Inhibitors of alphavirus entry and replication identified with a stable Chikungunya replicon cell line and virus-based assays

Leena Pohjala, Age Utt, Margus Varjak, Aleksei Lulla, Andres Merits, Tero Ahola, Päivi Tammela, Leena Pohjala, Age Utt, Margus Varjak, Aleksei Lulla, Andres Merits, Tero Ahola, Päivi Tammela

Abstract

Chikungunya virus (CHIKV), an alphavirus, has recently caused epidemic outbreaks and is therefore considered a re-emerging pathogen for which no effective treatment is available. In this study, a CHIKV replicon containing the virus replicase proteins together with puromycin acetyltransferase, EGFP and Renilla luciferase marker genes was constructed. The replicon was transfected into BHK cells to yield a stable cell line. A non-cytopathic phenotype was achieved by a Pro718 to Gly substitution and a five amino acid insertion within non-structural protein 2 (nsP2), obtained through selection for stable growth. Characterization of the replicon cell line by Northern blotting analysis revealed reduced levels of viral RNA synthesis. The CHIKV replicon cell line was validated for antiviral screening in 96-well format and used for a focused screen of 356 compounds (natural compounds and clinically approved drugs). The 5,7-dihydroxyflavones apigenin, chrysin, naringenin and silybin were found to suppress activities of EGFP and Rluc marker genes expressed by the CHIKV replicon. In a concomitant screen against Semliki Forest virus (SFV), their anti-alphaviral activity was confirmed and several additional inhibitors of SFV with IC₅₀ values between 0.4 and 24 µM were identified. Chlorpromazine and five other compounds with a 10H-phenothiazinyl structure were shown to inhibit SFV entry using a novel entry assay based on a temperature-sensitive SFV mutant. These compounds also reduced SFV and Sindbis virus-induced cytopathic effect and inhibited SFV virion production in virus yield experiments. Finally, antiviral effects of selected compounds were confirmed using infectious CHIKV. In summary, the presented approach for discovering alphaviral inhibitors enabled us to identify potential lead structures for the development of alphavirus entry and replication phase inhibitors as well as demonstrated the usefulness of CHIKV replicon and SFV as biosafe surrogate models for anti-CHIKV screening.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Construction and characterization of a…
Figure 1. Construction and characterization of a stable BHK cell line carrying CHIKV-replicon.
A) Schematic representation of the used CHIKV replicons (numbers and symbols are explained in the text). B) The process leading to selection of non-cytotoxic (NCT) CHIKV replicons, identification of mutations associated with the NCT phenotype and confirmation of their phenotypes. C) Phenotype of BHK-CHIKV-NCT cells; green fluorescence is caused by EGFP expression. Arrow indicates a cell in the process of division. D) Immunofluorescence images of BHK-CHIKV-NCT cells stained with anti-dsRNA (top), anti-SFV nsP3 (middle) and co-staining with anti-dsRNA and anti-SFV nsP3 (bottom). A representative optical slice from the middle of the cell is shown. Scale bar is 10 µm.
Figure 2. Effects of adaptive mutations on…
Figure 2. Effects of adaptive mutations on the CHIKV replicon.
A) Effects of the PG and NCT mutations on the accumulation of positive-strand replicon and corresponding sgRNAs in cells transfected with in vitro transcripts of CHIKV-LR, CHIKV-PG, CHIKV-NCT and their variants containing the Rluc marker in the nsP3 region. Total RNAs extracted from transfected cells at 16 h post-transfection; 10 µg aliquot from each sample was separated by electrophoresis in formaldehyde gel and analyzed by Northern blotting. The constructs are shown at the top; positions of replicon and sgRNAs are indicated with arrows. “Mock” indicates RNAs from mock-transfected control cells. B) Immunofluorescence analysis of nsP2 localization in cells transfected with in vitro transcripts of CHIKV-LR or CHIKV-PG at 8 h post-infection or with in vitro transcripts of CHIKV-NCT at 16 h post-infection. Cells were fixed and stained with anti-nsP2 and DAPI; EGFP was detected by fluorescence. Merge indicates co-staining of DAPI and nsP2.
Figure 3. Virus entry assays with SFVts9-…
Figure 3. Virus entry assays with SFVts9-Rluc.
A) The temperature-sensitive phenotype of SFVts9-Rluc as measured by Rluc activity of infected cell lysates at 1 h, 2 h and 3 h post-infection at 39°C. B) The effect of chloroquine on Rluc signals of SFVts9-Rluc at 39°C. C) Effects of 5,7-dihydroxyflavones and 10H-phenothiazines on the accumulation of Rluc in cells infected with SFVts9-Rluc at 39°C. Selected examples of results of hit compounds in SFV entry inhibition assay. The cell cultures were treated with 100 µM compounds and Rluc levels were determined 1 h post-infection. All results represent the average of three replicates.

References

    1. Gubler DJ. The global emergence/resurgence of arboviral diseases as public health problems. Arch Med Res. 2002;33:330–342.
    1. Weaver SC, Reisen WK. Present and future arboviral threats. Antiviral Res. 2010;85:328–345.
    1. Strauss JH, Strauss EG. The alphaviruses: gene expression, replication, and evolution. Microbiol Rev. 1994;58:491–562.
    1. Pialoux G, Gauzere BA, Jaureguiberry S, Strobel M. Chikungunya, an epidemic arbovirosis. Lancet Infect Dis. 2007;7:319–327.
    1. Powers AM, Logue CH. Changing patterns of Chikungunya virus: re-emergence of a zoonotic arbovirus. J Gen Virol. 2007;88:2363–2377.
    1. Panning M, Grywna K, van Esbroeck M, Emmerich P, Drosten C. Chikungunya fever in travelers returning to Europe from the Indian Ocean region, 2006. Emerg Infect Dis. 2008;14:416–422.
    1. Rezza G, Nicoletti L, Angelini R, Romi R, Finarelli AC, et al. Infection with Chikungunya virus in Italy: an outbreak in a temperate region. Lancet. 2007;370:1840–1846.
    1. Kam YW, Ong EK, Renia L, Tong JC, Ng LF. Immuno-biology of Chikungunya and implications for disease intervention. Microbes Infect. 2009;11:1186–1196.
    1. Sissoko D, Malvy D, Ezzedine K, Renault P, Moscetti F, et al. Post-epidemic Chikungunya disease on Reunion Island: course of rheumatic manifestations and associated factors over a 15–month period. PLoS Negl Trop Dis. 2009;3:e389.
    1. Lidbury BA, Rulli NE, Suhrbier A, Smith PN, McColl SR, et al. Macrophage-derived proinflammatory factors contribute to the development of arthritis and myositis after infection with an arthrogenic alphavirus. J Infect Dis. 2008;197:1585–1593.
    1. Couderc T, Chretien F, Schilte C, Disson O, Brigitte M, et al. A mouse model for Chikungunya: young age and inefficient type-I interferon signaling are risk factors for severe disease. PLoS Pathog. 2008;4:e29.
    1. Ozden S, Huerre M, Riviere JP, Coffey LL, Afonso PV, et al. Human muscle satellite cells as targets of Chikungunya virus infection. PLoS One. 2007;2:e527.
    1. Ziegler SA, Lu L, da Rosa AP, Xiao SY, Tesh RB. An animal model for studying the pathogenesis of Chikungunya virus infection. Am J Trop Med Hyg. 2008;79:133–139.
    1. Robin S, Ramful D, Le Seach F, Jaffar-Bandjee MC, Rigou G, et al. Neurologic manifestations of pediatric Chikungunya infection. J Child Neurol. 2008;23:1028–1035.
    1. De Lamballerie X, Boisson V, Reynier JC, Enault S, Charrel RN, et al. On chikungunya acute infection and chloroquine treatment. Vector Borne Zoonotic Dis. 2008;8:837–839.
    1. Briolant S, Garin D, Scaramozzino N, Jouan A, Crance JM. In vitro inhibition of Chikungunya and Semliki Forest viruses replication by antiviral compounds: synergistic effect of interferon-alpha and ribavirin combination. Antiviral Res. 2004;61:111–117.
    1. De Clercq E. Antiviral and antimetabolic activities of neplanocins. Antimicrob Agents Chemother. 1985;28:84–89.
    1. De Clercq E, Bernaerts R, Shealy YF, Montgomery JA. Broad-spectrum antiviral activity of carbodine, the carbocyclic analogue of cytidine. Biochem Pharmacol. 1990;39:319–325.
    1. Tsetsarkin K, Higgs S, McGee CE, De Lamballerie X, Charrel RN, et al. Infectious clones of Chikungunya virus (La Reunion isolate) for vector competence studies. Vector Borne Zoonotic Dis. 2006;6:325–337.
    1. Dryga SA, Dryga OA, Schlesinger S. Identification of mutations in a Sindbis virus variant able to establish persistent infection in BHK cells: the importance of a mutation in the nsP2 gene. Virology. 1997;228:74–83.
    1. Frolov I, Agapov E, Hoffman TA, Jr, Pragai BM, Lippa M, et al. Selection of RNA replicons capable of persistent noncytopathic replication in mammalian cells. J Virol. 1999;73:3854–3865.
    1. Tamm K, Merits A, Sarand I. Mutations in the nuclear localization signal of nsP2 influencing RNA synthesis, protein expression and cytotoxicity of Semliki Forest virus. J Gen Virol. 2008;89:676–686.
    1. Rikkonen M, Peränen J, Kääriäinen L. Nuclear and nucleolar targeting signals of Semliki Forest virus nonstructural protein nsP2. Virology. 1992;189:462–473.
    1. Spuul P, Balistreri G, Kääriäinen L, Ahola T. Phosphatidylinositol 3-kinase-, actin- and microtubule-dependent transport of Semliki Forest virus replication complexes from the plasma membrane to modified lysosomes. J Virol. 2010;84:7543–7557.
    1. Casales E, Rodriguez-Madoz JR, Ruiz-Guillen M, Razquin N, Cuevas Y, et al. Development of a new noncytopathic Semliki Forest virus vector providing high expression levels and stability. Virology. 2008;376:242–251.
    1. Helenius A, Marsh M, White J. Inhibition of Semliki forest virus penetration by lysosomotropic weak bases. J Gen Virol. 1982;58 Pt 1:47–61.
    1. Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects of chloroquine on viral infections: an old drug against today's diseases? Lancet Infect Dis. 2003;3:722–727.
    1. Diamond MS, Zachariah M, Harris E. Mycophenolic acid inhibits dengue virus infection by preventing replication of viral RNA. Virology. 2002;304:211–221.
    1. Leyssen P, Balzarini J, De Clercq E, Neyts J. The predominant mechanism by which ribavirin exerts its antiviral activity in vitro against flaviviruses and paramyxoviruses is mediated by inhibition of IMP dehydrogenase. J Virol. 2005;79:1943–1947.
    1. Pohjala L, Barai V, Azhayev A, Lapinjoki S, Ahola T. A luciferase-based screening method for inhibitors of alphavirus replication applied to nucleoside analogues. Antiviral Res. 2008;78:215–222.
    1. Balistreri G, Caldentey J, Kääriäinen L, Ahola T. Enzymatic defects of the nsP2 proteins of Semliki Forest virus temperature-sensitive mutants. J Virol. 2007;81:2849–2860.
    1. Lulla V, Merits A, Sarin P, Kääriäinen L, Keränen S, et al. Identification of mutations causing temperature-sensitive defects in Semliki Forest virus RNA synthesis. J Virol. 2006;80:3108–3111.
    1. Gould EA, Coutard B, Malet H, Morin B, Jamal S, et al. Understanding the alphaviruses: Recent research on important emerging pathogens and progress towards their control. Antiviral Res. 2010;87:111–124.
    1. Powers AM, Brault AC, Shirako Y, Strauss EG, Kang W, et al. Evolutionary relationships and systematics of the alphaviruses. J Virol. 2001;75:10118–10131.
    1. Frolov I, Schlesinger S. Comparison of the effects of Sindbis virus and Sindbis virus replicons on host cell protein synthesis and cytopathogenicity in BHK cells. J Virol. 1994;68:1721–1727.
    1. Garmashova N, Gorchakov R, Frolova E, Frolov I. Sindbis virus non-structural protein nsP2 is cytotoxic and inhibits cellular transcription. J Virol. 2006;80:5686–5696.
    1. Sawicki DL, Perri S, Polo JM, Sawicki SG. Role for nsP2 proteins in the cessation of alphavirus minus-strand synthesis by host cells. J Virol. 2006;80:360–371.
    1. Perri S, Driver SA, Gardner JP, Sherrill S, Belli BA, et al. Replicon vectors derived from Sindbis virus and Semliki forest virus that establish persistent replication in host cells. J Virol. 2000;74:9802–9807.
    1. Agapov EV, Frolov I, Lindenbach BD, Pragai BM, Schlesinger S, et al. Noncytopathic Sindbis virus RNA vectors for heterologous gene expression. Proc Natl Acad Sci U S A. 1998;95:12989–12994.
    1. Frolov I, Garmashova N, Atasheva S, Frolova EI. Random insertion mutagenesis of sindbis virus nonstructural protein 2 and selection of variants incapable of downregulating cellular transcription. J Virol. 2009;83:9031–9044.
    1. Fros J, Liu WJ, Prow NA, Geertsema C, Ligtenberg M, et al. Chikungunya virus nonstructural protein 2 inhibits type I/II interferon-stimulated JAK-STAT signaling. J Virol. 2010;84:10877–87.
    1. Asres K, Seyoum A, Veeresham C, Bucar F, Gibbons S. Naturally derived anti-HIV agents. Phytother Res. 2005;19:557–581.
    1. Jassim SA, Naji MA. Novel antiviral agents: a medicinal plant perspective. J Appl Microbiol. 2003;95:412–427.
    1. Blanchard E, Belouzard S, Goueslain L, Wakita T, Dubuisson J, et al. Hepatitis C virus entry depends on clathrin-mediated endocytosis. J Virol. 2006;80:6964–6972.
    1. Gastaminza P, Whitten-Bauer C, Chisari FV. Unbiased probing of the entire hepatitis C virus life cycle identifies clinical compounds that target multiple aspects of the infection. Proc Natl Acad Sci USA. 2010;107:291–296.
    1. Wang LH, Rothberg KG, Anderson RG. Mis-assembly of clathrin lattices on endosomes reveals a regulatory switch for coated pit formation. J Cell Biol. 1993;123:1107–1117.
    1. Das T, Jaffar-Bandjee MC, Hoarau JJ, Trotot PK, Denizot M, et al. Chikungunya fever: CNS infection and pathologies of a re-emerging arbovirus. Prog Neurobiol. 2010;91:121–129.
    1. Sourisseau M, Schilte C, Casartelli N, Trouillet C, Guivel-Benhassine F, et al. Characterization of reemerging Chikungunya virus. PLoS Patho. 2007;3:e89.
    1. Liljeström P, Lusa S, Huylebroeck D, Garoff H. In vitro mutagenesis of a full-length cDNA clone of Semliki Forest virus: the small 6,000-molecular-weight membrane protein modulates virus release. J Virol. 1991;65:4107–4113.
    1. Rice CM, Levis R, Strauss JH, Huang HV. Production of infectious RNA transcripts from Sindbis virus cDNA clones: mapping of lethal mutations, rescue of a temperature-sensitive marker, and in vitro mutagenesis to generate defined mutants. J Virol. 1987;61:3809–3819.
    1. Crouch SP, Kozlowski R, Slater KJ, Fletcher J. The use of ATP bioluminescence as a measure of cell proliferation and cytotoxicity. J Immunol Methods. 1993;160:81–88.
    1. Huffman JH, Sidwell RW, Khare GP, Witkowski JT, Allen LB. In vitro effect of 1-beta-D-ribofuranosyl-1,2,4-triazole-3-carboxamide (virazole, ICN 1229) on deoxyribonucleic acid and ribonucleic acid viruses. Antimicrob Agents Chemother. 1973;3:235–241.

Source: PubMed

3
購読する