INTRODUCTION
Hepatitis C virus (HCV) infects at least 130 million people worldwide and is the major cause of chronic liver disease. Infected patients are at risk of developing fibrosis, cirrhosis, and liver cancer (
1–3). Although HCV was identified in 1989, advances in treatment have been augmented since the development of cell culture-grown HCV (HCVcc) in 2005 (
4–6). No vaccine is available, and the current treatment for HCV infection involves a weekly injection of pegylated alpha interferon and a twice-daily weight-based dose of ribavirin for 24 to 48 weeks. This standard of care is plagued by a long duration, limited efficacy, and serious side effects (
7). Although the recent addition of new direct-acting antivirals (DAAs) targeting HCV NS3-4A protease—telaprevir and boceprevir—to the anti-HCV therapeutic arsenal have improved the cure rates, they must be used in combination with interferon, as HCV has a remarkable ability to overcome a single DAA. Telaprevir and boceprevir only work in patients infected with genotype 1 HCV and are both not very effective in patients who did not respond to pegylated interferon-ribavirin therapy (
8). In addition, both telaprevir and boceprevir appear to worsen the already problematic side effects of the standard therapy, such as rashes and anemia (
9). Currently approved DAAs and most molecules in the pipeline are protease inhibitors, nucleoside inhibitors, nonnucleoside inhibitors, and NS5A inhibitors (
10). A major obstacle in combating HCV is the low fidelity of the viral replication machinery, enabling the virus to quickly develop resistance (
11). To date, ITX-5061 is the only inhibitor of HCV entry that has entered clinical testing. ITX-5061 blocks a postbinding step in the viral entry process by directly interacting with the entry factor scavenger receptor B1 (SR-B1) (
12). New DAAs targeting entry steps critical to viral infection with additive potency when combined with existing DAAs and exhibiting low cytotoxicity are highly desirable.
HCV is an enveloped, positive-sense RNA virus belonging to the
Flaviviridae family. The 9.6-kb viral genome encodes a single large polyprotein that is processed by viral and cellular proteases to produce the virion structural proteins (core and glycoproteins E1 and E2), P7, and nonstructural proteins (NS2, NS3, NS4A, NS4B, NS5A, and NS5B). HCV infection involves multiple steps. Viruses first attach to target cells via glycosaminoglycans and low-density lipoprotein (LDL) receptors. After recruitment to the membrane, HCV binds sequentially to entry factors involving SR-B1, the tetraspanin CD81, the Niemann-Pick C1-like 1 (NPC1L1) cholesterol (Cho) uptake receptor, and proteins of tight junctions, i.e., CLDN1 and OCLD (
13). HCV then enters cells at the tight junction via clathrin-mediated endocytosis and fuses with the host membrane in the late endosome. Progress in defining the molecular mechanism of HCV entry raises the opportunity to exploit new viral and host targets for therapeutic intervention. Entry inhibitors have the potential to limit the expansion of the infected cell reservoir, prevent reinfection after liver transplantation, and complement the many protease and polymerase inhibitors currently under development. Although the discovery of drugs targeting the entry stage is still in its infancy, antibodies against SR-B1 (
14), CD81 (
15), and CLDN1 (
16), as well as a number of small-molecule inhibitors, have recently been developed and are able to effectively block HCV entry (
17–24).
Phenothiazines are a group of nitrogen- and sulfur-containing tricyclic compounds that were first synthesized by Bernthsen in 1883. Phenothiazines with dialkylaminoalkyl groups and small groups substituted at positions 10 and 2, respectively, were found to interact with the dopamine receptors and have exhibited valuable activities, such as neuroleptic, antiemetic, antihistaminic, antipruritic, analgesic, and anthelmintic activities (
25). To date, more than 100 phenothiazines have been used in clinics to treat psychotic disorders, and over 5,000 phenothiazine derivatives have been synthesized. Other receptors that can be modulated by phenothiazines include histamine H1, adrenergic α1 and α2, muscarinic (cholinergic), and serotonergic receptors (
25). In addition to neurotransmitter receptors, phenothiazines have also been reported to bind to calmodulin and block its calcium signal-transduction activity, inhibit clathrin-coated pit formation, and activate rynodine receptors (
26). Antiviral and antimicrobial activities have also been described for phenothiazines and related compounds (
27).
Our lab and others recently identified three phenothiazines—fluphenazine, trifluoperazine, and prochlorperazine—as potent HCV entry inhibitors (
28,
29). In this work, we wanted to understand the antiviral mode of action of this family of compounds, which presumably inhibit HCV entry through a common mechanism of action. This information will assist in future endeavors to identify new and more potent inhibitors of HCV entry. We found that phenothiazines inhibit the virus-cell fusion step of the HCV life cycle by intercalating into the host cholesterol-rich membrane. In the presence of phenothiazines, cholesterol-rich membranes become more permeable to water molecules, leading to increased membrane fluidity. We subsequently developed a high-throughput screening assay. We screened a library of 2,752 compounds and identified a molecule, topotecan, that dose-dependently inhibits HCVcc-liposome fusion. This study suggests that alteration of target cholesterol-rich membrane fluidity may be a novel mode for suppressing HCV entry and should facilitate the identification of new HCV inhibitors with unique modes of action.
MATERIALS AND METHODS
Cells, plasmids, compounds, and reagents.
Huh-7.5 cells and plasmids encoding HIV Gag-Pol (
30) and the envelope proteins of HCV H77/J6 (
30) and vesicular stomatitis virus (VSV) were kindly provided by Charles Rice (Rockefeller University, NY). HEK 293T cells were purchased from Invitrogen (Carlsbad, CA). Trifluoperazine, prochlorperazine, mesoridazine, promazine, triflupromazine, and
cis-flupentixol were purchased from Sigma-Aldrich (St. Louis, MO). Chlorpromazine and thioridazine were from MP Biomedicals (Solon, OH). Fluphenazine and bafilomycin were from Alfa Aesar (Ward Hill, MA) and Axxora (San Diego, CA), respectively. All phenothiazine compounds were dissolved in dimethyl sulfoxide (DMSO) to a 10 mM stock concentration. Bafilomycin was dissolved in DMSO to a 250 μM stock concentration. Laurdan and Prodan fluorescent probes were purchased from Anaspec (Fremont, CA). Laurdan and Prodan were dissolved in methanol and DMSO to final concentrations of 0.5 mM and 10 mM, respectively. The human anti-CD81 JS81 monoclonal antibody (MAb) was obtained from BD Biosciences (San Jose, CA). CellTiter-Glo luminescent cell viability assay kits and BioLux Gaussia luciferase assay kits were purchased from Promega (Madison, WI) and New England BioLabs (Ipswich, MA), respectively. The growth medium for all cell culture work was Dulbecco's modified Eagle's medium (DMEM) containing 4,500 mg/liter glucose, 4.0 mM
l-glutamine, and 110 mg/liter sodium pyruvate (Thermo Scientific HyClone, Logan, UT) supplemented with 10% fetal bovine serum (Atlanta Biologicals, Lawrenceville, GA) and 1× nonessential amino acids (Thermo Scientific HyClone, Logan, UT). Dulbecco's phosphate-buffered saline (DPBS) was purchased from Thermo Scientific HyClone (Logan, UT). Octadecyl rhodamine B chloride (R18) was purchased from Invitrogen (St. Aubin, France), and all other lipids (99% pure) were from Avanti Polar Lipids (Alabaster, AL).
Production of HCVcc and pseudotyped lentiviruses.
The production and titer determination of Jc1 Gluc HCVcc (
31) in Huh-7.5 cells were performed as previously described (
28). Jc1 Gluc HCV contains the
Gaussia luciferase (Gluc) reporter gene between the HCV genes encoding the p7 and NS2 proteins. Pseudotyped lentiviruses were produced by cotransfecting 293T cells with plasmids encoding HIV Gag-Pol (
30), a provirus (pTRIP-Gluc) (
28), and the appropriate envelope protein, using TransIT reagent (Mirus, Madison, WI) following the manufacturer's protocol. The supernatants containing the pseudoparticles were collected, pooled, and filtered (0.45 μm pore size) at 48 h posttransfection and then stored at 4°C for up to 1 week or at −80°C for long-term storage. For production of lentiviruses pseudotyped with the envelope proteins from HCV genotype 1a H77 (H77 HCVpp), HCV genotype 2b J6 (J6 HCVpp), and vesicular stomatitis virus (VSV-Gpp), plasmids H77 E1E2 pcDNA3, J6 E1E2 pcDNA3, and pVSVG, respectively, were used (
30). A control pseudotyped lentivirus lacking any envelope protein (Env
−pp) was generated using the same protocol, except that the envelope protein-encoding plasmid was replaced with empty vector (pcDNA3).
HCVcc infection assays.
To determine whether phenothiazines are virucidal, Jc1 Gluc HCVcc (6.4 × 105 50% tissue culture infective doses [TCID50]/ml) was incubated with phenothiazines (50 or 5 μM), PD 404,182 (150 μM), or DMSO (0.5%) for 1 h at 37°C, and the virus-compound mixtures were diluted 100-fold in growth medium and used to infect Huh-7.5 cells seeded 4 to 6 h earlier in 96-well plates at 3.2 × 104 cells/well. For controls, virus and drugs were diluted 100-fold separately and mixed before infecting Huh-7.5 cells. Cells were thoroughly washed at 14 to 16 h post-virus inoculation to remove residual drug and virus. Supernatant Gluc activity was measured at 48 h postinfection. The percentages of virus entry and spread were determined relative to those of the DMSO control.
To determine whether phenothiazines act on host cells, Huh-7.5 cells were infected with Jc1 Gluc HCVcc at various times after drug removal. Briefly, Huh-7.5 cells were seeded in 48-well plates at 4 × 104 cells/well. After attachment, these cells were treated with phenothiazines (5 μM), PD 404,182 (150 μM), bafilomycin (10 nM), or DMSO (0.5%) for 2 h at 37°C. For sets 1 to 3, these cells were washed thoroughly to remove residual drugs and then inoculated with Jc1 Gluc HCVcc (multiplicity of infection [MOI] = 1) or VSV-Gpp (100-fold dilution) at 0, 4, or 24 h post-drug removal at 37°C. At 15 minutes post-virus inoculation, these cells were thoroughly washed to remove any remaining viruses and returned to the 37°C and 5% CO2 incubator. For set 4, cells were infected with the same viruses, but in the presence of the drug, and were continuously incubated in drug-containing medium after the infection period. Supernatant Gluc activity was measured at 72 h postinfection, normalized to viable cell levels, and used as an indication of viral infection.
To determine the anti-HCV activities of phenothiazines (
Table 1) and topotecan (see
Fig. 6C), Huh-7.5 cells (1.6 × 10
4 cells/well) seeded 4 to 6 h earlier were infected with Jc1 Gluc HCVcc (MOI = 0.01) in the presence of increasing concentrations of the compounds. Supernatant Gluc activity was measured at 48 to 72 h postinfection and normalized to the DMSO (0.02 to 0.5%) treatment control. The cell viability was measured using the CellTiter-Glo assay (Promega, Madison, WI) to gauge the compound toxicity. The 50% inhibitor concentration (IC
50), 50% effective concentration (EC
50), and 50% cytotoxic concentration (CC
50) were calculated using the sigmoidal fit function in OriginLab (OriginLab, Northampton, MA).
Synchronized HCVcc infection assay.
To determine the step of entry inhibited by phenothiazines, we carried out a synchronized infection assay (
Fig. 1B). Huh-7.5 cells were seeded in 48-well plates at 4 × 10
4 cells/well. The next day, virus-cell attachment was initiated by incubating the cells with Jc1 Gluc HCVcc (MOI = 1) at 4°C for 1.5 h. Unbound viruses were removed by thorough washing with complete growth medium, and then infection of bound viruses was initiated by moving the cell culture plates to a 37°C and 5% CO
2 incubator. Fluphenazine (5 μM), bafilomycin (10 nM), JS81 (2 μg/ml), or DMSO (0.05%) was added to the cells at different time points after the temperature shift. Cells were washed thoroughly with complete growth medium at 5 h post-drug addition. Supernatant Gluc activity was measured at 48 h postinfection, using a BioLux Gaussia luciferase assay kit (New England BioLabs, Ipswich, MA), and was used as an indication of viral infection.
HCVpp/cc-liposome fusion assay.
To evaluate the ability of compounds to inhibit HCV fusion
in vitro, we carried out an HCV-liposome fusion assay. Fusion between HCVpp and liposomes was assayed as described elsewhere (
32). H77 HCVpp collected from the cell supernatant was purified and concentrated 100-fold by use of ultracentrifugation devices, to a titer of ∼10
7 IU/ml. One microliter of liposomes (25 μM final lipid concentration) composed of phosphatidylcholine (PC), cholesterol, and R18 (65:30:5 mol%) was added to a 37°C thermostated cuvette containing 20 μl concentrated H77 HCVpp (∼2 × 10
5 viral particles) in phosphate-buffered saline (PBS) at pH 7.2. Fluphenazine, trifluoperazine, or promazine dissolved in DMSO was added to the mixture at a final concentration of 2.5, 5, 10, or 20 μM. After thermic equilibration, fusion was initiated by adding 2 μl diluted HCl (final pH, 5.0). Lipid mixing was measured as the dequenching of R18 (excitation, 560 nm; emission, 590 nm), resulting in an increase of the fluorescence signal. Recordings were performed with a Tecan Infinite-1000 spectrofluorometer. Maximal R18 dequenching was measured after addition of Triton X-100 to the cuvette at a 0.1% final concentration. Fusion between JFH-1 HCVcc and liposomes was determined similarly. Freshly prepared JFH1 HCVcc, concentrated 100-fold through a 20% sucrose cushion, was used in the fusion experiment (
33).
For the modified HCVpp-liposome fusion experiment, 20 μl concentrated H77 HCVpp or liposomes (working suspension at a 1:20 dilution from stock) was preincubated with fluphenazine, trifluoperazine, or promazine at a 5 μM final concentration for 3 min at 37°C. This mixture was then diluted 10-fold with PBS to reach a 0.5 μM final drug concentration, and 1 μl untreated liposomes or 20 μl HCVpp was added, respectively. Fusion was determined in the same way as that described above.
The study of the effect of topotecan on HCVcc, membrane fusion was performed as described previously (
33). Briefly, JFH-1 HCVcc (∼1.6 × 10
5 particles), suspended in 150 mM NaCl, 10 mM Tricine-NaOH, pH 7.4, was added to a cuvette containing R18-labeled PC-cholesterol liposomes (15 μM final lipid concentration) in the absence or presence of increasing concentrations of topotecan. After temperature equilibration, fusion was initiated by HCl addition to the cuvette, and kinetics were recorded using a dual-channel PicoFluor handheld fluorometer (Turner Biosystems, Sunnyvale, CA) operated under the “rhodamine” channel (excitation and emission wavelengths of 540 ± 20 and >570 nm, respectively). Maximal R18 dequenching was measured after the addition of 0.1% Triton X-100 (final concentration) to the cuvette.
Fluorescence spectroscopy.
To determine whether phenothiazines affect membrane fluidity, we calculated the generalized polarization (GP) of the fluorescent dyes Laurdan and Prodan incorporated into the liposomes in the absence or presence of phenothiazines. POPC (11-palmitoyl-2-oleoyl-
sn-glycero-3-phosphocholine), sphingomyelin (SM), and cholesterol (Cho) (Avanti Polar Lipids, Inc., Alabaster, AL) dissolved in chloroform at 10 mg/ml were mixed at a molar ratio of 1:0:0 (100% POPC), 2.3:0:1 (70% POPC plus 30% Cho), or 3.9:1:2.3 (54% POPC plus 14% SM plus 32% Cho). To remove solvent, the lipids were first dried under a stream of nitrogen and then lyophilized. The lyophilized lipid mixtures were resuspended in DPBS at a 400 μM final lipid concentration, sonicated at room temperature in a water bath sonicator for 10 min, and extruded repeatedly through a 100-nm polycarbonate membrane filter (Avanti Polar Lipids, Inc.) to obtain uniformly sized liposomes. Extruded liposomes were stored at 4°C for up to a week. For GP determination, liposomes (200 μM final concentration) were first incubated with Laurdan (5 μM final concentration) or Prodan (15 μM final concentration) for 15 min at room temperature in the dark. Increasing concentrations of the compounds were added to the mixture, and 100 μl/well of liposome-drug mixture was transferred to a white 384-well plate. The plate was incubated at 23°C for 30 min in the dark, and fluorescence spectra were collected in a Gemini EM spectrofluorometer (Molecular Devices, San Francisco, CA) with an excitation wavelength of 310 to 350 nm for both dyes and the emission spectra recorded at 440 and 480 nm for Prodan and 440 and 490 nm for Laurdan. GP was calculated according to the following equation (
34): GP = (
IB −
IR)/(
IB +
IR), where
IB and
IR are the fluorescence intensities at the blue and red edges of the emission spectrum, respectively. Data were corrected for the background signal measured with liposomes deprived of a probe. After the fluorescence spectra were measured, the plate was returned to a 37°C incubator for another 30 min, after which the spectra were measured again.
Screen for inhibitors.
A high-throughput screening assay was developed based on the ability of a compound to selectively increase the GP of cholesterol-rich liposomes in comparison to the DMSO control. The library screening was conducted at The National Screening Laboratory for the Regional Centers of Excellence in Biodefense and Emerging Infectious Disease (NSRB). Liposomes composed of POPC alone or POPC, Cho, and SM at a 3.9:1:2.3 molar ratio (200 μM final concentration) were incubated with Prodan (15 μM final concentration) for 15 min at room temperature in the dark. Eighty microliters of the mixture was dispensed into each well of white 384-well plates by use of a Matrix WellMate liquid dispenser (Matrix, Hudson, NH). One hundred nanoliters of the drug library (with concentrations ranging from 2 to 5 mg/ml) was then added to each well by use of a Seiko D-TRAN XM3106-31 PN 4-axis Cartesian robot pin transferor (Caliper Life Science, Waltham, MA). The plates were incubated in the dark for 30 min at room temperature, and the fluorescence spectra were collected in an SGM 610 FlexStation III spectrofluorometer (Molecular Devices, San Francisco, CA) with an excitation wavelength of 310 nm and the emission spectra recorded at 440 and 480 nm. We screened the Biomol4 (Enzo Life Sciences, Plymouth Meeting, PA) and Chembridge 3 libraries for molecules whose GP differed from that of the 0.25% DMSO control by a value of >0.05 (positive hit). Although we screened only 2,752 compounds in this study, this assay is amenable to high-throughput screening.
Replication inhibition and qRT-PCR.
To determine the effect of topotecan on viral replication, we quantified the amounts of HCV RNA in the appropriate cells (see
Fig. 7A). Huh-7.5 cells were electroporated with Jc1 Gluc HCV genomic RNA according to a previously described protocol (
28) and seeded into 24-well plates (1.4 × 10
5 cells/well). The desired compounds were added to the medium at 6 h postelectroporation. The medium was replaced with fresh, compound-containing medium at 48 h postelectroporation, and the supernatant Gluc activity was determined using a BioLux Gaussia luciferase assay kit (New England BioLabs, Ipswich, MA) at 72 h postelectroporation. After removing all the supernatant, these cells were washed once with Dulbecco's phosphate-buffered saline (Thermo Scientific HyClone, Logan, UT) and underwent one freeze-thaw cycle at −80°C before RNA extraction using an EZNA Total RNA kit (Omega Bio-Tek, Norcross, GA). The amount of HCV RNA was quantified via TaqMan quantitative reverse transcription-PCR (qRT-PCR) (qScript One-Step Fast kit; Quanta Biosciences, Gaithersburg, MD), using previously described primers (
35).
Virus entry inhibition assay.
To determine the effect of topotecan on HCV entry, uptake of HCVpp in the presence of the compound was evaluated. Huh-7.5 cells were seeded in poly-l-lysine-treated 96-well flat-bottom tissue culture plates at 1.8 × 104 cells/well and incubated at 37°C and 5% CO2 for 5 to 6 h to allow cell attachment. Compounds were added to the appropriate wells 1 h before transduction. HCVpp and Env−pp were added to the cells at a 1:5 dilution. The next day, these cells were washed 4 times with 100 μl/well of complete growth medium to remove unbound pseudoparticles and preexisting Gluc reporter, and 100 μl of fresh medium was added to each well. Supernatant Gluc activity was quantified at 48 h postwashing by use of a BioLux Gaussia luciferase assay kit (New England BioLabs, Ipswich, MA). The presented HCVpp entry data represent differences in supernatant Gluc activities between HCVpp- and Env−pp-transduced cells at the specified compound concentrations.
Statistical analysis.
Statistical significance between different samples was evaluated using Student's t test. A P value of <0.01 was considered statistically significant. All analysis was done using Microsoft Excel.
DISCUSSION
Insight into HCV entry gained over the last few years has allowed for the discovery and development of inhibitors acting at different stages of viral uptake. Addition of new entry inhibitors to current therapies will increase the resistance barrier, inhibit expansion of the infected pool, and reduce the rate and extent of reinfection after liver transplantation (
44). We and others recently identified 3 phenothiazine compounds—trifluoperazine, fluphenazine, and prochlorperazine—as inhibitors of HCV entry (
28,
29). Phenothiazines are a large class of chemicals, many of which are currently used in clinics to treat psychotic disorders (
25). To determine whether other phenothiazines can also inhibit HCV infection, we tested 6 additional FDA-approved phenothiazines and similar molecules and discovered that all these molecules exhibit anti-HCV activity (
Table 1). The most potent inhibitor,
cis-flupentixol, exhibited an IC
50 of 0.25 μM and a therapeutic index of 20. The presence of a piperazine ring at position 10 enhances but is not required for HCV entry inhibition. The presence of a trifluoromethyl group at position 2 also appears to enhance anti-HCV activity. This information should assist future structure-activity relationship studies to identify more potent phenothiazine-based anti-HCV inhibitors.
We showed that phenothiazines inhibit HCV-cell fusion by specifically interacting with the host/target membrane. Incorporation of phenothiazines into the leaflets of the target membrane increases the water permeability/fluidity of cholesterol-rich membranes (
Fig. 5) and reduces the rate of virus-liposome lipid mixing and hemifusion (
Fig. 4). The effects of lipid composition on viral infection, particularly the influence of cholesterol and sphingolipids, have been studied widely. Many viruses enter host cells via cholesterol-rich microdomains (lipid rafts), such as West Nile, Ebola, Marburg, herpes simplex, and vaccinia viruses, retroviruses, and alphaviruses (
45). In some cases, the cholesterol dependence is due to clustering of viral receptors in the lipid raft, while in other cases it is due to specific interactions between the viral envelope glycoproteins and (a lipid of) the target membrane, as is the case for the fusion protein of alphaviruses and cholesterol (
46). Concerning HCV, both phenomena are believed to occur.
In vitro cell culture studies have shown that HCV entry is adversely affected by cholesterol depletion (
47,
48). The cholesterol absorption receptor NPC1L1 was recently identified as an HCV entry factor (
49) which forms cholesterol-enriched microdomains together with flotillins (
50). Concerning the tetraspanin HCV receptor CD81, two cholesterol binding sites have been mapped in the three-dimensional model of this molecule (
51). It therefore appears that cholesterol might play a role in HCV entry through the local mobilization of receptors at specific membrane regions.
Our
in vitro fusion studies showed that the presence of cholesterol significantly enhances the fusion of both HCV envelope protein-pseudotyped lentiviruses and cell culture-produced virions with liposomes, further confirming the important role of cholesterol in HCV-mediated fusion (
32,
33,
52). In these receptor-free assays, cholesterol is likely to play a direct role in the fusion process.
Cholesterol is one of the most important lipid species in eukaryotic cells and has several different functions. Two of the primary and essential roles of cholesterol are to decrease permeability and increase the stability of the membrane bilayer (
53). Membranes rich in cholesterol have a rough surface due to clustering of cholesterol molecules into small patches (microdomains) (
54) and the different membrane thicknesses of cholesterol-rich regions (
55,
56). The local inhomogeneity and curvature in the target membrane can influence the early interaction of a fusion protein/peptide with the target membrane and, ultimately, virus fusion (
57,
58). The ability of phenothiazines to significantly increase the fluidity of cholesterol-rich membranes indicates that these molecules may interfere with cholesterol clustering and decrease the packing of cholesterol-rich microdomains, leading to reduced local inhomogeneity, which is important for HCV fusion.
A second possibility, which is not completely independent of the first one, is that the incorporation of phenothiazines can affect interaction of the 3β-OH of cholesterol with the HCV envelope protein/fusion peptide. The 3β-OH molecule participates in H-bond interactions with the head groups of various lipids, water in the solvent, and membrane proteins. In addition, 3β-OH can influence the folding of peptides at the water-membrane interface (
59). The 3β-OH in cholesterol is required for the fusion of Semliki Forest virus (
60,
61) and the optimal fusion of HCV with liposomes (
32). Insertion of phenothiazines into the cholesterol-rich membrane may adversely affect the interaction of 3β-OH with HCV envelope proteins, thus inhibiting HCV-cell fusion.
Building upon our observation that HCV entry can be inhibited by increasing target membrane fluidity, we developed a screen using liposomes and the lipophilic dye Prodan. We screened a library of 2,752 compounds and identified a molecule, topotecan, that preferentially increases the fluidity of cholesterol-rich membranes (
Fig. 6A). Using a well-established HCVcc-liposome fusion assay (
32,
33), we showed that topotecan dose-dependently inhibits HCVcc-membrane fusion
in vitro, validating the screen as a tool for discovering inhibitors of virus-cell fusion (
Fig. 6B). However, in cell culture assays, the anti-HCV activity of topotecan appears to derive primarily from its inhibition of HCV replication rather than entry, with an estimated EC
50 of ∼0.2 μM (
Fig. 7). Topotecan (Hycamtin) is a topoisomerase I inhibitor that acts by stabilizing the single-stranded DNA (ssDNA)-topoisomerase I complex and causing DNA cleavage (
62). It is currently used in chemotherapy for various cancers. In addition to topoisomerase, topotecan is known to affect many other cellular pathways, including downregulation of the phosphatidylinositol 3-kinase (PI3K)–Akt signaling pathway (
63) and disruption of the hypoxia inducible factor 1 (HIF-1) signaling pathway (
64). Topotecan is also a substrate of the ABC transporters P-glycoprotein (P-gp/MDR1) and breast cancer resistance protein (BCRP) and is actively cleared by the cell (
65). The lack of HCV entry inhibition by topotecan in cell culture assays may be due in part to active cellular extrusion and/or intracellular trafficking of the compound. Nevertheless, the ability of topotecan to inhibit HCV replication may warrant additional clinical studies of this compound.
In conclusion, our studies shed light on the mechanism of action of phenothiazines as inhibitors of HCV entry and showed, for the first time, that alteration of target host cell membrane fluidity can inhibit HCV entry. It is possible that the same mechanism is responsible for the antiviral activities of phenothiazines toward other viruses, such as inhibiting the budding of measles and herpes simplex viruses (
27). Based on these insights, we developed a high-throughput screen for modulators of cholesterol-rich membrane fluidity and screened a library of 2,752 compounds. One hit from this screen—topotecan—was found to both increase the fluidity of cholesterol-rich membranes and inhibit the fusion of these membranes with HCV. Targeting an entry step independent of viral proteins may also be an effective way to retard the development of drug resistance and inhibit HCV deletion mutants, which were found to reduce the antiviral effects of interferon therapy for chronic hepatitis C patients (
66). This study represents an exciting new paradigm for exploring additional membrane-targeting antivirals.