Much of our knowledge of the early steps in RSV entry is by analogy to similar enveloped viruses. In general, enveloped viruses are divided into two classes, those for which fusion and entry are triggered by acidic pH and a second group that fuses to the cell membrane at neutral pH. This classification is based primarily on the sensitivity of virus infection to drugs that neutralize endosomal acidification, as well as the sensitivity of infectivity to acidic pH. For many types of virus, including the well-studied vesicular stomatitis virus (VSV) and influenza A virus, a drop in pH is necessary and sufficient to mediate envelope protein structural rearrangements (
8,
9,
19,
60) that bring about membrane fusion and delivery of the virus capsid into the cell cytosol (
24). For other viruses, like Ebola, the activation of pH-dependent proteases is required to cleave the envelope proteins before structural rearrangement and membrane fusion can occur (
10). For such viruses, endocytosis is required to bring the virus into the acidic environment of the early-to-late-stage endosome. The best-studied endocytic pathway involves clathrin-coated pit formation and is controlled by clathrin and the AP2 complex (
45). Caveolae form a second class of endocytic vesicle but are less-well characterized and are distinguished by the presence of caveolin instead of clathrin (
36). Still a third set of endocytic pathways are neither caveolin nor clathrin associated but may involve Arf-related proteins (
37) and, like caveolae, are involved in the uptake of lipid rafts. Each of these endocytic pathways appears to converge on acidified endosomes (
38), and drugs that block endosomal acidification can be potent antivirals (
56).
In contrast, most retroviruses and paramyxoviruses enter cells irrespective of treatment with drugs that inhibit endosomal acidification. For RSV, membrane fusion and infection of HEp-2 cells were resistant to treatment with a weak base, ammonium chloride, that buffers against acidification (
28,
48). This has been interpreted to mean that RSV does not require endocytosis and instead, like other pH-independent viruses, fuses directly with the plasma membrane. In other work, using video microscopy of HEp-2 cells, RSV viral filaments were seen being taken up through the plasma membrane by an undefined macropinocytic mechanism (
46). Such uptake would likely require extensive actin and cytoskeleton rearrangement to engulf the viral filament. Unfortunately, this work did not indicate if this adsorption of filaments led to productive infection or, instead, to a dead-end pathway. No similar studies have been reported with spherical particles. It therefore remains unclear what route is used by RSV to productively infect cells.
MATERIALS AND METHODS
Chemicals.
All chemicals were ultragrade from Sigma (St. Louis, MO) unless stated otherwise. Dharmafect cell culture reagent and DharmaFECT 1 transfection reagent were from Dharmacon (Lafayette, CO).
Antibodies.
Polyclonal serum raised against VSV Indiana strain, obtained from ATCC (VR-1238AF), was used for fluorescence-activated cell sorter (FACS) detection of virus infection. A monoclonal antibody against RSV F protein (MCA490) was obtained from Serotec (Raleigh, NC) and used for the detection of RSV infection by FACS analysis. The secondary antibody was Alexa Fluor647-labeled goat anti-mouse immunoglobulin G (A21235) from Invitrogen (Carlsbad, CA). Antibodies for confirmation of siRNA knockdown of protein expression were from Santa Cruz Biotech (Santa Cruz, CA) and were made against unique peptides in the targeted protein. Those that were used in this report were against AP2-alpha (sc-17771), Rab5 (isoforms A, B, and C were sc-309, sc-598, and sc-26570, respectively), clathrin light chain A (sc-28276), clathrin light chain B (sc-28277), dynamin I (sc-12724), dynamin II (sc-6400), PAK-1 (sc-882), or WASF2 (sc-10394). Both anti-dynamin 3 antibody (ab3458) and anti-beta-actin antibody (600-532A1; served as loading control) were from Novus Biologicals, Littleton, CO. Each was detected on blots by using appropriate secondary horseradish peroxidase-conjugated antibodies. Goat anti-mouse (Pierce, Rockford, IL), mouse anti-goat (Pierce), or donkey anti-rabbit (Amersham) antibodies were used where appropriate. Chemiluminescent substrate detection was used to visualize bound antibody on blots (ECL kit; Amersham).
Cell lines and culture conditions.
Cells were maintained in a humidified air-5% CO2 atmosphere incubator at 37°C. 293 FT cells were from Invitrogen. Both 293 and HeLa cells (ATCC CCL-2) were grown in Dulbecco's modified Eagle's medium (Gibco) supplemented with 10% fetal bovine serum (Gemini Bioproducts, West Sacramento, CA).
Virus stocks.
VSV Indiana strain was obtained from ATCC (VR-1238). VSV Indiana strain expressing green fluorescent protein (GFP) or dsRed was kindly provided by M. Whitt (University of Alabama). RSV expressing dsRed was provided by M. Peeples and P. Collins (
22). Wild-type RSV was from ATCC (VR-26). For RSV cultivation, HeLa cells were grown to 20% confluence and then infected at a multiplicity of infection (MOI) of 0.5. The cells were incubated at 37°C in a CO
2 incubator until a cytopathic effect was observed (typically 2 days postinfection). The culture supernatant was harvested and cell debris was pelleted for 5 min at 3,000 ×
g. The supernatant was used immediately or stored frozen at −80°C. For VSV, 293 cells were grown to 80% confluence and infected at an MOI of 0.5. The culture supernatant was collected the following day and filtered through a 0.45-μm syringe filter to remove cell debris. Virus was used immediately or stored frozen at −80°C.
GFP, red fluorescent protein, or virus envelope protein expression was used to monitor infection by each virus. For VSV, infection was detected 7 h after infection. For RSV, marker expression was detected 20 h postinfection. Infected cells were counted by using a Leica DMIRB inverted epifluorescence microscope.
Production of envelope protein-pseudotyped murine leukemia virus.
Virus envelope protein-pseudotyped murine leukemia viruses (MLV) were prepared as controls in the drug treatment experiments and for FACS analysis of virus infection. Pseudotypes were made bearing the envelope proteins of VSV type G (VSV-G), ecotropic MLV, or 10A1 MLV. Both the VSV-G- and ecotropic MLV-pseudotyped viruses encoded GFP as a marker of infection. The 10A1-pseudoyped virus encoded either GFP or a truncated CD4 (CD4.1) marker (Miltenyi Biotec, Auburn, CA) of infection. The latter was cloned into the retroviral packaging plasmid pFB (Stratagene, La Jolla, CA). Pseudotyped virus was assembled by transfecting 293 FT cells (Invitrogen) with plasmids encoding the MLV core structural proteins (pGag-Pol) together with plasmids carrying the reporter gene (pΨ-GFP or pFB-CD4.1) and encoding the envelope protein of VSV (pVSV-G; Clontech, Mountain View, CA); ecotropic MLV (pcDNA3:Fr-env), as detailed elsewhere (
30); or p10A1 (Clontech, Mountain View, CA). Calcium phosphate was used as the method of plasmid transfection. After 2 days, culture supernatants were collected and filtered through a 0.45-μm filter to remove cell debris, and the filtered virus was used immediately or stored frozen at −80°C. The expression of GFP was detected 36 h postinfection by counting fluorescent cells by microscopy.
Drug treatments.
To confirm the pH independence of RSV infection, cells were treated with lysosomotropic agents which inhibit endosomal acidification. VSV (a pH-dependent virus) and ecotropic MLV (a pH-independent virus) were used as controls. Cells were incubated for 1 h in the presence of chloroquine (10 μM), ammonium chloride (10 mM), or bafilomycin A1 (10 nM). Virus was then added, and each compound was maintained with cells for a further 5 h. If a drug inhibited virus infection, it was expected that this time of incubation would limit cell toxicity but delay infection sufficiently to reduce the virus titer to undetectable levels. The culture medium was then removed, and cells were cultivated for the times indicated below. In each case, the viruses, including RSV, encoded GFP as a marker of infection and the virus titer was determined by counting the GFP-positive cells after virus challenge.
Chlorpromazine was used to block clathrin-mediated endocytosis. Cells were treated as described above, with the indicated concentrations of the drug. Again, a VSV-G-pseudotyped virus was used as a clathrin-dependent control. 10A1 MLV was previously reported to be unaffected by chlorpromazine, and so, a pseudotype of this virus expressing GFP as a marker of infection was used to control for drug toxicity.
siRNA library.
The siRNA library used was a subset of the membrane-trafficking siRNA library (RTF H-005500; Dharmacon). Pools of four siRNAs targeting each gene were initially tested. A pooled approach allowed the identification of potentially important genes without requiring the testing of each siRNA individually. The other advantage is that combined siRNAs may work synergistically by targeting distinct regions of the same mRNA or multiple-splice variants. Since individual siRNA dosage can be reduced, off-target effects are minimized while maintaining specific gene suppression (
27).
To overcome issues of gene compensation that would mask the effect of an isotype-specific siRNA pool, genes were chosen that had two or fewer known gene isotypes or variants of overlapping function. The exception was Rab5a, -b, and -c, for which all three isoform-specific siRNAs were used in the screen. Other gene variants, such as dynamins 1, 2, and 3 and WASF1, -2, and -3, have distinct functions in different cellular compartments and for this reason were also included (
23,
51). The 79 targeted genes (excluding the control set) are listed in Table
1.
Reverse transfection and optimization of siRNA delivery.
To optimize siRNA transfection of HeLa cells, a VSV-G-pseudotyped lentivirus expression vector was used initially to generate a population of cells with stable firefly luciferase expression. These cells were then transfected with various doses of DharmaFECT 1 transfection reagent (Dharmacon) together with siRNA-targeting luciferase or a nontargeting siRNA. From this preliminary work, conditions were identified (indicated below) that produced consistent suppression of 95% of luciferase activity in HeLa cells without noticeable cell toxicity. The siRNA suppression was evident 1 day after transfection and peaked on day 2, reaching a plateau that lasted until day 4, after which it subsided (not shown). Therefore, cells were infected with virus 2 days after transfection and infection was measured on day 3.
All transfections were performed in a 96-well format. A 1.6% (vol/vol) stock of DharmaFECT 1 transfection reagent was made in Dharmafect cell culture reagent (Dharmacon) and incubated for 10 min. From this stock, 25 μl was added to the lyophilized siRNA in each well and incubated at room temperature for 30 min to allow the siRNAs to rehydrate and form siRNA-lipid complexes. Then, 104 cells in 100 μl of complete Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum was added and the medium was changed after 24 h. The final concentration of pooled siRNAs per well was 6.25 pmol. Individual siRNA duplexes were also used at 6.25 pmol per well.
Screening of the siRNA library.
For screening, recombinant RSV that expressed GFP as a marker of infection was used (
61). After HeLa cells were transfected with siRNA, virus was introduced at an MOI of 0.05. GFP expression was evident at 20 h postinfection, at which time monolayers were photographed (4 images per well) using a 10× lens objective. The impact of each siRNA on virus infection was analyzed by counting the cells expressing GFP. The controls included in each experiment were the use of transfection reagent alone, a nontargeting siRNA (Dharmacon), a red fluorescent nonspecific siRNA (siGLO, Dharmacon), and siRNA pools targeting glyceraldehyde-3-phosphate dehydrogenase or lamin A/C. The
kif11 gene product is required for cell survival, and siRNAs targeting this gene are cytotoxic. Therefore,
kif11 siRNA provided a measure of siRNA efficacy by measuring cell death. The other siRNAs served as negative controls for nonspecific effects of siRNA and/or the transfection reagents on cell viability and virus infection. For screening, the number of infected cells for each siRNA treatment was normalized to the average number of infected cells for the negative-control siRNAs. In all cases, cell viability was checked by visually inspecting cells using phase-contrast microscopy.
Use of dominant-negative forms of endocytic-trafficking mediators and virus challenge.
Plasmids encoding wild-type, dominant-negative, or constitutively active mutant forms of proteins important in endosomal trafficking were cloned into the mammalian expression plasmid pLENTI6 (Invitrogen). Each gene was tagged with GFP and has been characterized elsewhere (
30). The mutant genes included Eps15 Δ95-295 (a component of the AP2 clathrin adapter complex; the dominant-negative form inhibits clathrin-coated pit budding); Rab5 S34N (a dominant-negative form that inhibits early endosome formation); and Rab5 Q79L (a constitutively active Rab5 that drives the formation of enlarged early endosomes). To control for the effects of GFP expression on infection, a GFP-expressing plasmid was used.
Each expression construct was transfected into HeLa cells, at 20% confluence, using calcium phosphate. After overnight incubation, the medium was replaced. RSV, VSV or recombinant 10A1 MLV were added to cells 36 h after transfection and incubated with cells for 1 h. Unbound virus was removed by replacing the medium twice. RSV-infected cells were incubated for 20 h, VSV-challenged cells were incubated for 7 h, and 10A1 MLV-infected cells were incubated for 36 h before detection of virus infection.
Cell surface staining for detection of virus infection by FACS.
Virus infection of cells was detected by FACS analysis after staining cells for the expression of virus-specific envelope proteins or CD4.1 infection reporter for 10A1 MLV. The cells were washed twice in phosphate-buffered saline (PBS) and detached from the plate with 5 mM EDTA in PBS. The cells were fixed in freshly made 4% (wt/vol) paraformaldehyde (buffered to pH 7.4 with PBS) for 5 min, then washed in PBS and incubated for 30 min in PBS supplemented with 1% bovine serum albumin to block nonspecific binding of antibodies. All incubations with antibodies were performed in PBS and 1% bovine serum albumin. For RSV and VSV, cells were incubated for 1 h with virus-specific antibody at a 1:200 dilution, and then washed three times and incubated for 30 min with secondary Alexa Fluor647-labeled anti-mouse immunoglobulin G (Invitrogen) at a 1:500 dilution. For the recombinant 10A1 MLV, cells were directly stained with an anti-CD4 Alexa Fluor647 conjugate (Becton Dickinson, NJ) as recommended by the manufacturer. The cells were washed three times and analyzed by using a FACS.
FACS analysis.
Stained cells were analyzed on a FACSCanto instrument (Becton Dickinson). At least three independent experiments were performed, and at least 5 × 104 cells were sampled each time. The cells were gated based on forward and side light scatter to ensure the inclusion of individual cells only, without any debris or aggregates. To analyze the impact of recombinant gene expression, cells were gated by the expression level of the GFP-tagged gene, using a 488-nm laser as an excitation source. The transfection conditions were optimized to give approximately 20% of cells expressing the GFP-tagged protein at moderate to high levels. The cells were stained as described above to detect virus infection and then scanned with a 633-nm laser. As a control for cellular autofluorescence, HeLa cells were transfected with a plasmid vector encoding β-galactosidase. As a control for the contribution of debris in the virus inoculum and for nonspecific antibody binding to cells, infected cells were stained prior to the expression of virus envelope proteins (3, 6, or 12 h after infection with VSV, RSV, or recombinant 10A1 MLV, respectively). The use of the two different lasers (488 and 633 nm) for the excitation of fluors prevented problems of signal contamination from one fluorescence channel into the next. All data were analyzed by using FlowJo 7.1 software from Tree Star Inc. (Ashland, OR).
Counting of cell-bound virus particles.
The binding of virus to cells was optimized to permit the visualization of distinct punctate staining on the cell surface. Virus was concentrated by pelleting through 20% sucrose in 20 mM HEPES, pH 7.4. The concentrated virus was then bound to cells by incubation for 2 h at 4°C to prevent cellular endocytosis. The cells were then washed twice in ice-cold PBS and fixed in paraformaldehyde as described above for FACS experiments. The cells were then stained with anti-RSV F antiserum (1:200 dilution), followed by Alexa Fluor
594-labeled secondary antibody (1:200 dilution). Confocal microscopy was performed using a Zeiss LSM 510 META laser-scanning confocal microscope. At an MOI of 10, distinct virus particles were visible. To count the virus particles bound to cells, uncompressed TIF images were analyzed by the Wright Cell Imaging Facility (WCIF) version of the ImageJ software package (
http://www.uhnresearch.ca/facilities/wcif/imagej/ ). Counting of virus particles was performed on a cell-to-cell basis using the “analyze particles” function and was normalized to the total area of the cell. The margins of each cell were defined manually by referring to matched visible light phase-contrast images and using the “region of interest” function of the software.
Statistics.
Statistical analysis was performed using GraphPad Prism version 4.00 for Windows (GraphPad Software, San Diego, CA). Data were compared by one-way analysis of variance, and analysis included the Tukey-Kramer posttest.
DISCUSSION
We conclude that RSV is an enveloped virus that requires clathrin-mediated endocytosis but not endosomal acidification to infect cells. The studies established this observation with multiple corroborating experimental approaches, and concomitantly determined the value of targeted siRNA profiling to extend our understanding of viral pathogenesis. The concepts that are discussed may be relevant to other pH-independent viruses whose entry and productive infection requirements are not fully defined. Few other examples exist of pH-independent enveloped viruses that use endocytosis to efficiently infect cells. Human immunodeficiency virus (HIV) can inefficiently infect cells after being endocytosed (
12), but entry still appears to occur predominantly at the cell surface (
49). Until recent work was published, avian leucosis virus, a retrovirus afflicting birds, was also thought to enter at the cell surface because, as for other retroviruses, inhibitors of endosomal acidification did not block infection. It was not until a close scrutiny of infection kinetics that a pH-dependent step required after receptor engagement was revealed (
14,
47). However, we do not believe that RSV is the same, since the parameters of infection were set to specifically detect any lag in infection due to effects of the lysosomotropic drug treatment. It is now important to determine if RSV is an exception or if other paramyxoviruses share this pathway of entry and require endocytosis before infection can take place. This knowledge will help in the development of better agents for the treatment of paramyxovirus infection.
Aside from pH, few other endosomal triggers of virus entry into cells have been identified. Even the recently reported cathepsin cleavage of the Ebola virus envelope protein, required before virus entry can take place, uses a pH-dependent protease (
10). A few reports indicate that disulfide bond rearrangement may be important in HIV entry, but this is still believed to occur on the cell surface (
18). Thus, pH insensitivity of a virus is often equated with lack of need for endocytosis. The ability to form syncytia at neutral pH is also considered to be a strong indicator of competence for entry at the plasma membrane. This demonstrates that the envelope proteins are capable of fusing cell and virus membranes at the cell surface, but it does not indicate if such fusion leads to a productive virus infection. RSV drives syncytium formation at neutral pH (
48) and was shown to have a pH-insensitive infection mechanism in HEp-2 cells (
48). We showed that the infection of HeLa cells was similarly unaffected by the endosomal acidification inhibitors ammonium chloride, chloroquine, and bafilomycin A1. Historically, these observations have been interpreted to mean that RSV entered at the cell surface.
Recently, it was reported that internalization of receptor-bound MLV (a pH-insensitive retrovirus that is thought to enter at the cell surface) may precede entry and this was likely to be actin and myosin dependent (
31). We wanted to know if RSV showed a similar dependence on actin function. Indeed, RhoA activity, an activator of actin polymerization, was shown to be required for syncytium formation by RSV (
21). Also, actin disruption using cytochalasin inhibited RSV uptake in dendritic cells (
59). Unfortunately, these studies did not identify the actual role of actin in RSV infection itself. In the current work, actin-associated genes were identified as important for RSV infection but the identified genes were those predominantly associated with endocytosis.
Genes important in actin polymerization were ARRB1, ARPC2, DDEF2, PAK1 (also termed PAK-alpha), and WASF2 (WAVE2). Each is important in coupling clathrin-mediated endocytosis to actin. β-Arrestin (ARRB1) is involved in the desensitization of receptors by targeting them to clathrin-coated vesicles through a RhoA and actin-dependent mechanism (
3). WASF2 is an important down-stream effector of receptor-mediated signaling that triggers actin polymerization. WASF family members also play important roles late in clathrin-coated pit formation by coupling to the ARP2/3 actin-regulating complex and may act to move the vesicle through the cell (
35). This suggests a potential role of these genes in regulating RSV endocytosis after the binding of RSV receptors.
Consistent with a key role for endocytosis in RSV entry, specific siRNAs targeting genes known to be important in endocytosis itself were indicated. Most notable were those required for the assembly of clathrin-coated pits, including those encoding AP-2 subunits and clathrin light chain B. The alpha and beta subunits of AP-2 are important for clathrin-coated pit formation and are involved in clustering receptors to pit invaginations on the cell membrane. Clathrin light chains A and B are the main protein coating the cytoplasmic faces of clathrin-coated pits, and they control pit assembly. Together with clathrin heavy chains, the light chains form distinct structures, termed triskelions, upon which the clathrin coat is assembled (
33). Multiple isoforms and variants of each of the clathrin light chain genes are expressed within the same cell at the same time. These variants are believed to have different roles in controlling the endocytosis kinetics and possibly the origins and destinations of cargoes (
33).
The role of clathrin-mediated endocytosis was independently confirmed by chlorpromazine treatment of cells and by transfecting cells with a well-characterized dominant-negative mutant form of Eps15. Chlorpromazine treatment is reported to be a specific inhibitor of clathrin-mediated endocytosis (
57), while Eps15 is a critical component of clathrin pit formation and interacts with both AP2-alpha and -beta (
45). The dominant-negative form of Eps15 potently blocks the uptake of labeled transferrin by clathrin-mediated endocytosis but does not block other markers that are taken up by caveolae or macropinocytosis (
4). Both the drug treatment and the expression of dominant-negative Eps15 were effective inhibitors of RSV infection. The major technical hurdle in using this and other dominant-negative genes is that the phenotypes are dosage dependent and require moderate- to high-level expression in cells. In the current work, FACS analysis was used to overcome this limitation while providing statistical power to the analysis. The results of the FACS analysis confirmed what was suggested by microscopy and showed a strong correlation between Eps15 Δ95-295 expression and a statistically significant decrease in the level of RSV infection. The drop in infection seen with chlorpromazine and the mutant Eps15 was similar to that seen for VSV, a virus characterized as using clathrin-mediated endocytosis (
52). This observation, together with the siRNA data, strongly indicates a key role for clathrin function in RSV infection. However, this evidence alone does not indicate that endocytosis is also important, since clathrin is involved in other cellular functions, such as mitosis (
43).
The confirmation that endocytosis was required for RSV infection was obtained by using siRNAs and dominant-negative mutants targeting the endocytic machinery itself. The primary siRNA screen indicated that targeting of the endocytosis-regulating proteins ITSN1 and -2, dynamin 3, and Rab5A each resulted in significant decreases in infection. Except for ITSN1, each was confirmed in the secondary screen using individual siRNAs instead of pools. Both the dynamin 3 suppression and Rab5 suppression were confirmed by Western blotting. Dynamins are involved in pinching off endocytic vesicles, including clathrin-coated pits and caveolae, from the cell membrane (
13,
23). Intersectins (ITSN) bind dynamin and are involved in regulating clathrin-mediated endocytosis (
40). Rab5 directly controls the formation of early endosomes. The dominant-negative mutant forms of Rab5 confirmed a need for early endosome formation for RSV infection. Interestingly, while the dominant-negative form suppressed infection by both RSV and the VSV control, the constitutively active form of Rab5 (Q79L) did not affect RSV infection greatly. The difference between these two mutant proteins of Rab5 is that the negative form blocks early endosome formation, whereas the active form enhances early endosome formation but disrupts downstream maturation steps. These observations then suggest that, unlike VSV, RSV does not require steps immediately after early endosome formation or that another pathway not disrupted by the active mutant takes over. Caveolae are one such candidate. These endocytic vesicles intersect with the clathrin-dependent endosomes at the early endosome and play a role in JC virus infection (
42). Indeed, siRNA targeting caveolin 2 had some effect on RSV infection but fell below the threshold of significance for the screen. However, the great preponderance of siRNAs targeting clathrin-related genes that had significant effects on infection argues that if caveolae play a part in RSV infection, it must be at a step after the initial virus uptake into the cell. Additional studies with specific inhibitors of late endosomes, caveolae, and other early endosome markers will help to resolve this issue and determine the exact point of RSV exit from the endosomal pathway.
In this work, siRNAs that reduced infection were focused on and studied. Interestingly, treatment with some siRNAs elevated infection levels by as much as two- to threefold and included siRNAs targeting Stau, Bin1, AP1M2, and Rab7B genes. Stau is involved in RNA association with the cytoskeleton and could be important for the subcellular localization of the viral genome. Bin1 (amphiphysin 2) isoforms are involved in numerous processes, including endocytosis and cell cycle regulation (
58), and AP1M2 is implicated in the regulation of clathrin-mediated endocytosis and for the basolateral sorting of receptors (
17). Rab7 controls the maturation of late endosomes from early endosomes. Such genes, which raise the level of infection when suppressed, could be part of a host innate immune response against the virus, and could include PKR, which is an important mediator of the interferon response against RNA viruses (
2), or APOBEC, which degrades retroviral nucleic acids upon entry into the cell cytoplasm (
34). Suppressing the expression of each of these genes would be predicted to result in increased infection. However, the genes in the present screen are unlikely to be directly involved in known innate immunity pathways. Instead, it is more likely that at least Bin1 and AP1M2 are involved in regulating endocytic pathways that misdirect virus into parts of the cell that are not conducive for replication. Since Rab7 is key for late endosome formation, it is likely that reaching this compartment is deleterious to RSV. Consistent with this was the finding that the constitutively active Rab5 Q79L mutant, which blocks the maturation of early into late endosomes, did not inhibit RSV infection.
Among the siRNAs that targeted one of several isoforms or gene variants, typically one variant appeared more important for virus infection than others. Of the three Rab5 isoforms, three dynamins, three WASF variants, and two clathrin light chains targeted, only the Rab5A, dynamin 3, WASF2, and clathrin light chain B siRNA pools reduced infection below the cutoff threshold for the primary screen. The different activities of the Rab5, dynamin, and clathrin light chain variant-specific siRNAs were not due to lack of endogenous protein expression in the HeLa cells or lack of siRNA efficacy, since Western blots for at least Rab5A and 5B, as well as dynamin 2 and 3 and clathrin light chain A and B, were expressed and reduced to similar extents after siRNA treatment. Instead, the distinction may be that RSV prefers a pathway controlled by clathrin light chain B, dynamin 3, and Rab5A for optimal infection. Other similar pathways may operate in parallel, but subtle differences in the destination or the path taken by each may preclude virus from targeting sites needed for replication. Indeed, clathrin light chain A and B have been shown to have different kinetics of vesicle association and may control trafficking to different sites within the cell (
1). This level of selectivity of a ligand for a specific gene isoform is not typical. In previous work, studying the endocytic trafficking requirements of the EGF receptor, only after suppression of the expression of all three isoforms of Rab5 was EGFR trafficking interrupted (
25). It has been suggested by others that well-characterized viruses such as VSV and simian virus 40 may serve as superior probes for dissecting endocytic mechanisms (
39). The current work and that of others indicate that viruses have very specific trafficking requirements. Virus-encoded reporter assays also have the advantage of signal amplification. Taking advantage of these strengths, we demonstrate that it is also possible to use an siRNA approach to define the entry mechanism of an uncharacterized virus such as RSV.
The current work highlights the power of using siRNAs to identify new genes and pathways important for pathogen infection. We chose to focus on a subset of genes important in endocytosis and actin rearrangement. While the screen did not exhaustively target all such genes, it had the advantage of yielding information for a workable set of genes that could be confirmed through independent tests. This approach and the methods of validation used in these studies were recently recommended by experts in the RNA interference (RNAi) field to limit the reporting of false positives from RNAi screens (
16). The activity of siRNA pools was confirmed by testing individual siRNAs targeting the same gene, limiting potential false positives due to off-target effects. The next level of validation demonstrated that protein expression was specifically reduced after siRNA treatment. This portion of the work was limited by the availability of antibodies able to detect endogenous protein in cells. Independent tests using dominant-negative and constitutively active forms of proteins in the endocytic pathway then confirmed endocytosis as important for RSV. Larger (whole genome) siRNA screens will yield many potential genes involved in virus infection but will likely be hindered by off-target effects and means to independently validate hits. However, progress is being made toward increasing siRNA specificity, and an expanded repertoire of dominant-negative genes and specific drugs will make such screens increasingly effective for identifying potential targets of disease intervention.