs (siRNAs) have been identified from both plants and animals, yet little is known about their biochemical modes of action or biological functions. Here we report that 2′-O-methyl oligonucleotides can act as irreversible, stoichiometric inhibitors of small RNA function. We show that a 2′-O-methyl oligonucleotide complementary to an siRNA can block mRNA cleavage in Drosophila embryo lysates and HeLa cell S100 extracts and in cultured human HeLa cells. In Caenorhabditis elegans, injection of the 2′-O-methyl oligonucleotide complementary to the miRNA let-7 can induce a let-7 loss-of-function phenocopy. Using an immobilized 2′-O-methyl oligonucleotide, we show that the C. elegans Argonaute proteins ALG-1 and ALG-2, which were previously implicated in let-7 function through genetic studies, are constituents of a let-7-containing protein–RNA complex. Thus, we demonstrate that 2′-O-methyl RNA oligonucleotides can provide an efficient and straightforward way to block small RNA function in vivo and furthermore can be used to identify small RNA-associated proteins that mediate RNA silencing pathways.

Introduction

The endoribonuclease Dicer produces two types of small regulatory RNAs that regulate gene expression: small interfering RNAs (siRNAs) and microRNAs (miRNAs) (Bernstein et al. 2001; Grishok et al. 2001; Hutvágner et al. 2001; Ketting et al. 2001; Knight and Bass 2001). In animals, siRNAs direct target mRNA cleavage (Elbashir et al. 2001b, 2001c), whereas miRNAs block target mRNA translation (Lee et al. 1993; Reinhart et al. 2000; Brennecke et al. 2003; Xu et al. 2003). Recent data suggest that both siRNAs and miRNAs incorporate into similar, perhaps even identical, protein complexes and that a critical determinant of mRNA destruction versus translation regulation is the degree of sequence complementary between the small RNA and its mRNA target (Hutvágner and Zamore 2002; Mourelatos et al. 2002; Zeng et al. 2002; Doench et al. 2003; Saxena et al. 2003; Zeng et al. 2003).

Target RNA cleavage directed by siRNA is called RNA interference (RNAi). RNAi is a powerful method for the study of gene function in animals and plants and has even been proposed as a therapy for treating genetic disorders and viral infections. Biochemical studies in Drosophila S2 cells (Bernstein et al. 2001; Hammond et al. 2001a; Caudy et al. 2002; Liu et al. 2003) and affinity purification (Martinez et al. 2002) or immunoprecipitation (Hutvágner and Zamore 2002) from cultured human HeLa cells have identified protein components of the RNAi effector complex, the RNA-induced silencing complex (RISC). Genetic mutations that disrupt RNAi in Caenorhabditis elegans, Drosophila, green algae, fungi, and plants have likewise identified proteins required for RNAi (Cogoni and Macino 1997, 1999a, 1999b; Ketting et al. 1999; Tabara et al. 1999, 2002; Catalanotto et al. 2000, 2002; Dalmay et al. 2000, 2001; Fagard et al. 2000; Grishok et al. 2000; Ketting and Plasterk 2000; Mourrain et al. 2000; Wu-Scharf et al. 2000; Grishok and Mello 2002; Tijsterman et al. 2002a, 2002b). Key steps in the RNAi pathway have also emerged from studies of RNAi reconstituted in cell-free extracts (Tuschl et al. 1999; Zamore et al. 2000; Hammond et al. 2001b; Nykänen et al. 2001; Martinez et al. 2002; Schwarz et al. 2002; Tang et al. 2003).

Recently, hundreds of miRNAs have been identified in animals and plants (Lagos-Quintana et al. 2001, 2002; Lau et al. 2001; Lee and Ambros 2001; Reinhart et al. 2002; Ambros et al. 2003; Aravin et al. 2003; Brennecke and Cohen 2003; Lim et al. 2003). Of these, the biological functions of only four animal miRNAs are known. In C. elegans, the miRNAs lin-4 (Lee et al. 1993; Olsen and Ambros 1999) and let-7 (Reinhart et al. 2000) regulate developmental timing, whereas the Drosophila miRNAs bantam and miR-14 control cell survival by repressing translation of proapoptotic genes (Brennecke et al. 2003; Xu et al. 2003). Computational approaches promise to identify the mRNA targets of other miRNAs (Enright et al. 2003; Lewis et al. 2003; Stark et al. 2003), but these proposed miRNA/target mRNA pairs will require experimental confirmation. Despite the widespread use of RNAi to “knock down” gene function, the RNAi pathway itself remains poorly understood. Similarly, despite intensive efforts to identify all miRNAs in vertebrates, worms, and flies, the mechanisms underlying miRNA function remain mysterious and no biological function has been ascribed to the vast majority of miRNAs.

Here we show that 2′-O-methyl oligonucleotides are potent and irreversible inhibitors of small RNA-directed RNA silencing in vivo and in vitro. Our experiments using 2′-O-methyl oligonucleotides also demonstrate that the acquisition of a target RNA by an siRNA-programmed RISC is far more efficient than the binding of an antisense oligonucleotide to the same region of the target. To demonstrate the utility of 2′-O-methyl oligonucleotides in probing RNA silencing pathways, we show that 2′-O-methyl oligonucleotides efficiently block siRNA-directed RISC activity in cell extracts and in cultured human HeLa cells. When injected into C. elegans larvae, a let-7-complementary 2′-O-methyl oligonucleotide can efficiently suppress lin-41 translational repression by the let-7 miRNA. Finally, we use a tethered 2′-O-methyl oligonucleotide to demonstrate association of the C. elegans Argonaute proteins ALG-1 and ALG-2 with let-7.

Results

Inhibition of RNAi by 2′-O-Methyl Oligonucleotides

Although RNAi has proved a straightforward and cost-effective method to assess the function of protein-coding mRNAs (Fire et al. 1998; Caplen et al. 2000, 2001; Carthew 2001; Elbashir et al. 2001a) and even some noncoding RNAs (Liang et al. 2003), no comparable method allows the sequence-specific inactivation of the siRNA or miRNA components of the RISC. The ideal inhibitor of RISC function would be a nucleic acid that would be recognized by the RISC by nucleotide complementarity, but be refractory to RISC-directed endonucleolytic cleavage or translational control. In theory, such a molecule would titrate out RISCs containing a complementary siRNA or miRNA, but not affect the function of RISCs containing guide RNAs unrelated in sequence. Such a RISC inhibitor should also resist degradation by cellular ribonucleases so that it persists long enough to bind RISC and block its function. Finally, the ideal inhibitor of small RNA function would act at concentrations unlikely to elicit nonspecific responses to the inhibitor itself, i.e., in the low nanomolar range, the same concentration at which siRNAs themselves are effective.

At micromolar concentration, DNA antisense oligonucleotides may block miRNA function in Drosophila embryos (Boutla et al. 2003), but the poor stability of DNA oligonucleotides in vivo may limit their utility. Phosphorothioate-substituted DNA oligonucleotides, which show good in vivo stability, do not inhibit RISC function in vitro (data not shown). 2′-O-methyl oligonucleotides are also highly resistant to cellular ribonucleases (Inoue et al. 1987). To test whether 2′-O-methyl oligonucleotides can act as RISC inhibitors, we asked whether a 2′-O-methyl oligonucleotide, tethered to streptavidin paramagnetic beads via a 5′ biotin linkage, could be used to deplete siRNA-programmed RISC from the reaction. Drosophila embryo lysate was programmed with a synthetic siRNA duplex directed against a firefly (Photinus pyralis) luciferase (Pp-luc) mRNA target (Figure 1A). Then, a tethered 31-nt 2′-O-methyl oligonucleotide complementary to the 21-nt siRNA antisense strand was added. Finally, the beads were removed from the solution using a magnet, and the supernatant was tested for siRNA-programmed RISC activity. Under these conditions, the 2′-O-methyl oligonucleotide completely depleted the reaction of the RISC programmed with the antisense strand of the siRNA, but not of RISC programmed with the sense strand (Figure 1B). Thus, depletion occurred only when the siRNA strand contained within RISC was complementary to the tethered oligonucleotide.

thumbnail
Figure 1. A 2′-O-Methyl RNA Oligonucleotide Inhibits RNAi In Vitro in Drosophila Embryo Lysate

(A) Sequences of the sense and antisense Pp-luc target RNAs (black), the siRNA (red, antisense strand; black, sense strand), and the sense and antisense 2′-O-methyl oligonucleotides (blue) used.

(B) Sequence-specific depletion of RNAi activity by immobilized 2′-O-methyl oligonucleotides from Drosophila embryo lysate programmed with siRNA. siRNA was incubated with lysate to assemble RISC; then, immobilized 2′-O-methyl oligonucleotide was added. Finally, the beads were removed from the supernatant, and either sense or antisense 32P-radiolabeled target RNA was added to the supernatant to measure RISC activity for each siRNA strand.

Symbols and abbreviations: Ø, target RNA before incubation with siRNA-programmed lysate; T, total reaction before depletion; unbound, the supernatant after incubation with the immobilized antisense (AS) or sense (S) 2′-O-methyl oligonucleotides shown in (A). The absence of 5′ cleavage product demonstrates that the sense oligonucleotide depleted RISC containing antisense siRNA, but not sense siRNA, and the antisense oligonucleotide depleted the sense RISC, but not that containing antisense siRNA. Bi, 5′ biotin attached via a six-carbon linker.

https://doi.org/10.1371/journal.pbio.0020098.g001

We extended this method to measure the amount of RISC formed in the in vitro reaction at different concentrations of the siRNA duplex. An siRNA duplex in which the antisense strand was 5′-32P-radiolabeled was incubated in the reaction; then, the tethered 2′-O-methyl oligonucleotide was added to deplete the reaction of antisense siRNA-programmed RISC. The beads were then washed and the fraction of 32P-siRNA bound to the beads determined. Depletion was verified by testing the supernatant for RISC activity. Formally, the amount of 32P-siRNA retained on the beads for a given concentration of siRNA duplex places an upper limit on the concentration of RISC formed. However, our results using this assay are, within error, identical to the amount of RISC measured by two independent methods: the accumulation of single-stranded siRNA from functionally asymmetric siRNA duplexes (Schwarz et al. 2003), and the magnitude of the burst of target cleavage measured by pre-steady-state kinetics (B. Haley and P. D. Zamore, unpublished data). The simplest explanation for our results is that this assay directly measures siRNA incorporated into RISC. Figure 2A shows the results of this assay for six different concentrations of siRNA duplex (5 nM, 15 nM, 25 nM, 50 nM, 100 nM, and 200 nM siRNA). First, the data show that RISC assembly in vitro is inefficient; the majority of siRNA duplexes do not contribute to RISC production. Second, RISC assembly is saturable, suggesting that some component of RISC itself is limiting.

thumbnail
Figure 2. 2′-O-Methyl Oligonucleotides Act as Stoichiometric, Irreversible Inhibitors of RISC Function

(A) The immobilized sense 2′-O-methyl oligonucleotide was used to determine the concentration of 32P-radiolabeled antisense siRNA assembled into RISC in Drosophila embryo. The 2′-O-methyl oligonucleotide and siRNA duplex are shown in Figure 1A.

(B–G) Inhibition of RNAi was measured using free 2′-O-methyl oligonucleotide and 1.3 nM (B), 4.6 nM (C), 9.3 nM (D), 14.5 nM (E), 18 nM (F), and 23.5 nM (G) RISC. The concentration of 2′-O-methyl oligonucleotide required for half-maximal inhibition (IC50) was calculated by fitting each dataset to a sigmoidal curve using a Hill coefficient of 1.

(H) A plot of IC50 versus RISC concentration suggests that each 2′-O-methyl oligonucleotide binds a single RISC. The data suggest that binding is essentially irreversible.

https://doi.org/10.1371/journal.pbio.0020098.g002

To understand better the mechanism by which the 2′-O-methyl oligonucleotide interacted with RISC, we measured the concentration of free 2′-O-methyl oligonucleotide required for half-maximal inhibition of RISC activity (IC50; Figure 2B–2G) at the six different RISC concentrations determined in Figure 2A. The IC50 for inhibition by free 2′-O-methyl oligonucleotide is shown for each RISC concentration in Figure 2H. The IC50 for the 2′-O-methyl oligonucleotide was remarkably close to half the RISC concentration, suggesting that a single 31-nt 2′-O-methyl oligonucleotide binds each RISC and blocks its function. Consistent with this apparent 1:1 stoichiometry, the data for the 2′-O-methyl oligonucleotide titrations fit well to sigmoidal curves, with a Hill coefficient of 1 (Figure 2B–2G). The sequence specificity of 2′-O-methyl oligonucleotide inhibition of RISC function clearly shows that inhibition reflects binding of the oligo to the RISC. Our data are most easily explained if the concentration of the 2′-O-methyl oligonucleotide required for inhibition in these experiments is much greater than the KD for binding; i.e., the experiments were conducted in a stoichiometric binding regime. Under a stoichiometric binding regime, inhibition by the 2′-O-methyl oligonucleotides would be essentially irreversible.

In theory, the 2′-O-methyl oligonucleotide might act by displacing the passenger (sense) strand of the siRNA duplex, thereby blocking incorporation of the guide (antisense) strand into RISC (Elbashir et al. 2001c). We can exclude this possibility because a 5′ tethered 31-nt 2′-O-methyl oligonucleotide complementary to the passenger strand of the siRNA did not deplete the guide-strand RISC activity (see Figure 1B). Similarly, an antisense sequence 2′-O-methyl oligonucleotide cannot pair with antisense RISC, but can pair with a sense target mRNA. We anticipated that this antisense 2′-O-methyl oligonucleotide would pair with the sense target mRNA and occlude the antisense RISC from the target. Surprisingly, the antisense 2′-O-methyl oligonucleotide was a poor inhibitor of antisense RISC function when it was used to bind the target site, requiring 300 nM for half-maximal inhibition in a reaction containing 14.5 nM RISC and 3 nM sense target RNA (Figure 3A). By contrast, the same antisense 2′-O-methyl oligonucleotide was highly effective in blocking the activity of the sense RISC, to which it is complementary, acting with an IC50 of 8.2 nM in a reaction containing 16.8 nM sense-strand RISC and 3 nM antisense target RNA (Figure 3B). (In this experiment, sense-strand RISC was generated by changing the first nucleotide of the sense strand from C to U, thereby reversing its functional asymmetry [Schwarz et al. 2003].)

thumbnail
Figure 3. RISC Does Not Act through an Antisense Mechanism

(A) Inhibition of sense target cleavage by an antisense 2′-O-methyl oligonucleotide requires an approximately 40-fold higher concentration than by a sense oligonucleotide. The antisense oligonucleotide can pair completely with the sense target RNA, but not with the antisense siRNA-programmed RISC. The IC50 value and the RISC concentration are indicated. Also shown are the sequences of the sense Pp-luc RNA target (black), the siRNA (red, antisense strand; black, sense strand), and the 2′-O-methyl oligonucleotide (blue).

(B) The same antisense 2′-O-methyl oligonucleotide is an effective competitor of antisense target cleavage. In this experiment, inhibition occurs via binding of the antisense oligonucleotide to the sense siRNA-programmed RISC, not the target RNA. The IC50 value and the RISC concentration are indicated. Also shown are the sequences of the Pp-luc antisense RNA target (black), the siRNA (red, antisense strand; black, sense strand), and the 2′-O-methyl oligonucleotide (blue). The G:U wobble in the siRNA duplex in (B) acts to direct the sense strand into RISC and improving its efficacy in target cleavage.

https://doi.org/10.1371/journal.pbio.0020098.g003

Thus, the interaction of 2′-O-methyl oligonucleotide with RISC is dramatically different from the interaction of 2′-O-methyl oligonucleotide with target RNA; RISC has a more than 40-fold greater affinity for the 2′-O-methyl oligonucleotide than the oligonucleotide has for an RNA target (compare Figures 2E and 3A). These data imply that the interaction of RISC with target is more complex than simple nucleic acid hybridization. Inhibition of the siRNA-programmed RISC by a 2′-O-methyl oligonucleotide with the sequence of the target RNA is more effective than inhibition mediated by binding of an oligonucleotide to the target RNA itself. Thus, the RISC is more adept at finding or remaining bound (or both) to the target RNA than a 2′-O-methyl oligonucleotide. These data suggest that specific proteins in the RISC facilitate either target finding, target binding, or both. Consistent with this idea, inhibition of RISC function was incomplete using 21-nt 2′-O-methyl oligonucleotides (data not shown). Thus, target sequence flanking the site of complementarity to the siRNA guide strand may play a role in target RISC binding. Perhaps an active mechanism that involves target sequences flanking the siRNA facilitates the search for the target sequence; future studies will clearly be needed to test this idea.

Inhibition of RNAi in Cultured Human Cells

Our data show that 2′-O-methyl oligonucleotides are stoichiometric, irreversible, sequence-specific inhibitors of siRNA function in RNAi reactions using Drosophila embryo lysate. Can 2′-O-methyl oligonucleotides block siRNA function in vivo? To address this question, we carried out sequential transfection experiments using 1 nM, 5 nM, 10 nM, or 25 nM siRNA duplex. siRNA was transfected on the first day; then, reporter and control plasmids were cotransfected together with various amounts of 2′-O-methyl oligonucleotide on the second day. Silencing of Pp-luc, relative to the sea pansy (Renilla reniformis) luciferase (Rr-luc) control, was measured on the third day. For each siRNA concentration, we determined the concentration of 2′-O-methyl required for half-maximal inhibition of RNAi (Figure 4A–4D). Increasing amounts of the 2′-O-methyl oligonucleotide gradually extinguished the ability of the siRNA to silence Pp-luc in all four experiments. The inhibition of silencing in the cultured cells cannot be a consequence of the 2′-O-methyl oligonucleotide displacing the sense strand of the siRNA duplex, because assembly of siRNA into RISC occurred a full day before the oligonucleotide was introduced. When 10 nM siRNA was used in the transfection, approximately 1 nM 2′-O-methyl RNA was required for half-maximal inhibition of RNAi (Figure 4C and 4E). At 25 nM siRNA, approximately 1.1 nM 2′-O-methyl RNA was required to inhibit half the RNAi activity (Figure 4D and 4E). In Figure 4E, we plot the siRNA concentration versus the amount of 2′-O-methyl oligonucleotide required for half-maximal inhibition of silencing (IC50). The data fit well to a sigmoidal curve, consistent with the idea that, at these concentrations, increasing amounts of siRNA do not produce a corresponding increase in RISC activity. Higher concentrations of siRNA were not examined because they produce sequence-independent changes in gene expression (Persengiev et al. 2003; Semizarov et al. 2003). We conclude that both cells and extracts have a limited capacity to assemble RISC on exogenous siRNA. Our data suggest that the use of siRNA concentrations greater than that required to produce the maximum amount of RISC will lead to the accumulation of double-stranded siRNA in vivo and may thus contribute to the undesirable, sequence-independent responses sometimes observed in cultured mammalian cells (Persengiev et al. 2003; Semizarov et al. 2003; Sledz et al. 2003).

thumbnail
Figure 4. A 2′-O-Methyl Oligonucleotide Is a Potent Inhibitor of RNAi in Human Cultured HeLa Cells

(A–D) HeLa cells were transfected with 1 nM (A), 5 nM (B), 10 nM (C), or 25 nM (D) siRNA-targeting Pp-luc mRNA. The next day the cells were cotransfected with Rr-luc-and Pp-luc-expressing plasmids together with various amounts of a 31-nt 2′-O-methyl oligonucleotide complementary to the antisense strand of the siRNA. The half-maximal concentration of 2′-O-methyl oligonucleotide required to inhibit (IC50) was determined by fitting the data to a sigmoidal curve using a Hill coefficient of 1.

(E) IC50 plotted as a function of the concentration of transfected siRNA.

https://doi.org/10.1371/journal.pbio.0020098.g004

Inhibition of miRNA Function In Vitro and In Vivo

In animal cells, miRNAs are thought predominantly to function as translational regulators. Nonetheless, a growing body of evidence suggests that they function through a similar, if not identical, RISC as siRNAs (Hutvágner and Zamore 2002; Zeng et al. 2002, 2003; Doench et al. 2003; Khvorova et al. 2003; Schwarz et al. 2003). Because 2′-O-methyl oligonucleotides block siRNA function in vitro and cultured human cells, we asked whether these oligonucleotides might likewise disrupt the function of a specific miRNA in vitro and in vivo. An ideal candidate for such an miRNA is let-7. Classical genetic mutations in C. elegans let-7 produce well-characterized, readily scored phenotypes. Furthermore, human HeLa cells express multiple let-7 family members, and endogenous let-7 is present naturally in RISC (Hutvágner and Zamore 2002; Zeng and Cullen 2003). We tested whether a 31-nt 2′-O-methyl oligonucleotide complementary to let-7 could block target cleavage guided by the endogenous let-7-programmed RISC present in HeLa S100 extract (Figure 5A). (Our assay detects the target-cleaving activity of let-7; we have not examined endogenous human mRNA targets whose translation may be repressed by let-7.) As a control, we also tested whether the oligonucleotide could block the activity of a let-7-containing RISC assembled in vitro in Drosophila embryo lysate. Addition of this 2′-O-methyl oligonucleotide efficiently blocked target RNA cleavage directed by the endogenous let-7-programmed RISC in the HeLa S100 extract and by the RISC programmed with exogenous let-7 siRNA duplex in Drosophila embryo lysate (Figure 5C).

thumbnail
Figure 5. A Complementary 2′-O-Methyl Oligonucleotide Blocks Endogenous let-7-Containing RISC Function

(A) Sequence of the let-7-complementary site in the target RNA (black), of the siRNA (red, antisense strand; black, sense strand), and of the let-7-complementary 2′-O-methyl oligonucleotide (blue).

(B) Schematic representation of the target RNA, which contained both Pp-luc and antisense let-7 sequences.

(C) Drosophila embryo lysate (left) was programmed with let-7 siRNA; then, the target RNA and the 2′-O-methyl oligonucleotide were added together. Target RNA and 2′-O-methyl oligonucleotide (right) were added to HeLa S100 extract, which contains endogenous human let-7-programmed RISC.

(D) An RNA target containing both Pp-luc and antisense let-7 sequence can be simultaneously targeted by Pp-luc siRNA and endogenous let-7 in HeLa S100 lysate. The let-7-complementary 2′-O-methyl oligonucleotide blocks let-7-programmed, but not Pp-luc siRNA-programmed, RISC function. The bottom panel shows the same samples analyzed separately to better resolve the let-7 5′ cleavage product.

(E) Drosophila embryo lysate was programmed with let-7 siRNA and then incubated with biotinylated 2′-O-methyl oligonucleotide tethered to paramagnetic streptavidin beads. The beads were removed and the supernatant tested for RNAi activity.

Symbols and abbreviations: Ø, target RNA before incubation with siRNA-programmed lysate; T, total reaction before depletion; unbound, the supernatant after incubation with the paramagnetic beads. “Mock” indicates that no oligonucleotide was used on the beads; “let-7” indicates that the beads contained the let-7-complementary oligonucleotide shown in (A).

https://doi.org/10.1371/journal.pbio.0020098.g005

In addition to containing endogenous let-7-programmed RISC, HeLa S100 can be programmed with exogenous siRNA duplexes (Martinez et al. 2002; Schwarz et al. 2002). The target RNA used in Figure 5B also contains sequence from the Pp-luc mRNA and can therefore be targeted by a Pp-luc-specific siRNA duplex (see Figures 1A and 5C). We incubated the Pp-luc siRNA duplex with the human HeLa S100 to form Pp-luc-directed RISC, then added the let-7-complementary 2′-O-methyl oligonucleotide and the target RNA. The oligonucleotide blocked cleavage by the endogenous let-7-programmed RISC, but had no effect on cleavage directed by the exogenous Pp-luc siRNA in the same reaction (Figure 5D). When tethered to a paramagnetic bead, this oligonucleotide could also quantitatively deplete the let-7-programmed RISC from the Drosophila embryo lysate (Figure 5E), demonstrating that, again, the interaction between the 2′-O-methyl oligonucleotide and the RISC was apparently irreversible. The 2′-O-methyl oligonucleotide was a specific and potent inhibitor of target cleavage directed by a naturally occurring, miRNA-programmed RISC. Furthermore, our data demonstrate that individual RISCs act independently even when they target the same RNA.

Next we asked whether 2′-O-methyl oligonucleotides can inhibit miRNA function in vivo. Translational repression directed by miRNAs occurs in C. elegans, where both the lin-4 and let-7 miRNAs have been shown to block translation of their target mRNAs without altering mRNA stability (Wightman et al. 1993; Ha et al. 1996; Moss et al. 1997; Olsen and Ambros 1999; Reinhart et al. 2000; Seggerson et al. 2002). The genetics of lin-4 and let-7 function are well-characterized in worms, where they are required during larval development to control the timing and pattern of cell division in the hypodermis (Lee et al. 1993; Reinhart et al. 2000). First, we tested whether injection into the germline of wild-type adult hermaphrodites of 2′-O-methyl oligonucleotides complementary to either lin-4 or let-7 could block lin-4 or let-7 function during the larval development of the resulting progeny. Although the 2′-O-methyl oligonucleotides were not toxic and when coinjected with an unrelated DNA transformation reporter did not prevent the uptake and expression of the coinjected DNA, we did not observe inhibition of lin-4 or let-7 activity (data not shown). This finding suggests that single-stranded 2′-O-methyl oligonucleotides are not efficiently transmitted to the progeny of injected animals. To circumvent this problem, we next injected 2′-O-methyl oligonucleotides directly into larvae and examined the phenotypes of the injected animals. The lin-4 miRNA functions in L1/L2 larvae, and we have found that, in our hands, L1 larvae do not survive microinjection (data not shown); thus, it was not possible to assay for inhibition of lin-4 function by direct injection. In contrast, let-7 functions during the L4 stage, and we found that L2 and L3 larvae survive the microinjection procedure (see Materials and Methods). Loss of let-7 function causes worms to reiterate the L4 larval molt and inappropriately produce larval cuticle at the adult stage. Loss-of-function let-7 phenotypes include weak cuticles prone to bursting at the vulva, defects in egg-laying, and loss of adult-specific cuticular structures that run the length of the animal's body, the alae (Reinhart et al. 2000). After larvae were injected with the let-7-specific 2′-O-methyl oligonucleotide, 80% of the adult worms lacked alae; 77% lacked alae and also exhibited bursting vulvae (Figure 6A). In contrast, animals injected with an unrelated control 2′-O-methyl oligonucleotide displayed no abnormal phenotypes (Figure 6A).

thumbnail
Figure 6. Injection of a 2′-O-Methyl Oligonucleotide Complementary to let-7 miRNA Can Phenocopy the Loss of let-7 Function in C. elegans

(A) Wild-type and lin-41(ma104) L2-stage C. elegans larvae were injected with either a 2′-O-methyl oligonucleotide complementary to let-7 miRNA (Figure 5A) or an unrelated Pp-luc 2′-O-methyl oligonucleotide. Absence of alae and presence of bursting vulvae were scored when the injected animals reached adulthood.

(B) Isolation of let-7-associated proteins with a tethered 2′-O-methyl oligonucleotide. Northern blot analysis of let-7 miRNA remaining in the supernatant of the worm lysate after incubation with the let-7-complementary (let-7) or Pp-luc (unrelated) oligonucleotide. Input represents the equivalent of 50% of the total extract incubated with tethered oligonucleotide.

(C) Western blot analysis of the GFP-tagged ALG-1 and ALG-2 proteins associated with let-7. The upper band corresponds to GFP-tagged ALG-1 and the lower to GFP-tagged ALG-2. Extracts from a transgenic strain expressing the tagged proteins was incubated with the indicated tethered 2′-O-methyl oligonucleotide; then, the beads were washed and bound proteins were fractionated on an 8% SDS-polyacrylamide gel. Western blots were probed using anti-GFP monoclonal or anti-RDE-4 polyclonal antibody. The RDE-4-specific band is marked with an asterisk (Tabara et al. 2002).

(D and E) Analysis of let-7 miRNA in ALG-1/ALG-2 complexes (D). Extracts prepared from mixed-stage wild-type worms (N2) or from GFP::ALG-1/ALG-2 transgenic worms were immunoprecipitated using anti-GFP monoclonal antibodies. The unbound and immunoprecipitated RNAs were analyzed by Northern blot hybridization for let-7 (D), and 5% of the immunoprecipitated protein was analyzed by Western blotting for GFP to confirm recovery of the GFP-tagged ALG-1/ALG-2 proteins (E).

https://doi.org/10.1371/journal.pbio.0020098.g006

All of the phenotypes associated with injection of the let-7-complementary 2′-O-methyl oligonucleotide are consistent with a loss of let-7 activity. let-7 represses translation of lin-41 mRNA by binding to a partially complementary site in the lin-41 3′-untranslated region (Reinhart et al. 2000; Slack et al. 2000; Vella et al. 2004). Consequently, many of the phenotypes associated with the loss of let-7 reflect overexpression of LIN-41 protein; let-7 mutants are partially suppressed by mutations in lin-41. We reasoned that if the phenotypes observed in the injected larvae reflect a loss of let-7 activity, then they should likely be partially suppressed by a lin-41 mutation (Reinhart et al. 2000; Slack et al. 2000). To test this possibility, we injected the let-7-specific 2′-O-methyl oligonucleotide into the lin-41(ma104) strain and compared the penetrance of the phenotypes with those observed for injection into wild-type. Consistent with the idea that the injected oligonucleotide specifically inactivates let-7, the absence of alae- and vulval-bursting phenotypes were both suppressed in the lin-41(ma104) mutant strain (Figure 6A). The number of worms lacking alae was reduced from 80% to 16%, and worms with bursting vulvae were dramatically reduced (74% in wild-type compared to 3.8% in the lin-41(ma104) strain). The observed suppression (64%) was nearly identical to that reported for a let-7, lin-41 genetic double mutant (70%; Reinhart et al. 2000; Slack et al. 2000). Together, our data support the idea that 2′-O-methyl oligonucleotides are potent inhibitors of miRNA function that can be used to probe the function of specific miRNAs in C. elegans.

Isolation of Protein–miRNA Complex Using a Tethered 2′-O-Methyl Oligonucleotide

Our in vitro experiments suggest that both siRNA- and miRNA-containing RISCs are stably bound by 2′-O-methyl oligonucleotides. In theory, tethered 2′-O-methyl oligonucleotides could be used to isolate cellular factors associated with specific miRNAs. In human cells, miRNAs such as let-7 are in a protein complex that contains Argonaute proteins (Hutvágner and Zamore 2002; Mourelatos et al. 2002; Dostie et al. 2003). In C. elegans, the Argonaute protein-encoding genes alg-1 and alg-2 are required for the biogenesis or function (or both) of the miRNAs lin-4 and let-7 (Grishok et al. 2001), but it has not been shown whether ALG-1 and ALG-2 proteins are directly associated with let-7.

We prepared extracts from wild-type adult worms carrying a transgene expressing GFP-tagged ALG-1 and ALG-2 proteins. The extracts were then incubated with the let-7-complementary 2′-O-methyl oligonucleotide tethered by a 5′ biotin to streptavidin-conjugated paramagnetic beads. As a control, the experiment was performed in parallel using an oligonucleotide not complementary to let-7. The let-7-complementary, but not the control, oligonucleotide depleted nearly all the let-7 miRNA from the extract (Figure 6B). Western blotting using an anti-GFP antibody revealed that both GFP-tagged ALG-1 and ALG-2 protein copurified with the let-7-complementary oligonucleotide, but not the control oligonucleotide (Figure 6C). In contrast, the RNA-binding protein RDE-4, which is required for RNAi but not for miRNA function in C. elegans, did not copurified with the let-7-complementarity oligonucleotide, providing further support for the specificity of the let-7:ALG-1/ALG-2 interaction (Figure 6C).

Finally, we used a coimmunoprecipitation assay to examine the interaction between let-7 and ALG-1/ALG-2. In this assay, a monoclonal anti-GFP antibody was used to coimmunoprecipitate ALG-1/ALG-2 and small RNAs from the GFP::ALG-1/GFP::ALG-2 strain, which expresses GFP::ALG-1/ALG-2 fusion proteins. Northern blot analysis of the immune complex showed that it contained mature 22-nt let-7 miRNA (Figure 6D). No detectable let-7 was recovered with the anti-GFP antibody from the N2 wild-type strain. By comparing the fraction of let-7 associated with GFP::ALG-1/ALG-2 with the unbound fraction of let-7 miRNA, we estimate that approximately 30% of the 22-nt let-7 RNAs coimmunoprecipitate with GFP::ALG-1 and GFP::ALG-2. These data support a model in which that ALG-1 and ALG-2 form a complex, in vivo, that contains a substantial fraction of the mature let-7 miRNA.

Discussion

Our studies indicate that 2′-O-methyl oligonucleotides bind efficiently and essentially irreversibly to RISC by basepairing with the small guide RNA. These findings provide a rapid and reliable method to measure programmed RISC concentration in vitro and to identify the in vivo functions of small RNA and the identities of their associated proteins. The ability to measure RISC concentration should enable detailed kinetic studies of the enzymatic activity of RISC, an essential step in understanding RISC function. In fact, this method was recently put to use in analyzing the molecular basis of asymmetry in siRNA function (Schwarz et al. 2003). In this study, we have used a tethered 2′-O-methyl oligonucleotide to demonstrated the association of ALG-1/ALG-2, two C. elegans Argonaute proteins, with the endogenous worm miRNA let-7.

Our in vitro and in vivo studies using 2′-O-methyl oligonucleotides demonstrate that cells and extracts have a limited capacity to assemble RISC on exogenous siRNA. Our in vitro experiments suggest that inhibition of RISC by 2′-O-methyl oligonucleotides is stoichiometric and essentially irreversible. Using a sequential transfection protocol in cultured cells, we find that the half-maximal amount of 2′-O-methyl oligonucleotide required to inhibit silencing (IC50) is less than the amount of siRNA transfected. These data suggest that only a fraction of the transfected siRNA forms RISC. Furthermore, the data are consistent with stoichiometric and irreversible binding of the 2′-O-methyl oligonucleotide to RISC in vivo.

Our data hint that recognition of the 2′-O-methyl oligonucleotide by RISC and, by inference, recognition of target RNA by RISC are qualitatively different from the simple binding of two complementary nucleic acids by basepairing. We observed that RISC function was far more readily inhibited by binding a 2′-O-methyl oligonucleotide to RISC than by binding the same 2′-O-methyl oligonucleotide to the site of RISC recognition on a target RNA. A clear implication of this finding is that RISC does not acquire its RNA target by a passive basepairing mechanism that zippers together 21 nt of complementary RNA. Thus, RNAi is not merely a form of antisense inhibition in which the antisense strand is stabilized in a duplex. Rather, an active mechanism—perhaps involving target sequences flanking the region of complementarity—underlies the specificity and efficiency of RISC targeting.

Finally, we have shown the utility of 2′-O-methyl oligonucleotides to probe miRNA function in vivo. Injection of a 2′-O-methyl oligonucleotide complementary to the let-7 miRNA into C. elegans larvae phenocopied a let-7 loss-of-function mutation, demonstrating that 2′-O-methyl oligonucleotides can disrupt the function of a single miRNA in vivo. These data, combined with our studies in vitro and in cultured cells, show the promise of 2′-O-methyl oligonucleotides as a tool for dissecting the function of the numerous miRNAs found in a wide range of organisms. In this regard, 2′-O-methyl oligonucleotides provide a tool similar in practice, but mechanistically distinct from, RNAi itself and thus may facilitate the study of small RNA function in cases in which classical genetic mutations in miRNA genes are unavailable.

Materials and Methods

General methods.

Drosophila embryo lysate preparation, in vitro RNAi reactions, and cap-labeling of target RNAs were as described elsewhere (Haley et al. 2003). Target RNAs were used at approximately 3 nM concentration. Cleavage products of RNAi reactions were analyzed by electrophoresis on 5% or 8% denaturing polyacrylamide gels. Gels were dried, exposed to image plates, and then scanned with a FLA-5000 phosphorimager (Fuji Photo Film Company, Tokyo, Japan). Images were analyzed using Image Reader FLA-5000 version 1.0 (Fuji) and Image Gauge version 3.45 (Fuji). Data analysis was performed using Excel (Microsoft, Redmond, Washington, United States) and IgorPro 5.0 (Wavemetrics, Lake Oswego, Oregon, United States).

siRNA and 2′-O-methyl oligonucleotides.

Synthetic siRNA (Dharmacon, Lafayette, Colorado, United States) was deprotected according to the manufacturer, annealed (Elbashir et al. 2001b, 2001c), and used at 50 nM final concentration unless otherwise noted. 2′-O-methyl oligonucleotides (either from IDT, Santa Clara, California, United States, or from Dharmacon) were 5′-CAU CAC GUA CGC GGA AUA CUU CGA AAU GUC C-3′ and 5′-Bio-CAU CAC GUA CGC GGA AUA CUU CGA AAU GUC C-3′ (complementary to the Pp-luc siRNA sense strand); 5′-GGA CAU UUC GAA GUA UUC CGC GUA CGU GAU G-3′ and 5′-Bio-A CAU UUC GAA GUA UUC CGC GUA CGU GAU GUU-3′ (complementary to the Pp-luc antisense strand); and 5′-Bio-UCU UCA CUA UAC AAC CUA CUA CCU CAA CCU U-3′ (complementary to let-7); 5′ biotin was attached via a six-carbon spacer arm.

Immobilized 2′-O-methyl oligonucleotide capture of RISC.

Biotinylated 2′-O-methyl oligonucleotide (10 pmol) was incubated for 1 h on ice in lysis buffer containing 2 mM DTT with 50 μl of Dynabeads M280 (as a suspension as provided by the manufacturer; Dynal, Oslo, Norway) to immobilize the oligonucleotide on the beads. To ensure that the tethered oligonucleotide remained in excess when more than 50 nM siRNA was used, 20 pmol of biotinylated 2′-O-methyl oligonucleotide was immobilized. For RISC capture assays, siRNA was preincubated in a standard 50 μl in vitro RNAi reaction for 15 min at 25°C. Then, the immobilized 2′-O-methyl oligonucleotide was added to the reaction and incubation continued for 1 h at 25°C. After incubation, beads were collected using a magnetic stand (Dynal). The unbound supernatant was recovered and an aliquot assayed for RISC activity as previously described (Elbashir et al. 2001b; Nykänen et al. 2001) to confirm that RISC depletion was complete. The beads were then washed three times with ice-cold lysis buffer containing 0.1% (w/v) NP-40 and 2 mM DTT, followed by a wash without NP-40. To determine the amount of RISC formed, input and bound radioactivity was determined by scintillation counting (Beckman Instruments, Fullerton, California, United States). To isolate let-7-containing complexes from C. elegans adults, we incubated 20 pmol of immobilized 2′-O-methyl oligonucleotide with 1 mg of total protein.

Sequential transfection.

HeLa S3 cells were transfected in a 24-well plate (200 mm2 per well) using Lipofectamine 2000 (GIBCO, San Diego, California, United States) according to the manufacturer's protocol, first with various concentrations of siRNA targeting Pp-luc mRNA. After 6 h, the cells were washed with PBS and the media replaced. On the next day, the cells were cotransfected with Rr-luc-expressing (0.1 μg/well) and Pp-luc-expressing (0.25 μg/well) plasmids and 2′-O-methyl oligonucleotides using Lipofectamine 2000 (GIBCO) according to the manufacturer's protocol. The luciferase activity was measured 24 h later with the Dual Luciferase assay kit (Promega, Madison, Wisconsin, United States) using a Mediators Diagnostika (Vienna, Austria) PhL luminometer.

Worm injection.

For in vivo inhibition of let-7 function, 1 mg/ml let-7-complementary 2′-O-methyl oligonucleotide in water (100 μM) was injected into either wild-type (N2) or lin-41(ma104) L2 larvae. Injection of L2 larvae was essentially as described elsewhere (Conte and Mello 2003). The 2′-O-methyl oligonucleotide solution was injected into the body cavity of the larvae using the low flow and pressure setting to prevent animals from dying. Despite these precautions, approximately 60% of the animals do not survive injection, irrespective of the oligonucleotide injected. let-7 phenotypes were also observed at 10 μM oligonucleotide, but were less penetrant. Phenotypes were scored after the injected animals survived to adulthood.

Other methods.

Synchronized transgenic animals carrying GFP::ALG-1, GFP::ALG-2 were harvested at adulthood and homog-enized in ice-cold buffer (25 mM HEPES–NaOH [pH 7.4], 150 mM NaCl, 1 mM EDTA, 1 mM DTT, 10% [v/v] glycerol, 0.5% [v/v] Triton X-100, 2% [v/v] SUPERaseIn [Ambion, Austin, Texas, United States]) and Mini Complete Protease Inhibitor cocktail (1 tablet/10 ml solution) (Roche, Basel, Switzerland) using a stainless-steel Dounce homogenizer (Wheaton Incorporated, Millville, New Jersey, United States). The homogenized extract was clarified by a centrifugation at 13,817 × g for 10 min at 4°C.

To recover the proteins associated with the let-7 miRNA, the beads were boiled for 10 min in 20 μl of SDS loading buffer (10 mM Tris–HCl [pH 6.8], 2% [w/v] SDS, 100 mM DTT, and 10% [v/v] glycerol). Proteins were resolved by SDS-PAGE on an 8% gel and transferred to Hybond-C membrane (Amersham Biosciences, Little Chalfont, United Kingdom). To detect GFP-tagged ALG-1, ALG-2, and RDE-4 proteins, the membrane was incubated overnight at 4°C with either monoclonal anti-GFP (Roche) or an affinity-purified polyclonal anti-RDE-4 antibody (Tabara et al. 2002) diluted 1:1000 into TBST-milk solution (100 mM Tris–HCl [pH 7.5], 150 mM NaCl, 0.1% [v/v] Tween-20, and 5% [w/v] dried milk), incubated 1 h at room temperature with either anti-mouse (GFP-tagged ALG-1/ALG-2) or anti-rabbit (RDE-4) HRP-conjugated secondary antibody (Jackson Laboratory, Bar Harbor, Maine, United States) diluted 1:5,000 in TBST and then visualized by enhanced chemulinescence (New England Nuclear, Boston, Massachusetts, United States).

Immunoprecipitation of GFP-tagged ALG-1/ALG-2 complexes was performed by preclearing worm extract with 50 μl of protein G–agarose beads (Roche) per 5 mg of total protein for 1 h at 4°C. The cleared extract was then incubated with 10 μg of monoclonal antibody anti-AFP 3E6 (Qbiogene, Montreal, Quebec, Canada) for 1 h at 4°C followed by 50 μl of protein G–agarose. The agarose beads were then washed three times with ice-cold homogenization buffer.

Depletion of let-7 miRNA was monitored by Northern blotting. RNA was eluted from the immobilized 2′-O-methyl oligonucleotide by digestion with 1 mg/ml proteinase K in 200 mM Tris–HCl (pH 7.5), 25 mM EDTA, 300 mM NaCl, 2% (w/v) SDS at 50°C for 30 min, followed by extraction with phenol–chloroform, and recovered by precipitation with ethanol. Recovered RNA was resuspended in 10 μl of formamide-loading buffer (98% [v/v] deionized formamide, 10 mM EDTA, 0.025% [w/v] xylene cyanol, 0.025 % [w/v] bromophenol blue), heated to 100°C for 2 min. RNA was resolved on a 15% denaturing polyacrylamide gel, transferred to Hybond-N membrane (Amersham Biosciences), and detected by Northern blot analysis using a 5′-32P-radiolabeled antisense let-7 RNA probe (UAU ACA ACC UAC UAC CUC AUU) as described elsewhere (Hutvágner and Zamore 2002).

Supporting Information

Accession Numbers

The GenBank (http://www.ncbi.nlm.nih.gov/Genbank/) accession number for Photinus pyralis is X65324 and for Renilla reniformis is AF025846.

Rfam (http://www.sanger.ac.uk/Software/Rfam/index.shtml) accession numbers for the let-7 family members are MI0000060–MI0000068, MI0000433, and MI0000434.

The LocusLink (http://www.ncbi.nlm.nih.gov/LocusLink/) ID numbers for the genes discussed in this paper are alg-1 (181504), alg-2 (173468), bantam (117376), let-7 (266954), lin-4 (266860), lin-41 (172760), miR-14 (170868), and rde-4 (176438).

Acknowledgments

We thank Darryl Conte, Jr., for the gfp::alg-1 and gfp::alg-2 plasmids; Erbay Yigit for generating the transgenic line expressing GFP::ALG-1, GFP::ALG-2; and members of the Zamore and Mello laboratories for helpful discussion and encouragement. Research support was provided by National Institutes of Health to PDZ (GM62862-01 and GM65236-01) and CCM (GM58800). GH is a Charles A. King Trust fellow of the Medical Foundation; MJS is a Canadian Institutes of Health Research postdoctoral fellow; CCM is a Howard Hughes Medical Institute assistant investigator; and PDZ is a Pew Scholar in the Biomedical Sciences and a W. M. Keck Foundation Young Scholar in Medical Research.

References

  1. 1. Ambros V, Lee RC, Lavanway A, Williams PT, Jewell D (2003) MicroRNAs and other tiny endogenous RNAs in C. elegans. Curr Biol 13: 807–818.
  2. 2. Aravin AA, Lagos-Quintana M, Yalcin A, Zavolan M, Marks D, et al. (2003) The small RNA profile during Drosophila melanogaster development. Dev Cell 5: 337–350.
  3. 3. Bernstein E, Caudy AA, Hammond SM, Hannon GJ (2001) Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409: 363–366.
  4. 4. Boutla A, Delidakis C, Tabler M (2003) Developmental defects by antisense-mediated inactivation of micro-RNAs 2 and 13 in Drosophila and the identification of putative target genes. Nucleic Acids Res 31: 4973–4980.
  5. 5. Brennecke J, Cohen S (2003) Towards a complete description of the microRNA complement of animal genomes. Genome Biol 4: 228.
  6. 6. Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen SM (2003) bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell 113: 25–36.
  7. 7. Caplen NJ, Fleenor J, Fire A, Morgan RA (2000) dsRNA-mediated gene silencing in cultured Drosophila cells: A tissue culture model for the analysis of RNA interference. Gene 252: 95–105.
  8. 8. Caplen NJ, Parrish S, Imani F, Fire A, Morgan RA (2001) Specific inhibition of gene expression by small double-stranded RNAs in invertebrate and vertebrate systems. Proc Natl Acad Sci U S A 98: 9742–9747.
  9. 9. Carthew RW (2001) Gene silencing by double-stranded RNA. Curr Opin Cell Biol 13: 244–248.
  10. 10. Catalanotto C, Azzalin G, Macino G, Cogoni C (2000) Transcription: Gene silencing in worms and fungi. Nature 404: 245.
  11. 11. Catalanotto C, Azzalin G, Macino G, Cogoni C (2002) Involvement of small RNAs and role of the qde genes in the gene silencing pathway in Neurospora. Genes Dev 16: 790–795.
  12. 12. Caudy AA, Myers M, Hannon GJ, Hammond SM (2002) Fragile X-related protein and VIG associate with the RNA interference machinery. Genes Dev 16: 2491–2496.
  13. 13. Cogoni C, Macino G (1997) Isolation of quelling-defective (qde) mutants impaired in posttranscriptional transgene-induced gene silencing in Neurospora crassa. Proc Natl Acad Sci U S A 94: 10233–10238.
  14. 14. Cogoni C, Macino G (1999a) Gene silencing in Neurospora crassa requires a protein homologous to RNA-dependent RNA polymerase. Nature 399: 166–169.
  15. 15. Cogoni C, Macino G (1999b) Posttranscriptional gene silencing in Neurospora by a RecQ DNA helicase. Science 286: 2342–2344.
  16. 16. Conte D, Mello CC (2003) RNA interference in Caenorhabditis elegans. In: Asubel FM, Brent R, Kingston RE, Moore DD, Seidman JG, editors. Current protocols in molecular biology. New York: John Wiley and Sons. pp. 26.23.1–26.23.20. et al.
  17. 17. Dalmay T, Hamilton A, Rudd S, Angell S, Baulcombe DC (2000) An RNA-dependent RNA polymerase gene in Arabidopsis is required for posttranscriptional gene silencing mediated by a transgene but not by a virus. Cell 101: 543–553.
  18. 18. Dalmay T, Horsefield R, Braunstein TH, Baulcombe DC (2001) SDE3 encodes an RNA helicase required for post-transcriptional gene silencing in Arabidopsis. EMBO J 20: 2069–2078.
  19. 19. Doench JG, Petersen CP, Sharp PA (2003) siRNAs can function as miRNAs. Genes Dev 17: 438–442.
  20. 20. Dostie J, Mourelatos Z, Yang M, Sharma A, Dreyfuss G (2003) Numerous microRNPs in neuronal cells containing novel microRNAs. RNA 9: 180–186.
  21. 21. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, et al. (2001a) Duplexes of 21-nucleotide RNAs mediate RNA interference in mammalian cell culture. Nature 411: 494–498.
  22. 22. Elbashir SM, Lendeckel W, Tuschl T (2001b) RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev 15: 188–200.
  23. 23. Elbashir SM, Martinez J, Patkaniowska A, Lendeckel W, Tuschl T (2001c) Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO J 20: 6877–6888.
  24. 24. Enright A, John B, Gaul U, Tuschl T, Sander C, et al. (2003) MicroRNA targets in Drosophila. Genome Biol 5: R1.
  25. 25. Fagard M, Boutet S, Morel J-B, Bellini C, Vaucheret H (2000) AGO1, QDE-2, and RDE-1 are related proteins required for post-transcriptional gene silencing in plants, quelling in fungi, and RNA interference in animals. Proc Natl Acad Sci U S A 97: 11650–11654.
  26. 26. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, et al. (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391: 806–811.
  27. 27. Grishok A, Mello CC (2002) RNAi (Nematodes: Caenorhabditis elegans). Adv Genet 46: 339–360.
  28. 28. Grishok A, Tabara H, Mello C (2000) Genetic requirements for inheritance of RNAi in C. elegans. Science 287: 2494–2497.
  29. 29. Grishok A, Pasquinelli AE, Conte D, Li N, Parrish S, et al. (2001) Genes and mechanisms related to RNA interference regulate expression of the small temporal RNAs that control C. elegans developmental timing. Cell 106: 23–34.
  30. 30. Ha I, Wightman B, Ruvkun G (1996) A bulged lin-4/lin-14 RNA duplex is sufficient for Caenorhabditis elegans lin-14 temporal gradient formation. Genes Dev 10: 3041–3050.
  31. 31. Haley B, Tang G, Zamore PD (2003) In vitro analysis of RNA interference in Drosophila melanogaster. Methods 30: 330–336.
  32. 32. Hammond SM, Boettcher S, Caudy AA, Kobayashi R, Hannon GJ (2001a) Argonaute2, a link between genetic and biochemical analyses of RNAi. Science 293: 1146–1150.
  33. 33. Hammond SM, Caudy AA, Hannon GJ (2001b) Post-transcriptional gene silencing by double-stranded RNA. Nat Rev Genet 2: 110–119.
  34. 34. Hutvágner G, Zamore PD (2002) A microRNA in a multiple-turnover RNAi enzyme complex. Science 297: 2056–2060.
  35. 35. Hutvágner G, McLachlan J, Pasquinelli AE, Balint É, Tuschl T, et al. (2001) A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science 293: 834–838.
  36. 36. Inoue H, Hayase Y, Imura A, Iwai S, Miura K, et al. (1987) Synthesis and hybridization studies on two complementary nona(2′-O-methyl)ribonucleotides. Nucleic Acids Res 15: 6131–6148.
  37. 37. Ketting RF, Plasterk RH (2000) A genetic link between co-suppression and RNA interference in C. elegans. Nature 404: 296–298.
  38. 38. Ketting RF, Haverkamp TH, van Luenen HG, Plasterk RH (1999) Mut-7 of C. elegans, required for transposon silencing and RNA interference, is a homolog of Werner syndrome helicase and RNaseD. Cell 99: 133–141.
  39. 39. Ketting RF, Fischer SE, Bernstein E, Sijen T, Hannon GJ, et al. (2001) Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes Dev 15: 2654–2659.
  40. 40. Khvorova A, Reynolds A, Jayasena SD (2003) Functional siRNAs and miRNAs exhibit strand bias. Cell 115: 209–216.
  41. 41. Knight SW, Bass BL (2001) A role for the RNase III enzyme DCR-1 in RNA interference and germ line development in Caenorhabditis elegans. Science 293: 2269–2271.
  42. 42. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T (2001) Identification of novel genes coding for small expressed RNAs. Science 294: 853–858.
  43. 43. Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, et al. (2002) Identification of tissue-specific microRNAs from mouse. Curr Biol 12: 735–739.
  44. 44. Lau NC, Lim LP, Weinstein EG, Bartel DP (2001) An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 294: 858–862.
  45. 45. Lee RC, Ambros V (2001) An extensive class of small RNAs in Caenorhabditis elegans. Science 294: 862–864.
  46. 46. Lee RC, Feinbaum RL, Ambros V (1993) The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75: 843–854.
  47. 47. Lewis B, Shih I, Jones-Rhoades M, Bartel D, Burge C (2003) Prediction of mammalian microRNA targets. Cell 115: 787–798.
  48. 48. Liang XH, Liu Q, Michaeli S (2003) Small nucleolar RNA interference induced by antisense or double-stranded RNA in trypanosomatids. Proc Natl Acad Sci U S A 100: 7521–7526.
  49. 49. Lim LP, Lau NC, Weinstein EG, Abdelhakim A, Yekta S, et al. (2003) The microRNAs of Caenorhabditis elegans. Genes Dev 17: 991–1008.
  50. 50. Liu Q, Rand TA, Kalidas S, Du F, Kim HE, et al. (2003) R2D2, a bridge between the initiation and effector steps of the Drosophila RNAi pathway. Science 301: 1921–1925.
  51. 51. Martinez J, Patkaniowska A, Urlaub H, Lührmann R, Tuschl T (2002) Single-stranded antisense siRNA guide target RNA cleavage in RNAi. Cell 110: 563–574.
  52. 52. Moss EG, Lee RC, Ambros V (1997) The cold shock domain protein LIN-28 controls developmental timing in C. elegans and is regulated by the lin-4 RNA. Cell 88: 637–646.
  53. 53. Mourelatos Z, Dostie J, Paushkin S, Sharma AK, Charroux B, et al. (2002) miRNPs: A novel class of ribonucleoproteins containing numerous microRNAs. Genes Dev 16: 720–728.
  54. 54. Mourrain P, Beclin C, Elmayan T, Feuerbach F, Godon C, et al. (2000) Arabidopsis SGS2 and SGS3 genes are required for posttranscriptional gene silencing and natural virus resistance. Cell 101: 533–542.
  55. 55. Nykänen A, Haley B, Zamore PD (2001) ATP requirements and small interfering RNA structure in the RNA interference pathway. Cell 107: 309–321.
  56. 56. Olsen PH, Ambros V (1999) The lin-4 regulatory RNA controls developmental timing in Caenorhabditis elegans by blocking LIN-14 protein synthesis after the initiation of translation. Dev Biol 216: 671–680.
  57. 57. Persengiev SP, Zhu X, Green MR (2003) Nonspecific, concentration-dependent stimulation and repression of mammalian gene expression by small interfering RNAs (siRNAs). RNA 10: 12–18.
  58. 58. Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, et al. (2000) The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403: 901–906.
  59. 59. Reinhart BJ, Weinstein EG, Rhoades MW, Bartel B, Bartel DP (2002) MicroRNAs in plants. Genes Dev 16: 1616–1626.
  60. 60. Saxena S, Jonsson ZO, Dutta A (2003) Small RNAs with imperfect match to endogenous mRNA repress translation: Implications for off-target activity of siRNA in mammalian cells. J Biol Chem 278: 44312–44319.
  61. 61. Schwarz DS, Hutvágner G, Haley B, Zamore PD (2002) Evidence that siRNAs function as guides, not primers, in the Drosophila and human RNAi pathways. Mol Cell 10: 537–548.
  62. 62. Schwarz DS, Hutvágner G, Du T, Xu Z, Aronin N, et al. (2003) Asymmetry in the assembly of the RNAi enzyme complex. Cell 115: 199–208.
  63. 63. Seggerson K, Tang L, Moss EG (2002) Two genetic circuits repress the Caenorhabditis elegans heterochronic gene lin-28 after translation initiation. Dev Biol 243: 215–225.
  64. 64. Semizarov D, Frost L, Sarthy A, Kroeger P, Halbert DN, et al. (2003) Specificity of short interfering RNA determined through gene expression signatures. Proc Natl Acad Sci U S A 100: 6347–6352.
  65. 65. Slack FJ, Basson M, Liu Z, Ambros V, Horvitz HR, et al. (2000) The lin-41 RBCC gene acts in the C. elegans heterochronic pathway between the let-7 regulatory RNA and the LIN-29 transcription factor. Mol Cell 5: 659–669.
  66. 66. Sledz CA, Holko M, de Veer MJ, Silverman RH, Williams BR (2003) Activation of the interferon system by short-interfering RNAs. Nat Cell Biol 5: 834–839.
  67. 67. Stark A, Brennecke J, Russell RB, Cohen SM (2003) Identification of Drosophila microRNA targets. PLoS Biol 1: e60.
  68. 68. Tabara H, Sarkissian M, Kelly WG, Fleenor J, Grishok A, et al. (1999) The rde-1 gene, RNA interference, and transposon silencing in C. elegans. Cell 99: 123–132.
  69. 69. Tabara H, Yigit E, Siomi H, Mello CC (2002) The dsRNA binding protein RDE-4 interacts with RDE-1, DCR-1, and a DexH-box helicase to direct RNAi in C. elegans. Cell 109: 861–871.
  70. 70. Tang G, Reinhart BJ, Bartel DP, Zamore PD (2003) A biochemical framework for RNA silencing in plants. Genes Dev 17: 49–63.
  71. 71. Tijsterman M, Ketting RF, Okihara KL, Sijen T, Plasterk RH (2002a) RNA helicase MUT-14-dependent gene silencing triggered in C. elegans by short antisense RNAs. Science 295: 694–697.
  72. 72. Tijsterman M, Okihara KL, Thijssen K, Plasterk RH (2002b) PPW-1, a PAZ/PIWI protein required for efficient germline RNAi, is defective in a natural isolate of C. elegans. Curr Biol 12: 1535–1540.
  73. 73. Tuschl T, Zamore PD, Lehmann R, Bartel DP, Sharp PA (1999) Targeted mRNA degradation by double-stranded RNA in vitro. Genes Dev 13: 3191–3197.
  74. 74. Vella MC, Choi EY, Lin SY, Reinert K, Slack FJ (2004) The C. elegans microRNA let-7 binds to imperfect let-7 complementary sites from the lin-41 3′UTR. Genes Dev 18: 132–137.
  75. 75. Wightman B, Ha I, Ruvkun G (1993) Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 75: 855–862.
  76. 76. Wu-Scharf D, Jeong B, Zhang C, Cerutti H (2000) Transgene and transposon silencing in Chlamydomonas reinhardtii by a DEAH-box RNA helicase. Science 290: 1159–1163.
  77. 77. Xu P, Vernooy SY, Guo M, Hay BA (2003) The Drosophila microRNA Mir-14 suppresses cell death and is required for normal fat metabolism. Curr Biol 13: 790–795.
  78. 78. Zamore PD, Tuschl T, Sharp PA, Bartel DP (2000) RNAi: Double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell 101: 25–33.
  79. 79. Zeng Y, Cullen BR (2003) Sequence requirements for micro RNA processing and function in human cells. RNA 9: 112–123.
  80. 80. Zeng Y, Wagner EJ, Cullen BR (2002) Both natural and designed micro RNAs can inhibit the expression of cognate mRNAs when expressed in human cells. Mol Cell 9: 1327–1333.
  81. 81. Zeng Y, Yi R, Cullen BR (2003) MicroRNAs and small interfering RNAs can inhibit mRNA expression by similar mechanisms. Proc Natl Acad Sci U S A 100: 9779–9784.