Abstract
Virus replication strongly depends on cellular factors, in particular, on host proteins. Here we report that the replication of the arteriviruses equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) is strongly affected by low-micromolar concentrations of cyclosporine A (CsA), an inhibitor of members of the cyclophilin (Cyp) family. In infected cells, the expression of a green fluorescent protein (GFP) reporter gene inserted into the PRRSV genome was inhibited with a half-maximal inhibitory concentration (IC50) of 5.2 μM, whereas the GFP expression of an EAV-GFP reporter virus was inhibited with an IC50 of 0.95 μM. Debio-064, a CsA analog that lacks its undesirable immunosuppressive properties, inhibited EAV replication with an IC50 that was 3-fold lower than that of CsA, whereas PRRSV-GFP replication was inhibited with an IC50 similar to that of CsA. The addition of 4 μM CsA after infection prevented viral RNA and protein synthesis in EAV-infected cells, and CsA treatment resulted in a 2.5- to 4-log-unit reduction of PRRSV or EAV infectious progeny. A complete block of EAV RNA synthesis was also observed in an in vitro assay using isolated viral replication structures. The small interfering RNA-mediated knockdown of Cyp family members revealed that EAV replication strongly depends on the expression of CypA but not CypB. Furthermore, upon fractionation of intracellular membranes in density gradients, CypA was found to cosediment with membranous EAV replication structures, which could be prevented by CsA treatment. This suggests that CypA is an essential component of the viral RNA-synthesizing machinery.
INTRODUCTION
The replication of RNA viruses strongly depends on their successful interplay with the host cell at multiple levels. By now, a wide variety of host cell proteins have been implicated in RNA virus replication, and some of these might, in fact, constitute interesting targets for antiviral therapy (1). Thus, the possibility to target host factors rather than viral proteins is receiving increasing attention as an alternative and promising antiviral approach (reviewed in references 2 and 3). In contrast to antiviral therapy that aims to inhibit viral protein functions, the use of drugs targeting host factors should not lead to drug resistance, which is a common problem when combating RNA viruses, due to their high mutation rate and potential for rapid adaptation.
The drug cyclosporine (CsA) was previously found to inhibit the replication of a number of RNA viruses (4–8). Recently, multiple laboratories, including our own, reported that the replication of various (human) coronaviruses, including the severe acute respiratory syndrome coronavirus (SARS-CoV), can also be inhibited by CsA treatment (9–11). This drug affects the function of several members of the cellular cyclophilin (Cyp) protein family, which consists of peptidyl-prolyl isomerases (PPIases) that act as chaperones to facilitate protein folding and are involved in protein trafficking and immune cell activation (12, 13). Although Cyps share many similarities in terms of structure and activity, important differences in specific functions and subcellular localization have been documented (12). In line with the inhibition of virus replication by CsA, Cyp family members were identified to be essential host factors in the replicative cycle of several virus groups (reviewed in reference 14).
The drug CsA has been widely used as an immunosuppressant, e.g., in organ transplant patients (15), as its binding to various Cyps impairs calcineurin activity and abrogates the T-cell response. As Cyps appeared to be relevant targets for antiviral therapy, several Cyp inhibitors that lack the immunosuppressive properties of CsA, which would be an undesirable side effect during antiviral therapy, have been developed. The efficacy of several such compounds, e.g., Debio-025 and NIM811, is currently being explored in clinical trials for the treatment of hepatitis C virus (HCV) infection (16–18).
Since the replication of coronaviruses like SARS-CoV is inhibited by CsA (9, 10), we investigated whether this drug also inhibits the distantly related arteriviruses, which together with the coronavirus and ronivirus families constitute the order Nidovirales (19). The arterivirus porcine reproductive and respiratory syndrome virus (PRRSV) is one of the leading veterinary pathogens, causing an estimated annual loss of $664 million in the swine industry in the United States alone (20). Equine arteritis virus (EAV), in addition to being a relevant horse pathogen, has been used for decades as a model to dissect the molecular details of arterivirus and nidovirus replication (21).
Arteriviruses are positive-stranded RNA viruses with a genome size of about 13 to 16 kb (22). Their complex genome expression strategy involves genome translation to produce the polyprotein precursors for the viral nonstructural proteins (nsps) as well as the synthesis of a nested set of subgenomic (sg) mRNAs to express the structural proteins (23). The viral nsps, presumably together with various host factors, are thought to assemble into membrane-associated replication and transcription complexes (RTCs) that drive viral RNA synthesis (for recent reviews, see references 22 and 24). Arterivirus RNA synthesis was reported to be associated with a virus-induced network of endoplasmic reticulum (ER)-derived membrane structures, including large numbers of double-membrane vesicles (25). Many arteriviral proteins were found to be associated with these membrane structures, on which viral RNA synthesis was found to depend (26, 27). However, thus far, the identity and role of proviral host factors involved in the replicative cycle of arteriviruses have remained largely unexplored.
Using EAV and PRRSV, our studies on the inhibition of nidovirus replication by CsA have now been extended to arteriviruses and explored the mechanism of action of the compound in more detail. We show that low-micromolar concentrations of CsA can fully block arterivirus RNA synthesis and that the nonimmunosuppressive cyclophilin inhibitor Debio-064 is an even more potent inhibitor. These compounds probably exert their effect through their inhibition of CypA, as RNA interference (RNAi)-mediated knockdown of CypA strongly affected EAV RNA synthesis and CypA was found to cosediment with EAV replication structures.
MATERIALS AND METHODS
Cell culture, infection, and virus titration.
BHK-21 (28), Vero E6 (29), and MARC-145 (30) cells were cultured as described previously. 293/ACE2 cells (31) were cultured in Dulbecco's modified Eagle's medium (DMEM; Lonza) supplemented with 8% fetal calf serum, 100 U/ml of penicillin, 100 μg/ml of streptomycin, 2 mM l-glutamine, and 12 μg/ml blasticidin (PAA Laboratories). A cell culture-adapted derivative of the EAV Bucyrus isolate (32) and green fluorescent protein (GFP)-expressing recombinant EAV (33) were used to infect monolayers of BHK-21, Vero E6, and 293/ACE2 cells at a multiplicity of infection (MOI) of 5, as described previously (28, 29). MARC-145 cells were infected with a GFP-expressing recombinant PRRSV (SD01-08-GFP) at an MOI of 0.1, as previously described (34). EAV titers in cell culture supernatants were determined by plaque assay on BHK-21 cells (28), whereas PRRSV titers were determined by fluorescent focus assay (FFA) on MARC-145 cells, as described previously (35). For half-maximal inhibitory concentration (IC50) determinations, cells were grown in black 96-well plates (Greiner), infected with EAV-GFP or PRRSV-GFP, and treated with compounds in octuplet. GFP reporter expression was quantified by measuring the fluorescence in a 96-well plate reader, using an excitation wavelength of 485 nm and an emission wavelength of 535 nm.
Antibodies and drugs.
Rabbit polyclonal antibodies against CypA (Abcam), CypB (Abcam), and calnexin (BD), a goat polyclonal antiserum against GAPDH (glyceraldehyde-3-phosphate dehydrogenase; Santa Cruz), and a mouse monoclonal antibody (MAb) against β-actin (Sigma) were used. Rabbit antisera recognizing the EAV replicase subunits nsp3 (36) and nsp9 (27) and the EAV membrane (M) protein (29) and a MAb against the EAV nucleocapsid (N) protein (37) have been described previously. The cyclophilin inhibitors CsA (Sigma) and Debio-064 (Debiopharm, Switzerland) were dissolved in dimethyl sulfoxide (DMSO). CsA was stored as a 50-mg/ml stock at −20°C, and Debio-064 was stored as a 10 mM stock at 4°C in aliquots for single use. The IC50 of inhibitors was calculated with GraphPad Prism (version 5) software using a nonlinear regression model.
Immunofluorescence microscopy.
EAV-infected or mock-infected BHK-21 cells, grown on coverslips at 39.5°C, were fixed with 3% paraformaldehyde in phosphate-buffered saline (PBS), permeabilized with 0.1% Triton X-100, and processed for immunofluorescence microscopy as described previously (26). Specimens were examined with a Zeiss Axioskop 2 fluorescence microscope with an Axiocam HRc camera and Zeiss Axiovision (version 4.4) software.
Western blot analysis.
After SDS-PAGE, proteins were transferred to Hybond-LFP membranes (GE Healthcare) by semidry blotting. Membranes were blocked with 1% casein in PBS containing 0.1% Tween 20 (PBST) and were incubated with anti-nsp3 (1:2,000), anti-nsp9 (1:2,000), anti-M (1:2,000), anti-N (1:10,000), anti-CypA (1:1,000), anti-CypB (1:2,000), or anti-β-actin (1:50,000) antiserum diluted in PBST with 0.5% casein. Biotin-conjugated swine anti-rabbit IgG (1:2,000) or goat anti-mouse IgG (1:1,000) antibodies (Dako) and Cy3-conjugated mouse anti-biotin (1:2,500) antibodies were used for detection. Blots were scanned with a Typhoon 9410 imager (GE Healthcare) and analyzed with ImageQuant TL software.
Isolation of EAV RTC-containing replication structures and in vitro RNA synthesis assays.
EAV replication structures were isolated from BHK-21 or Vero E6 cells, and in vitro RNA synthesis assays were performed essentially as described previously (27). In short, approximately 1 × 108 EAV-infected BHK-21 or Vero E6 cells were harvested at 6 or 7 h postinfection (p.i.), and cells were lysed to obtain a postnuclear supernatant (PNS) (27). A standard in vitro RNA synthesis assay mixture contained 20 μl of PNS (the equivalent of 6 × 104 cells) from EAV-infected BHK-21 cells, 5 μl of an inhibitor solution, or 5 μl of RTC dilution buffer (control). Following gel electrophoresis, 32P-labeled reaction products were analyzed by denaturing agarose gel electrophoresis and by exposing a PhosphorImager screen directly to the dried gel, after which screens were scanned with a Typhoon 9410 imager (GE Healthcare), and incorporation of label was quantified using ImageQuant TL software.
Density gradient fractionation.
Subcellular fractionation of PNS was performed in continuous 0 to 30% OptiPrep density gradients in RTC dilution buffer. The gradients were prepared in 13.2 ml Ultra-Clear centrifugation tubes (Beckman Coulter) using a Gradient Master gradient former (Biocomp). One milliliter of PNS from Vero E6 cells was carefully loaded on top of the preformed gradient. After centrifugation for 17 h at 48,000 × g in an SW41 rotor at 4°C, the gradient was fractionated into 0.5-ml fractions. The density of each fraction was determined with a refractometer (GETI).
Metabolic labeling of viral RNA synthesis.
Labeling of viral RNA with [3H]uridine was performed essentially as described previously (38). Briefly, at 4.5 h p.i., 4 × 105 EAV-infected BHK-21 cells in 4-cm2 dishes were given medium containing 10 μg/ml actinomycin D (ActD; Sigma-Aldrich) and either 4 μM CsA or 0.01% DMSO as a solvent control. After 1 h, viral RNA synthesis was labeled by adding 100 μCi of [3H]uridine to the medium. The 3H-labeled RNAs were isolated, separated in denaturing agarose gels, and visualized by fluorography. To verify that equal amounts of total RNA were loaded, the gel was hybridized with a 32P-labeled oligonucleotide probe (5′-TTCACGCCCTCTTGAACTCTCTCTTC-3′) recognizing 28S rRNA, as described previously (27).
RNA interference.
ON-TARGETplus smart-pool small interfering RNA (siRNA) duplexes (Dharmacon) against CypA (PPIA; catalog no. L-004979-04) and CypB (PPIB; catalog no. L-004606-00) were used to silence CypA and CypB expression in 293/ACE2 cells. A nontargeting siRNA (D-001810-10) was used as a control, and a GAPDH-targeting siRNA (D-001830-10) was used to monitor transfection and knockdown efficiency. Stock solutions of 2 μM were prepared by dissolving siRNAs in 1× siRNA buffer (Dharmacon). For transfection of cells in 96-well clusters, 1 × 104 293/ACE2 cells per well were transfected with a 100-μl mixture containing 100 nM siRNA, 0.2 μg DharmaFECT1 transfection reagent (Dharmacon), Opti-MEM reduced-serum medium (Invitrogen), and antibiotic-free culture medium, according to the manufacturers' instructions. For cells in 12-well clusters, 600-μl transfection mixtures were used. Medium was replaced at 24 h posttransfection (p.t.) by antibiotic-free culture medium, and at 48 h p.t., cells were infected with EAV-GFP or wild-type (wt) EAV. Duplicate cultures were used either to prepare lysates to analyze protein expression levels or to monitor cell viability using a CellTiter 96 AQueous nonradioactive cell proliferation assay (Promega), according to the manufacturer's instructions.
RESULTS
EAV-GFP and PRRSV-GFP replication is inhibited by CsA.
The effect of CsA on arterivirus replication was investigated in cell culture using two representatives of the arterivirus family, EAV and PRRSV (European genotype). For the initial experiments, we employed GFP-expressing recombinant viruses, since quantification of GFP expression provides a rapid and reliable method to detect inhibition of virus replication. BHK-21 cells were grown in 96-well plates and infected at an MOI of 5 with GFP-expressing recombinant EAV (33). Upon removal of the inoculum (at 1 h p.i.), medium containing 0.03 to 4 μM CsA was given, and at 18 h p.i., cells were fixed and GFP expression was quantified. We observed a strong dose-dependent inhibition of EAV-GFP replication (Fig. 1A) in the absence of significant cytotoxic effects at the CsA concentrations used (Fig. 1B). The IC50 of CsA for EAV-GFP replication in BHK-21 cells was determined to be 0.95 μM.
Fig 1.
Cyclosporine treatment blocks EAV-GFP and PRRSV-GFP replication. (A) BHK-21 cells were infected with EAV-GFP at an MOI of 5, and at 1 h p.i., the inoculum was replaced by medium containing different concentrations of CsA, as indicated on the x axis. Cells were fixed at 18 h p.i., and GFP reporter expression was quantified and normalized to the GFP signal of control cells (100%) treated with DMSO (the solvent concentration was equal to that in cultures that received 4 μM CsA). (B) The effect of CsA on cell viability compared to the viability of untreated control cells (100%) was determined using a CellTiter 96 AQueous nonradioactive cell proliferation assay (Promega). Graphs show the results (average and SD) of a representative experiment performed in quadruplicate. (C) MARC-145 cells were infected with PRRSV-GFP at an MOI of 0.1, and at 1 h p.i., the inoculum was replaced by medium with CsA. Cells were fixed at 24 h p.i., and GFP reporter expression was quantified and normalized to the signal in solvent-treated control cells (100%). (D) Effect of CsA on the viability of the MARC-145 cells compared to the viability of untreated control cells (100%). Graphs show the results (average and SD) of a representative experiment (n = 8). All experiments were repeated at least twice.
A similar experiment was performed with PRRSV-GFP in MARC-145 cells (Fig. 1C). Although less sensitive to CsA treatment than EAV-GFP, the replication of PRRSV-GFP was completely blocked at 16 μM CsA, and an IC50 of 5.22 μM was determined. Cell viability was only slightly affected by CsA concentrations above 4 μM (Fig. 1D).
The cyclophilin inhibitor Debio-064 blocks EAV-GFP and PRRSV-GFP replication.
Although CsA has been found to effectively block the replication of various RNA viruses in cell culture (14), its use in antiviral therapy would be complicated by the immune suppression (39) that is a major side effect. Therefore, several alternative Cyp inhibitors that lack the immune-suppressive properties of CsA, like SCY-635, NIM811, and Debio-025, which all block HCV replication, have been developed (17, 18, 40).
In this study, we tested whether the nonimmunosuppressive Cyp inhibitor Debio-064 is able to block EAV-GFP and PRRSV-GFP replication (Fig. 2A). Debio-064 is a structurally modified cyclosporine exhibiting an approximately 5-fold higher affinity for CypA than CsA. Debio-064 is 300-fold less active than CsA at inhibiting mouse T-cell proliferation induced by concanavalin A, suggesting that the compound does not inhibit calcineurin (41). EAV-GFP-infected BHK-21 cells (Fig. 2B) or PRRSV-GFP-infected MARC-145 cells (Fig. 2D) were treated with various noncytotoxic concentrations of Debio-064, and viral replication was quantified as described for CsA treatment. Compared to CsA, Debio-064 had a stronger inhibitory effect on EAV-GFP replication. At a concentration of 0.5 μM Debio-064, the EAV-GFP signal was hardly detectable (Fig. 2A), and an IC50 of 0.29 μM, which is about 3-fold lower than that of CsA, was determined. For PRRSV-GFP, an almost complete block in GFP expression was observed at 8 μM, and an IC50 of 5.14 μM (Fig. 2C), which is comparable to the inhibitory effect of CsA on PRRSV-GFP replication, was determined.
Fig 2.
The CsA analog Debio-064 blocks EAV-GFP and PRRSV-GFP replication. (A) EAV-GFP-infected BHK-21 cells (MOI, 5) were incubated with various concentrations of Debio-064 from 1 h p.i. onwards. Cells were fixed at 18 h p.i., and GFP reporter expression was quantified and normalized to the GFP signal of control cells (100%) that were treated with DMSO (the solvent concentration was equal to that in medium containing 4 μM Debio-064). (B) Effect of Debio-064 on BHK-21 cell viability compared to the viability of untreated control cells (100%). (C) PRRSV-GFP-infected MARC-145 cells (MOI, 0.1) were incubated with various concentrations of Debio-064 from 1 h p.i. onwards. Cells were fixed at 24 h p.i., and GFP reporter expression was quantified and normalized to the GFP signal of control cells (100%) that were treated with DMSO. (D) Effect of Debio-064 on MARC-145 cell viability compared to the viability of control cells (100%). Results (average and SD) of a representative experiment performed in quadruplicate are shown, and all experiments were repeated at least twice.
CsA and Debio-064 prevent arterivirus protein expression.
In our initial experiments, we tested the effect of CsA on the replication of a GFP-expressing recombinant EAV. To verify that wt EAV replication could also be inhibited by the drug, we analyzed wt EAV-infected BHK-21 cells that were treated with 0.25 to 8 μM CsA. At 6 h p.i., cells were lysed and lysates were subjected to Western blot analysis. The expression of viral nonstructural proteins (the nsp5-8 precursor and nsp9) and the structural M and N proteins was hardly detectable after 4 μM CsA treatment, while a clear reduction in protein expression could already be observed at 2 μM CsA (Fig. 3A). As observed for EAV-GFP, Debio-064 had a stronger inhibitory effect than CsA on the replication of wt EAV. Viral protein expression was clearly reduced in the presence of 0.5 μM the drug and became almost undetectable at 1 μM Debio-064 (Fig. 3B).
Fig 3.
CsA and Debio-064 treatment block viral protein expression in cells infected with wild-type EAV. BHK-21 cells were infected with EAV (MOI, 5) and treated from 1 h p.i. on with CsA (A) or Debio-064 (B) at the concentration indicated above each lane. Cells were lysed at 6 h p.i., and viral protein expression was analyzed by SDS-PAGE and Western blotting with antibodies against nsp9 and nsp5-8, the M protein, and the N protein. β-Actin was used as a loading control. For immunofluorescence microscopy, mock-infected or EAV-infected and CsA-treated (C) or Debio-064-treated (D) cells were fixed at 6 h p.i. and stained with an anti-nsp3 antiserum. The drug concentrations used are indicated in each panel. Bars, 50 μm.
The effect of CsA and Debio-064 was further characterized by immunofluorescence microscopy of infected cells. For wt EAV, double-stranded RNA (dsRNA) (data not shown) and viral proteins were undetectable after a dose of 4 μM CsA (Fig. 3C) or 1 μM Debio-064 (Fig. 3D). In the case of PRRSV-GFP-infected MARC-145 cells (data not shown), maximal inhibition was observed at a 16 μM CsA dose. However, as previously observed for coronavirus-infected Vero E6, 17Cl1, or Huh7 cells (9), a small fraction of the MARC-145 cells remained capable of supporting PRRSV-GFP replication, even at high CsA doses of up to 64 μM (data not shown). We did not observe such a small, residual population of apparently drug-insensitive cells in EAV-infected BHK-21 cultures treated with either CsA or Debio-064 (Fig. 3C and D).
CsA and Debio-064 block the production of arterivirus infectious progeny.
To assess the extent to which CsA and Debio-064 treatment affected infectious progeny titers, we performed plaque assays to measure EAV titers at 12 h p.i. using supernatants from infected (MOI, 5) BHK-21 cells that had been treated with CsA or Debio-064 (Fig. 4A). CsA strongly reduced EAV progeny titers, with an almost 4-log-unit reduction at 4 μM CsA. Treatment probably completely abolished virus production, as the titers observed after treatment with 4 μM CsA were similar to those measured at 1 h p.i., which likely reflected the remainder of the high-MOI inoculum used (data not shown). These data correlated well with the barely detectable expression of the nsp5-8, nsp9, and the M and N proteins and the lack of dsRNA after treatment with 4 μM CsA (Fig. 3A and C). Treatment with Debio-064 also resulted in an ∼4-log-unit reduction of infectious progeny at 2 μM, while a 2- to 3-log-unit reduction was already achieved by treatment with 1 μM the compound.
Fig 4.
Treatment of infected cells with cyclophilin inhibitors strongly reduces arterivirus yields. (A) EAV-infected BHK-21 cells (MOI, 5) were treated with various concentrations of CsA (gray bars) or Debio-064 (white bars) from 1 h p.i. onwards, and virus titers in the culture medium at 12 h p.i were determined by plaque assay. (B) MARC-145 cells infected with PRRSV-GFP (MOI, 0.1) were treated from 1 h p.i. onwards with CsA (gray bars) or Debio-064 (white bars) at the concentrations indicated below the x axis, and virus titers in the medium at 24 h p.i. were determined by fluorescent focus assay.
Using a fluorescent focus assay, we also analyzed the production of PRRSV-GFP infectious progeny in culture supernatants of CsA-treated MARC-145 cells at 24 h p.i. As observed for EAV, the production of PRRSV-GFP infectious progeny was affected by CsA treatment, although significantly higher concentrations were required. At 16 μM CsA, an ∼1.5-log-unit reduction in the yield of infectious progeny was observed, while an apparently complete block (2.5-log-unit reduction) required a dose of 32 μM (Fig. 4B, gray bars). Treatment with Debio-064 resulted in an ∼1.5-log-unit reduction of infectious progeny at 16 μM and an ∼2.5-log-unit reduction of infectious progeny at 32 μM (Fig. 4B, white bars), which are comparable to the reduction in PRRSV progeny observed upon CsA treatment.
Cyclophilin inhibitors affect EAV RNA synthesis both in vivo and in vitro.
The above-described experiments showed that CsA can effectively block both EAV and PRRSV replication in cell culture. To establish that this lack of viral protein synthesis was due to a block of viral RNA synthesis, we measured the effect of CsA treatment on EAV RNA synthesis in infected Vero E6 cells in vivo, by metabolic labeling with [3H]uridine (in the presence of actinomycin D). When 4 μM CsA was given at 4.5 h p.i., 3H incorporation into viral RNA during a pulse-labeling from 5.5 to 6.5 h p.i. was reduced to 8% of the incorporation measured for nontreated control cells (Fig. 5A). To obtain more insight into the mechanism by which CsA inhibits arterivirus replication, we tested its effect in a previously developed in vitro assay to study the RNA-synthesizing activity of semipurified EAV RTCs (27). These assays were performed with PNS from EAV-infected BHK-21 cells, and the reactions, during which 32P-labeled CTP is incorporated into viral RNA, were conducted in the presence of various concentrations of CsA. In the absence of the drug, in vitro synthesis of EAV genomic and sg RNAs was observed (Fig. 5B, lane 1), as documented previously (27). RNA-synthesizing activity was completely abolished when the reaction was performed in the presence of 12 μM CsA (lane 4), while a >50% reduction of viral RNA synthesis was observed in the presence of 8 μM CsA (lane 3). Comparable results were obtained with Debio-064, which also caused a >50% reduction of EAV RTC activity at about 8 μM and a complete inhibition at 16 μM (Fig. 5C, lanes 3 and 5). These data strongly suggest that Cyp inhibitors can directly affect the RNA-synthesizing capacity of the membrane-associated EAV RTCs in PNS samples. We recognize that the concentrations needed to fully block EAV RTC activity in vitro are ∼3-fold higher than those required to block virus replication in cell culture. This might be due to differences in the experimental setup, as the PNS used for the in vitro reaction constituted a concentrated preparation of RTCs (and host factors), and reaction conditions might influence the interaction between Cyps and their inhibitor.
Fig 5.
The in vitro and in vivo RNA-synthesizing activity of EAV RTCs can be blocked by CsA or Debio-064 treatment. (A) Metabolic labeling of EAV-infected cells with [3H]uridine at between 5.5 and 6.5 h p.i. in the presence or absence of 4 μM CsA. Total RNA was isolated, analyzed in denaturing agarose gels, and detected by fluorography. The amount of [3H]uridine that was incorporated into viral genomic RNA was quantified and normalized to that in EAV-infected, untreated control cells (100%). 28S rRNA detected by hybridization with a 32P-labeled probe (bottom) was used as a control to correct for variations in loading during viral RNA quantification. (B and C) Semipurified RTCs isolated from EAV-infected BHK-21 cells at 6 h p.i. were used in an in vitro RNA synthesis assay in which [32P]CTP was incorporated into viral RNA. Reactions, performed in the presence of various concentrations of CsA (B) or Debio-064 (C), as indicated above the lanes, were terminated after 100 min. RNA was isolated, and reaction products were analyzed in denaturing formaldehyde agarose gels. The positions of the genomic RNA (position 1) and subgenomic RNAs (positions 2 to 7) are indicated next to the gels.
EAV replication depends on cyclophilin A.
CsA is known to inhibit the PPIase activity of several members of the cyclophilin family. In particular, CypA and CypB have been implicated in the replication of several viruses (reviewed in reference 14). We therefore analyzed the effect of siRNA-mediated knockdown of CypA and CypB expression levels on the replication of EAV-GFP. We made use of the same human 293/ACE2 cells that we previously used to study the role of Cyps in SARS-CoV replication (9). This cell line was also susceptible to EAV infection, although only ∼40% of the cells became GFP positive after infection with EAV-GFP at a high MOI, as judged by immunofluorescence microscopy of infected cells fixed at 8 h p.i. (data not shown).
Knockdown of CypA and CypB expression was monitored by Western blotting, and an ∼80% reduction of expression compared to the level in control cells transfected with a nontargeting control siRNA was typically observed (Fig. 6A). Depletion of CypA or CypB did not have a significant effect on cell viability during the 48 h of the knockdown experiment (Fig. 6B). Compared to control cells, knockdown of CypB expression did not influence GFP reporter expression when these cells were infected with EAV-GFP, in which GFP fluorescence was measured at 24 h p.i. (Fig. 6C). In contrast, knockdown of CypA resulted in an ∼60% reduction of the GFP signal in EAV-GFP-infected cells compared to that in the control cells (Fig. 6C). Furthermore, at 32 h p.i., wt EAV titers in the culture medium of infected 293/ACE2 cells (MOI, 0.01) that had been depleted for CypA showed an ∼4-fold decrease in virus progeny compared to control cells (Fig. 6D). These data strongly suggest that EAV replication and the production of virus progeny depend on the availability of the host factor CypA.
Fig 6.
RNAi-mediated knockdown of CypA, but not CypB, strongly affects EAV replication. 293/ACE2 cells were transfected with a nontargeting control siRNA or siRNAs targeting CypA or CypB. (A) Knockdown of CypA and CypB levels was monitored by Western blotting with CypA- and CypB-specific antisera. β-Actin was used as a loading control. (B) Viability of cells at 48 h posttransfection with the various siRNAs normalized to that of cells transfected with the nontargeting control siRNA (100%). (C) GFP reporter expression of cells transfected with the siRNAs indicated below the graph and infected at 48 h posttransfection with EAV-GFP at an MOI of 5. Cells were fixed at 24 h p.i., and GFP fluorescence was quantified and normalized to that in infected cells transfected with nontargeting siRNA. (D) Virus titers at 32 h p.i. in the culture medium of cells transfected with the siRNAs indicated below the graph and infected at 48 h posttransfection with wt EAV at an MOI of 0.01.
Cyclophilin A cosediments with EAV RTCs.
Since our RNAi experiments suggested that EAV RNA synthesis depends on the availability of CypA, we investigated whether CypA cosediments with EAV RTC-containing membranes. We therefore fractionated PNSs from EAV-infected and mock-infected Vero E6 cells in a 0 to 30% OptiPrep density gradient. Gradient fractions were analyzed by Western blotting using antisera against CypA, CypB, several EAV nsps, and various organelle marker proteins (Fig. 7). Densities in the gradient ranged from 1.04 g/ml to 1.18 g/ml (Fig. 7A), and the low-density fractions of both EAV-infected and mock-infected PNSs contained the cytosolic marker GAPDH, while several organelle markers, like the ER protein calnexin and the mitochondrial marker CoxIV (data not shown), were found in higher-density fractions (Fig. 7B and C). This confirmed the separation of membrane-containing fractions from the cytosol. The membrane-associated EAV RTCs, detected with an antiserum directed against nsp9 (RNA-dependent RNA polymerase [RdRp]), sedimented at densities of about 1.15 g/ml (Fig. 7C). The nsp9-containing fraction also contained significant amounts of the normally cytosolic CypA (Fig. 7C). Upon density gradient fractionation of PNSs from uninfected cells, CypA was found only in the low-density cytosolic fractions (Fig. 7B), but in PNSs from EAV-infected cells, a fraction of CypA was found to cosediment with the RTC-containing membranes (compare fractions 3 in Fig. 7B and C). CypB, being an ER-associated protein, was observed in the high-density gradient fractions of both mock- and EAV-infected cell lysates (Fig. 7B). Therefore, the protein was present in the fractions containing the EAV RTCs, in particular, in the nsp9-containing fraction (Fig. 7C, fractions 3 and 4), but was clearly more dispersed in the gradient with mock-infected PNSs.
Fig 7.
Cosedimentation of CypA with EAV RTCs in density gradients. The postnuclear supernatant of EAV- and mock-infected Vero E6 cells was fractionated using a continuous 0 to 30% OptiPrep density gradient. (A) Densities of the fractions were determined with a refractometer. (B) Distribution of CypA, CypB, the cytosolic marker protein GAPDH, and the ER marker protein calnexin in density gradient fractions of mock-infected cell lysates analyzed by Western blotting. (C) Distribution of CypA, CypB, the cytosolic marker protein GAPDH, the ER marker protein calnexin, and EAV RdRp nsp9 in density gradient fractions of EAV-infected cells analyzed by Western blotting. (D) Before loading on a 0 to 30% OptiPrep gradient, PNS was pretreated with 12 μM CsA for 30 min on ice. Western blot analysis was used to determine the sedimentation of CypA, the ER marker protein calnexin, and the cytosolic marker protein GAPDH in nsp9-containing membrane fractions of CsA-treated and untreated PNSs and a fraction with the same density prepared from mock-infected cells.
To analyze whether CsA can prevent the cosedimentation of CypA with EAV RTCs, we pretreated the PNSs from EAV-infected cells with CsA at 12 μM—the concentration that completely inhibited EAV RTC activity in vitro—for 30 min on ice before separating the material in an OptiPrep density gradient. Subsequently, the high-density nsp9-containing membrane fractions were analyzed for the presence of CypA by Western blotting (Fig. 7D). In the absence of CsA, a clear cosedimentation of CypA and nsp9 was observed, while CypA was no longer detectable in the high-density nsp9-containing fraction of CsA-treated lysates. This suggests that CsA can prevent the cosedimentation of CypA with the membrane-associated EAV RTCs.
DISCUSSION
Our study shows that arterivirus replication can be inhibited by the cyclophilin inhibitor CsA and the nonimmunosuppressive CsA analog Debio-064, which inhibit EAV RNA synthesis, likely through their effect on the host protein CypA that appears to be recruited to EAV RTCs. CsA inhibits the PPIase function of CsA-sensitive Cyp family members, like CypA, by binding to their active site (13). We show here that low-micromolar concentrations of CsA can block the replication of both EAV and PRRSV, two prominent representatives of the arterivirus family. PRRSV-GFP replication was inhibited with an IC50 of 5.22 μM, and an almost complete block was observed upon treatment with 16 μM CsA (Fig. 1C). These values are comparable to those previously observed for the inhibition of coronavirus replication by CsA (9, 10). Compared to PRRSV and coronaviruses, the inhibitory effect of CsA was even stronger for EAV, for which we calculated an IC50 of 0.95 μM (Fig. 1A). The IC50s obtained for arteriviruses are in the range of those observed for other viruses, like HCV (7, 42), several flaviviruses (8), vaccinia virus (43), and HIV-1 (5). A remarkable and yet not understood phenomenon is that a small fraction (1 to 5%) of PRRSV-infected MARC-145 cells and, previously, coronavirus-infected Vero E6, 17Cl1, or Huh7 cells (9) appeared to be refractive to CsA treatment, even at high concentrations. This effect was not observed for EAV-infected BHK-21 cells (Fig. 3C), which might be explained by the higher sensitivity of EAV to the compound. In any case, the fact that the distantly related coronaviruses (9, 10) and arteriviruses can both be inhibited by CsA suggests the nidovirus-wide conservation of a cyclophilin-dependent function in viral replication.
Previously, CsA was found to inhibit the replication of a variety of RNA viruses, including important human pathogens like HCV, HIV-1, and dengue virus (reviewed in reference 14). For example, both CypA and CypB were found to specifically interact with the flaviviral nonstructural proteins NS5A and NS5B, and these interactions are sensitive to CsA treatment (6, 44–46). In the case of the interaction of CypA with HCV NS5A, the PPIase activity of the former was proposed to induce a conformational change in the latter (47) that promotes RNA binding to NS5A and enhances RNA replication (48). Chatterji et al. reported that, in addition to the CypA-NS5A interaction, HCV replication also depends on the binding of CypA to the enzymatic pocket of NS5B, the viral RdRp, thus enhancing its affinity for RNA. On the other hand, PPIase-defective CypA failed to interact with HCV NS5B (49), suggesting that the isomerase activity of CypA is an essential factor in the interaction with NS5B that promotes HCV replication. Liu et al. showed that the binding of CypA to NS5B mediates the proper folding of enzymatically active NS5B and facilitates the incorporation of the latter into replication complexes. The interaction between CypA and NS5B can be inhibited by CsA (50). In addition, Kaul et al. showed that the development of resistance against the Cyp inhibitor Debio-025 involved mutations (V2440A and V2440L) in HCV NS5B that are close to the NS5A/NS5B cleavage site. These are thought to delay processing of the NS5A/NS5B junction, thus extending the time during which the CypA binding site in NS5B is accessible (51). As a result, smaller amounts of CypA would suffice to mediate the proper folding of NS5B and its incorporation into replication complexes. Similar functions were attributed to CypB, since the interaction between CypB and NS5B was also found to be essential for RNA binding by NS5B and for HCV replication as a whole (52). Furthermore, Japanese encephalitis virus replication depends on the binding of CypB to NS4A and on CypB isomerase activity (6). For a number of RNA viruses, CypA was found to be incorporated into newly formed virions, although the functional relevance of this finding remains to be addressed in more detail (4, 53, 54). CypA also interacts with the SARS-CoV N protein (55), suggesting that the protein could be incorporated into virions (55), although coronavirus N proteins have also been implicated in viral RNA synthesis (56, 57) and are associated with intracellular replication structures (58).
We show here that expression of CypA is required for efficient EAV replication, as siRNA-mediated knockdown of CypA drastically reduced EAV-GFP replication (Fig. 6), while targeting CypB or Cyp40 (Fig. 6 and data not shown) had no effect. The importance of the normally cytosolic CypA was further substantiated by its cosedimentation with RTC-containing membrane structures in the high-density gradient fractions of EAV-infected cell lysates. In such gradients, the sedimentation of the ER marker protein calnexin was essentially similar when comparing infected and mock-infected PNSs (Fig. 7B and C). Furthermore, CsA treatment was able to prevent the sedimentation of CypA to the part of the gradient that also contained the EAV RTCs, following their biochemical isolation from infected cells (Fig. 7D), and the in vitro RNA-synthesizing activity of such replication structures was found to be inhibited by CsA and Debio-064 (Fig. 5B and C). The distribution of CypB appeared to be less dispersed in gradients containing infected cell lysates than in those containing mock-infected lysates, even though EAV-GFP replication was not affected by the siRNA-mediated knockdown of CypB levels (Fig. 6). This suggests that although the subcellular localization of CypB might be affected by the extensive EAV-driven modification of intracellular membranes (25), this does not have a measurable effect on virus replication. By using fluorescence microscopy, colocalization of CypA and viral RTCs could not be observed, presumably because the fraction of CypA that localizes to replication structures is too small (data not shown). Interestingly, we previously could not measure an effect on SARS-CoV replication when CypA or CypB expression was (largely) silenced (9) in the same 293/ACE2 cells used here for our EAV studies. The ∼20% residual Cyp expression that remained after siRNA-mediated knockdown may have been sufficient to support normal SARS-CoV replication, whereas it appears to have been insufficient to support the efficient replication of the apparently more sensitive EAV, in line with the higher sensitivity of this arterivirus to CsA treatment.
As reported for HCV (48–50), the association of CypA with the EAV replication structures suggests the existence of a functional—presumably PPIase activity-dependent—interaction that is essential for virus replication. This member of the cyclophilin protein family appears to directly promote the RNA-synthesizing activity of the EAV RTC (Fig. 5). Based on studies that analyzed binding sites for CypA using a set of 40 potential CypA-inhibiting peptides (59), we identified several potential CypA binding sites in EAV nsp10, the viral helicase protein. A functionally important interaction with such a key enzyme in arterivirus RNA synthesis could certainly explain that efficient EAV replication depends on the availability of sufficient CypA. Clearly, at this moment, we cannot exclude (direct or indirect) interactions with any of the other viral proteins, including—in analogy to HCV (49)—the viral RdRp subunit (nsp9). In line with the ideas regarding the influence of CypA on the RNA-binding capacity of HCV NS5A and NS5B, EAV RTC-associated CypA may be involved in the proper folding or activation of viral enzymes and/or their binding to viral RNA, which might directly affect their function in RNA synthesis.
CsA analogs like Debio-025, NIM811, and SCY635, which have an increased affinity for Cyps and lack the undesired immunosuppressive effect of CsA (17, 18, 40), can be considered promising antiviral compounds, as they could block HCV replication almost completely and resistance to these compounds does not easily develop compared to the ease of resistance development by inhibitors directly targeting viral enzymes (60). In our study, we compared the inhibition of EAV and PRRSV replication by CsA with that caused by the nonimmunosuppressive CsA analog Debio-064. For Debio-064, we obtained an IC50 of 0.32 μM, 3-fold lower than that of CsA (Fig. 2), which is in line with Debio-064's higher affinity for Cyps. We also observed an inhibitory effect of Debio-064 on PRRSV-GFP replication, although in contrast to EAV, its IC50 was similar to that of CsA. Therefore, not only do more potent (nonimmunosuppressive) CsA analogs constitute a promising class of molecules for the treatment of viral infections, but also, these compounds are valuable research tools for mechanistic studies into the role of cyclophilins in the replication of nidoviruses and other positive-stranded RNA viruses.
ACKNOWLEDGMENTS
We thank Johan Neyts for helpful discussions and Corrine Beugeling for excellent technical assistance.
This research was supported in part by TOP grant 700.57.301 from the Council for Chemical Sciences of The Netherlands Organization for Scientific Research (NWO-CW) and by the European Union Seventh Framework Programme (FP7/2007-2013) under SILVER grant agreement no. 260644.
Footnotes
Published ahead of print 14 November 2012
REFERENCES
- 1. Nagy PD, Pogany J. 2012. The dependence of viral RNA replication on co-opted host factors. Nat. Rev. Microbiol. 10:137–149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Kellam P. 2006. Attacking pathogens through their hosts. Genome Biol. 7:201 doi:10.1186/gb-2006-7-1-201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Schwegmann A, Brombacher F. 2008. Host-directed drug targeting of factors hijacked by pathogens. Sci. Signal. 1:re8 doi:10.1126/scisignal.129re8 [DOI] [PubMed] [Google Scholar]
- 4. Bose S, Mathur M, Bates P, Joshi N, Banerjee AK. 2003. Requirement for cyclophilin A for the replication of vesicular stomatitis virus New Jersey serotype. J. Gen. Virol. 84:1687–1699 [DOI] [PubMed] [Google Scholar]
- 5. Briggs CJ, Ott DE, Coren LV, Oroszlan S, Tozser J. 1999. Comparison of the effect of FK506 and cyclosporin A on virus production in H9 cells chronically and newly infected by HIV-1. Arch. Virol. 144:2151–2160 [DOI] [PubMed] [Google Scholar]
- 6. Kambara H, Tani H, Mori Y, Abe T, Katoh H, Fukuhara T, Taguwa S, Moriishi K, Matsuura Y. 2011. Involvement of cyclophilin B in the replication of Japanese encephalitis virus. Virology 412:211–219 [DOI] [PubMed] [Google Scholar]
- 7. Nakagawa M, Sakamoto N, Enomoto N, Tanabe Y, Kanazawa N, Koyama T, Kurosaki M, Maekawa S, Yamashiro T, Chen CH, Itsui Y, Kakinuma S, Watanabe M. 2004. Specific inhibition of hepatitis C virus replication by cyclosporin A. Biochem. Biophys. Res. Commun. 313:42–47 [DOI] [PubMed] [Google Scholar]
- 8. Qing M, Yang F, Zhang B, Zou G, Robida JM, Yuan Z, Tang H, Shi PY. 2009. Cyclosporine inhibits flavivirus replication through blocking the interaction between host cyclophilins and viral NS5 protein. Antimicrob. Agents Chemother. 53:3226–3235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. de Wilde AH, Zevenhoven-Dobbe JC, van der Meer Y, Thiel V, Narayanan K, Makino S, Snijder EJ, van Hemert MJ. 2011. Cyclosporin A inhibits the replication of diverse coronaviruses. J. Gen. Virol. 92:2542–2548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Pfefferle S, Schopf J, Kogl M, Friedel CC, Muller MA, Carbajo-Lozoya J, Stellberger T, von Dall'armi E, Herzog P, Kallies S, Niemeyer D, Ditt V, Kuri T, Zust R, Pumpor K, Hilgenfeld R, Schwarz F, Zimmer R, Steffen I, Weber F, Thiel V, Herrler G, Thiel HJ, Schwegmann-Wessels C, Pohlmann S, Haas J, Drosten C, von Brunn A. 2011. The SARS-coronavirus-host interactome: identification of cyclophilins as target for pan-coronavirus inhibitors. PLoS Pathog. 7:e1002331 doi:10.1371/journal.ppat.1002331 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Tanaka Y, Sato Y, Osawa S, Inoue M, Tanaka S, Sasaki T. 2012. Suppression of feline coronavirus replication in vitro by cyclosporin A. Vet. Res. 43:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Barik S. 2006. Immunophilins: for the love of proteins. Cell. Mol. Life Sci. 63:2889–2900 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Davis TL, Walker JR, Campagna-Slater V, Finerty PJ, Paramanathan R, Bernstein G, MacKenzie F, Tempel W, Ouyang H, Lee WH, Eisenmesser EZ, Dhe-Paganon S. 2010. Structural and biochemical characterization of the human cyclophilin family of peptidyl-prolyl isomerases. PLoS Biol. 8:e1000439 doi:10.1371/journal.pbio.1000439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Nagy PD, Wang RY, Pogany J, Hafren A, Makinen K. 2011. Emerging picture of host chaperone and cyclophilin roles in RNA virus replication. Virology 411:374–382 [DOI] [PubMed] [Google Scholar]
- 15. Tedesco D, Haragsim L. 2012. Cyclosporine: a review. J. Transplant. 2012:230386 doi:10.1155/2012/230386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Flisiak R, Feinman SV, Jablkowski M, Horban A, Kryczka W, Pawlowska M, Heathcote JE, Mazzella G, Vandelli C, Nicolas-Metral V, Grosgurin P, Liz JS, Scalfaro P, Porchet H, Crabbe R. 2009. The cyclophilin inhibitor Debio 025 combined with PEG IFNalpha2a significantly reduces viral load in treatment-naive hepatitis C patients. Hepatology 49:1460–1468 [DOI] [PubMed] [Google Scholar]
- 17. Flisiak R, Horban A, Gallay P, Bobardt M, Selvarajah S, Wiercinska-Drapalo A, Siwak E, Cielniak I, Higersberger J, Kierkus J, Aeschlimann C, Grosgurin P, Nicolas-Metral V, Dumont JM, Porchet H, Crabbe R, Scalfaro P. 2008. The cyclophilin inhibitor Debio-025 shows potent anti-hepatitis C effect in patients coinfected with hepatitis C and human immunodeficiency virus. Hepatology 47:817–826 [DOI] [PubMed] [Google Scholar]
- 18. Lawitz E, Godofsky E, Rouzier R, Marbury T, Nguyen T, Ke J, Huang M, Praestgaard J, Serra D, Evans TG. 2011. Safety, pharmacokinetics, and antiviral activity of the cyclophilin inhibitor NIM811 alone or in combination with pegylated interferon in HCV-infected patients receiving 14 days of therapy. Antiviral Res. 89:238–245 [DOI] [PubMed] [Google Scholar]
- 19. de Groot RJ, Cowley JA, Enjuanes L, Faaberg KS, Perlman S, Rottier PJ, Snijder EJ, Ziebuhr J, Gorbalenya AE. 2012. Order of Nidovirales, p 785–795 In King A, Adams M, Carstens E, Lefkowitz EJ. (ed), Virus taxonomy, the 9th Report of the International Committee on Taxonomy of Viruses. Academic Press, New York, NY [Google Scholar]
- 20. Holtkamp DJ, Kliebenstein JB, Neumann EJ, Zimmerman JJ, Rotto H, Yoder TK, Wang C, Yeske P, Mowrer C, Haley C. 2011. Abstr. 2011 Int. PRRS Symp., Chicago, IL, abstr. 86 [Google Scholar]
- 21. Snijder EJ, Spaan WJM. 2007. Arteriviruses, p 1337–1355 In Knipe DM, et al. (ed), Fields virology, 5th ed Lippincott Williams & Wilkins, Philadelphia, PA [Google Scholar]
- 22. Gorbalenya AE, Enjuanes L, Ziebuhr J, Snijder EJ. 2006. Nidovirales: evolving the largest RNA virus genome. Virus Res. 117:17–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Pasternak AO, Spaan WJ, Snijder EJ. 2006. Nidovirus transcription: how to make sense? J. Gen. Virol. 87:1403–1421 [DOI] [PubMed] [Google Scholar]
- 24. Fang Y, Snijder EJ. 2010. The PRRSV replicase: exploring the multifunctionality of an intriguing set of nonstructural proteins. Virus Res. 154:61–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Knoops K, Barcena M, Limpens RW, Koster AJ, Mommaas AM, Snijder EJ. 2012. Ultrastructural characterization of arterivirus replication structures: reshaping the endoplasmic reticulum to accommodate viral RNA synthesis. J. Virol. 86:2474–2487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. van der Meer Y, van Tol H, Krijnse Locker J, Snijder EJ. 1998. ORF1a-encoded replicase subunits are involved in the membrane association of the arterivirus replication complex. J. Virol. 72:6689–6698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. van Hemert MJ, de Wilde AH, Gorbalenya AE, Snijder EJ. 2008. The in vitro RNA synthesizing activity of the isolated arterivirus replication/transcription complex is dependent on a host factor. J. Biol. Chem. 283:16525–16536 [DOI] [PubMed] [Google Scholar]
- 28. Nedialkova DD, Gorbalenya AE, Snijder EJ. 2010. Arterivirus Nsp1 modulates the accumulation of minus-strand templates to control the relative abundance of viral mRNAs. PLoS Pathog. 6:e1000772 doi:10.1371/journal.ppat.1000772 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. de Vries AA, Chirnside ED, Horzinek MC, Rottier PJ. 1992. Structural proteins of equine arteritis virus. J. Virol. 66:6294–6303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Kim HS, Kwang J, Yoon IJ, Joo HS, Frey ML. 1993. Enhanced replication of porcine reproductive and respiratory syndrome (PRRS) virus in a homogeneous subpopulation of MA-104 cell line. Arch. Virol. 133:477–483 [DOI] [PubMed] [Google Scholar]
- 31. Kamitani W, Narayanan K, Huang C, Lokugamage K, Ikegami T, Ito N, Kubo H, Makino S. 2006. Severe acute respiratory syndrome coronavirus nsp1 protein suppresses host gene expression by promoting host mRNA degradation. Proc. Natl. Acad. Sci. U. S. A. 103:12885–12890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Bryans JT, Crowe ME, Doll ER, McCollum WH. 1957. Isolation of a filterable agent causing arteritis of horses and abortion by mares; its differentiation from the equine abortion (influenza) virus. Cornell Vet. 47:3–41 [PubMed] [Google Scholar]
- 33. van den Born E, Posthuma CC, Knoops K, Snijder EJ. 2007. An infectious recombinant equine arteritis virus expressing green fluorescent protein from its replicase gene. J. Gen. Virol. 88:1196–1205 [DOI] [PubMed] [Google Scholar]
- 34. Fang Y, Rowland RR, Roof M, Lunney JK, Christopher-Hennings J, Nelson EA. 2006. A full-length cDNA infectious clone of North American type 1 porcine reproductive and respiratory syndrome virus: expression of green fluorescent protein in the Nsp2 region. J. Virol. 80:11447–11455 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Sun Z, Li Y, Ransburgh R, Snijder EJ, Fang Y. 2012. Nonstructural protein 2 of porcine reproductive and respiratory syndrome virus inhibits the antiviral function of interferon-stimulated gene 15. J. Virol. 86:3839–3850 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Pedersen KW, van der Meer Y, Roos N, Snijder EJ. 1999. Open reading frame 1a-encoded subunits of the arterivirus replicase induce endoplasmic reticulum-derived double-membrane vesicles which carry the viral replication complex. J. Virol. 73:2016–2026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. MacLachlan NJ, Balasuriya UB, Hedges JF, Schweidler TM, McCollum WH, Timoney PJ, Hullinger PJ, Patton JF. 1998. Serologic response of horses to the structural proteins of equine arteritis virus. J. Vet. Diagn. Invest. 10:229–236 [DOI] [PubMed] [Google Scholar]
- 38. Knoops K, Swett-Tapia C, van den Worm SH, Te Velthuis AJ, Koster AJ, Mommaas AM, Snijder EJ, Kikkert M. 2010. Integrity of the early secretory pathway promotes, but is not required for, severe acute respiratory syndrome coronavirus RNA synthesis and virus-induced remodeling of endoplasmic reticulum membranes. J. Virol. 84:833–846 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Schreiber SL, Crabtree GR. 1992. The mechanism of action of cyclosporin A and FK506. Immunol. Today 13:136–142 [DOI] [PubMed] [Google Scholar]
- 40. Hopkins S, Dimassimo B, Rusnak P, Heuman D, Lalezari J, Sluder A, Scorneaux B, Mosier S, Kowalczyk P, Ribeill Y, Baugh J, Gallay P. 2012. The cyclophilin inhibitor SCY-635 suppresses viral replication and induces endogenous interferons in patients with chronic HCV genotype 1 infection. J. Hepatol. 57:47–54 [DOI] [PubMed] [Google Scholar]
- 41. Wenger R, Mutter M, Garrouste P, Lysek R, Turpin O, Vuagniaux G, Nicolas V, Novaroli Zanolari L, Crabbé R. November 2009. Cycloundecadepsipeptide compounds and use of said compounds as a medicament. Patent WO 2010/052559
- 42. Watashi K, Hijikata M, Hosaka M, Yamaji M, Shimotohno K. 2003. Cyclosporin A suppresses replication of hepatitis C virus genome in cultured hepatocytes. Hepatology 38:1282–1288 [DOI] [PubMed] [Google Scholar]
- 43. Damaso CR, Keller SJ. 1994. Cyclosporin A inhibits vaccinia virus replication in vitro. Arch. Virol. 134:303–319 [DOI] [PubMed] [Google Scholar]
- 44. Chatterji U, Lim P, Bobardt MD, Wieland S, Cordek DG, Vuagniaux G, Chisari F, Cameron CE, Targett-Adams P, Parkinson T, Gallay PA. 2010. HCV resistance to cyclosporin A does not correlate with a resistance of the NS5A-cyclophilin A interaction to cyclophilin inhibitors. J. Hepatol. 53:50–56 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Fernandes F, Ansari IU, Striker R. 2010. Cyclosporine inhibits a direct interaction between cyclophilins and hepatitis C NS5A. PLoS One 5:e9815 doi:10.1371/journal.pone.0009815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Gaither LA, Borawski J, Anderson LJ, Balabanis KA, Devay P, Joberty G, Rau C, Schirle M, Bouwmeester T, Mickanin C, Zhao S, Vickers C, Lee L, Deng G, Baryza J, Fujimoto RA, Lin K, Compton T, Wiedmann B. 2010. Multiple cyclophilins involved in different cellular pathways mediate HCV replication. Virology 397:43–55 [DOI] [PubMed] [Google Scholar]
- 47. Coelmont L, Hanoulle X, Chatterji U, Berger C, Snoeck J, Bobardt M, Lim P, Vliegen I, Paeshuyse J, Vuagniaux G, Vandamme AM, Bartenschlager R, Gallay P, Lippens G, Neyts J. 2010. DEB025 (alisporivir) inhibits hepatitis C virus replication by preventing a cyclophilin A induced cis-trans isomerisation in domain II of NS5A. PLoS One 5:e13687 doi:10.1371/journal.pone.0013687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Foster TL, Gallay P, Stonehouse NJ, Harris M. 2011. Cyclophilin A interacts with domain II of hepatitis C virus NS5A and stimulates RNA binding in an isomerase-dependent manner. J. Virol. 85:7460–7464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Chatterji U, Bobardt M, Selvarajah S, Yang F, Tang H, Sakamoto N, Vuagniaux G, Parkinson T, Gallay P. 2009. The isomerase active site of cyclophilin A is critical for hepatitis C virus replication. J. Biol. Chem. 284:16998–17005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Liu Z, Yang F, Robotham JM, Tang H. 2009. Critical role of cyclophilin A and its prolyl-peptidyl isomerase activity in the structure and function of the hepatitis C virus replication complex. J. Virol. 83:6554–6565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Kaul A, Stauffer S, Berger C, Pertel T, Schmitt J, Kallis S, Zayas M, Lohmann V, Luban J, Bartenschlager R. 2009. Essential role of cyclophilin A for hepatitis C virus replication and virus production and possible link to polyprotein cleavage kinetics. PLoS Pathog. 5:e1000546 doi:10.1371/journal.ppat.1000546 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Watashi K, Ishii N, Hijikata M, Inoue D, Murata T, Miyanari Y, Shimotohno K. 2005. Cyclophilin B is a functional regulator of hepatitis C virus RNA polymerase. Mol. Cell 19:111–122 [DOI] [PubMed] [Google Scholar]
- 53. Castro AP, Carvalho TM, Moussatche N, Damaso CR. 2003. Redistribution of cyclophilin A to viral factories during vaccinia virus infection and its incorporation into mature particles. J. Virol. 77:9052–9068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Liu X, Sun L, Yu M, Wang Z, Xu C, Xue Q, Zhang K, Ye X, Kitamura Y, Liu W. 2009. Cyclophilin A interacts with influenza A virus M1 protein and impairs the early stage of the viral replication. Cell. Microbiol. 11:730–741 [DOI] [PubMed] [Google Scholar]
- 55. Luo C, Luo H, Zheng S, Gui C, Yue L, Yu C, Sun T, He P, Chen J, Shen J, Luo X, Li Y, Liu H, Bai D, Yang Y, Li F, Zuo J, Hilgenfeld R, Pei G, Chen K, Shen X, Jiang H. 2004. Nucleocapsid protein of SARS coronavirus tightly binds to human cyclophilin A. Biochem. Biophys. Res. Commun. 321:557–565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Almazan F, Galan C, Enjuanes L. 2004. The nucleoprotein is required for efficient coronavirus genome replication. J. Virol. 78:12683–12688 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Schelle B, Karl N, Ludewig B, Siddell SG, Thiel V. 2006. Nucleocapsid protein expression facilitates coronavirus replication. Adv. Exp. Med. Biol. 581:43–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. van der Meer Y, Snijder EJ, Dobbe JC, Schleich S, Denison MR, Spaan WJ, Krijnse Locker J. 1999. Localization of mouse hepatitis virus nonstructural proteins and RNA synthesis indicates a role for late endosomes in viral replication. J. Virol. 73:7641–7657 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Pang X, Zhang M, Zhou L, Xie F, Lu H, He W, Jiang S, Yu L, Zhang X. 2011. Discovery of a potent peptidic cyclophilin A inhibitor Trp-Gly-Pro. Eur. J. Med. Chem. 46:1701–1705 [DOI] [PubMed] [Google Scholar]
- 60. Puyang X, Poulin DL, Mathy JE, Anderson LJ, Ma S, Fang Z, Zhu S, Lin K, Fujimoto R, Compton T, Wiedmann B. 2010. Mechanism of resistance of hepatitis C virus replicons to structurally distinct cyclophilin inhibitors. Antimicrob. Agents Chemother. 54:1981–1987 [DOI] [PMC free article] [PubMed] [Google Scholar]