1887

Abstract

Hepatitis C virus (HCV) affects about 170 million people worldwide. The current treatment has a high cost and variable response rates according to the virus genotype. Acridones, a group of compounds extracted from natural sources, showed potential antiviral actions against HCV. Thus, this study aimed to evaluate the effect of a panel of 14 synthetic acridones on the HCV life cycle. The compounds were screened using an Huh7.5 cell line stably harbouring the HCV genotype 2a subgenomic replicon SGR-Feo-JFH-1. Cells were incubated in the presence or absence of compounds for 72 h and cell viability and replication levels were assessed by MTT and luciferase assays, respectively. At a concentration of 5 µM the acridone Fac4 exhibited a >90 % inhibition of HCV replication with no effect on cell viability. The effects of Fac4 on virus replication, entry and release steps were evaluated in Huh7.5 cells infected with the JFH-1 isolate of HCV (HCVcc). Fac4 inhibited JFH-1 replication to approximately 70 %, while no effect was observed on virus entry. The antiviral activity of Fac4 was also observed on viral release, with almost 80 % of inhibition. No inhibitory effect was observed against genotype 3 replication. Fac4 was able to intercalate into dsRNA, however did not inhibit NS5B polymerase activity or translation driven by the HCV IRES. Although its mode of action is partly understood, Fac4 presents significant inhibition of HCV replication and can therefore be considered as a candidate for the development of a future anti-HCV treatment.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.000808
2017-07-01
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/jgv/98/7/1693.html?itemId=/content/journal/jgv/10.1099/jgv.0.000808&mimeType=html&fmt=ahah

References

  1. Alter HJ, Seeff LB. Recovery, persistence, and sequelae in hepatitis C virus infection: a perspective on long-term outcome. Semin Liver Dis 2000; 20:17–35 [View Article][PubMed]
    [Google Scholar]
  2. Houghton M. The long and winding road leading to the identification of the hepatitis C virus. J Hepatol 2009; 51:939–948 [View Article][PubMed]
    [Google Scholar]
  3. Baltimore D. Expression of animal virus genomes. Bacteriol Rev 1971; 35:235–241[PubMed]
    [Google Scholar]
  4. Penin F, Dubuisson J, Rey FA, Moradpour D, Pawlotsky JM. Structural biology of hepatitis C virus. Hepatology 2004; 39:5–19 [View Article][PubMed]
    [Google Scholar]
  5. Simmonds P, Becher P, Bukh J, Gould EA, Meyers G et al. ICTV virus taxonomy profile: Flaviviridae. J Gen Virol 2017; 98:2–3 [View Article][PubMed]
    [Google Scholar]
  6. Suzuki T, Ishii K, Aizaki H, Wakita T. Hepatitis C viral life cycle. Adv Drug Deliv Rev 2007; 59:1200–1212 [View Article][PubMed]
    [Google Scholar]
  7. Dustin LB, Rice CM. Flying under the radar: the immunobiology of hepatitis C. Annu Rev Immunol 2007; 25:71–99 [View Article][PubMed]
    [Google Scholar]
  8. Lindenbach BD, Rice CM. Unravelling hepatitis C virus replication from genome to function. Nature 2005; 436:933–938 [View Article][PubMed]
    [Google Scholar]
  9. Argentini C, Genovese D, Dettori S, Rapicetta M. HCV genetic variability: from quasispecies evolution to genotype classification. Future Microbiol 2009; 4:359–373 [View Article][PubMed]
    [Google Scholar]
  10. Murphy D, Chamberland J, Dandavino R, Sablon E. A new genotype of hepatitis C virus orginating from central Africa. Hepatology 2007; 46:623
    [Google Scholar]
  11. Simmonds P, Bukh J, Combet C, Deléage G, Enomoto N et al. Consensus proposals for a unified system of nomenclature of hepatitis C virus genotypes. Hepatology 2005; 42:962–973 [View Article][PubMed]
    [Google Scholar]
  12. Simmonds P, Holmes EC, Cha TA, Chan SW, Mcomish F et al. Classification of hepatitis C virus into six major genotypes and a series of subtypes by phylogenetic analysis of the NS-5 region. J Gen Virol 1993; 74:2391–2399 [View Article][PubMed]
    [Google Scholar]
  13. Cristina J, del Pilar Moreno M, Moratorio G. Hepatitis C virus genetic variability in patients undergoing antiviral therapy. Virus Res 2007; 127:185–194 [View Article][PubMed]
    [Google Scholar]
  14. Davis GL. Hepatitis C virus genotypes and quasispecies. Am J Med 1999; 107:21–26 [View Article]
    [Google Scholar]
  15. Martell M, Esteban JI, Genescà J, Weiner A, Weiner A et al. Hepatitis C virus (HCV) circulates as a population of different but closely related genomes: quasispecies nature of HCV genome distribution. J Virol 1992; 66:3225–3229[PubMed]
    [Google Scholar]
  16. Pawlotsky JM. Hepatitis C virus population dynamics during infection. Curr Top Microbiol Immunol 2006; 299:261–284[PubMed]
    [Google Scholar]
  17. Le Guillou-Guillemette H, Vallet S, Gaudy-Graffin C, Payan C, Pivert A et al. Genetic diversity of the hepatitis C virus: impact and issues in the antiviral therapy. World J Gastroenterol 2007; 13:2416–2426 [View Article][PubMed]
    [Google Scholar]
  18. Gao M, Nettles RE, Belema M, Snyder LB, Nguyen VN et al. Chemical genetics strategy identifies an HCV NS5A inhibitor with a potent clinical effect. Nature 2010; 465:96–100 [View Article][PubMed]
    [Google Scholar]
  19. Lawitz E, Lalezari JP, Hassanein T, Kowdley KV, Poordad FF et al. Sofosbuvir in combination with peginterferon alfa-2a and ribavirin for non-cirrhotic, treatment-naive patients with genotypes 1, 2, and 3 hepatitis C infection: a randomised, double-blind, phase 2 trial. Lancet Infect Dis 2013; 13:401–408 [View Article][PubMed]
    [Google Scholar]
  20. Rosenquist Å, Samuelsson B, Johansson PO, Cummings MD, Lenz O et al. Discovery and development of simeprevir (TMC435), a HCV NS3/4A protease inhibitor. J Med Chem 2014; 57:1673–1693 [View Article][PubMed]
    [Google Scholar]
  21. Jacobson IM, Gordon SC, Kowdley KV, Yoshida EM, Rodriguez-Torres M et al. Sofosbuvir for hepatitis C genotype 2 or 3 in patients without treatment options. N Engl J Med 2013; 368:1867–1877 [View Article][PubMed]
    [Google Scholar]
  22. Munir S, Saleem S, Idrees M, Tariq A, Butt S et al. Hepatitis C treatment: current and future perspectives. Virol J 2010; 7:296 [View Article][PubMed]
    [Google Scholar]
  23. Newman DJ, Cragg GM. Natural products as sources of new drugs from 1981 to 2014. J Nat Prod 2016; 79:629–661 [View Article][PubMed]
    [Google Scholar]
  24. Michael JP. Quinoline, quinazoline and acridone alkaloids. Nat Prod Rep 2008; 25:166–187 [View Article][PubMed]
    [Google Scholar]
  25. Alwan WS, Mahajan AA, Rane RA, Amritkar AA, Naphade SS et al. Acridone-based antitumor agents: a mini-review. Anticancer Agents Med Chem 2015; 15:1012–1025 [View Article][PubMed]
    [Google Scholar]
  26. Fujiwara M, Okamoto M, Okamoto M, Watanabe M, Machida H et al. Acridone derivatives are selective inhibitors of HIV-1 replication in chronically infected cells. Antiviral Res 1999; 43:189–199 [View Article][PubMed]
    [Google Scholar]
  27. Turpin JA, Buckheit RW, Derse D, Hollingshead M, Williamson K et al. Inhibition of acute-, latent-, and chronic-phase human immunodeficiency virus type 1 (HIV-1) replication by a bistriazoloacridone analog that selectively inhibits HIV-1 transcription. Antimicrob Agents Chemother 1998; 42:487–494[PubMed]
    [Google Scholar]
  28. Yamamoto N, Furukawa H, Ito Y, Yoshida S, Maeno K et al. Anti-herpesvirus activity of citrusinine-I, a new acridone alkaloid, and related compounds. Antiviral Res 1989; 12:21–36 [View Article][PubMed]
    [Google Scholar]
  29. Stankiewicz-Drogoń A, Dörner B, Erker T, Boguszewska-Chachulska AM. Synthesis of new acridone derivatives, inhibitors of NS3 helicase, which efficiently and specifically inhibit subgenomic HCV replication. J Med Chem 2010; 53:3117–3126 [View Article][PubMed]
    [Google Scholar]
  30. Stankiewicz-Drogon A, Palchykovska LG, Kostina VG, Alexeeva IV, Shved AD et al. New acridone-4-carboxylic acid derivatives as potential inhibitors of hepatitis C virus infection. Bioorg Med Chem 2008; 16:8846–8852 [View Article][PubMed]
    [Google Scholar]
  31. Sepúlveda CS, Fascio ML, García CC, D'Accorso NB, Damonte EB. Acridones as antiviral agents: synthesis, chemical and biological properties. Curr Med Chem 2013; 20:2402–2414 [View Article][PubMed]
    [Google Scholar]
  32. Chansriniyom C, Ruangrungsi N, Lipipun V, Kumamoto T, Ishikawa T. Isolation of acridone alkaloids and N-[(4-monoterpenyloxy)phenylethyl]-substituted sulfur-containing propanamide derivatives from Glycosmis parva and their anti-herpes simplex virus activity. Chem Pharm Bull 2009; 57:1246–1250 [View Article][PubMed]
    [Google Scholar]
  33. Itoigawa M, Ito C, Wu TS, Enjo F, Tokuda H et al. Cancer chemopreventive activity of acridone alkaloids on Epstein-Barr virus activation and two-stage mouse skin carcinogenesis. Cancer Lett 2003; 193:133–138 [View Article][PubMed]
    [Google Scholar]
  34. Takemura Y, Ju-Ichi M, Ito C, Furukawa H, Tokuda H. Studies on the inhibitory effects of some acridone alkaloids on Epstein-Barr virus activation. Planta Med 1995; 61:366–368 [View Article][PubMed]
    [Google Scholar]
  35. Houe H. Economic impact of BVDV infection in dairies. Biologicals 2003; 31:137–143 [View Article][PubMed]
    [Google Scholar]
  36. Mazzucco MB, Talarico LB, Vatansever S, Carro AC, Fascio ML et al. Antiviral activity of an N-allyl acridone against dengue virus. J Biomed Sci 2015; 22:29 [View Article][PubMed]
    [Google Scholar]
  37. Raney KD, Sharma SD, Moustafa IM, Cameron CE. Hepatitis C virus non-structural protein 3 (HCV NS3): a multifunctional antiviral target. J Biol Chem 2010; 285:22725–22731 [View Article][PubMed]
    [Google Scholar]
  38. Sepúlveda CS, Fascio ML, Mazzucco MB, Palacios ML, Pellón RF et al. Synthesis and evaluation of N-substituted acridones as antiviral agents against haemorrhagic fever viruses. Antivir Chem Chemother 2008; 19:41–47 [View Article][PubMed]
    [Google Scholar]
  39. Tabarrini O, Manfroni G, Fravolini A, Cecchetti V, Sabatini S et al. Synthesis and anti-BVDV activity of acridones as new potential antiviral agents. J Med Chem 2006; 49:2621–2627 [View Article][PubMed]
    [Google Scholar]
  40. Gentile I, Buonomo AR, Borgia F, Zappulo E, Castaldo G et al. MK-5172 : a second-generation protease inhibitor for the treatment of hepatitis C virus infection. Expert Opin Investig Drugs 2014; 23:719–728 [View Article][PubMed]
    [Google Scholar]
  41. Hayashi N, Izumi N, Kumada H, Okanoue T, Tsubouchi H et al. Simeprevir with peginterferon/ribavirin for treatment-naïve hepatitis C genotype 1 patients in Japan: CONCERTO-1, a phase III trial. J Hepatol 2014; 61:219–227 [View Article][PubMed]
    [Google Scholar]
  42. Summa V, Ludmerer SW, McCauley JA, Fandozzi C, Burlein C et al. MK-5172, a selective inhibitor of hepatitis C virus NS3/4a protease with broad activity across genotypes and resistant variants. Antimicrob Agents Chemother 2012; 56:4161–4167 [View Article][PubMed]
    [Google Scholar]
  43. Adams A. Crystal structures of acridines complexed with nucleic acids. Curr Med Chem 2002; 9:1667–1675 [View Article][PubMed]
    [Google Scholar]
  44. Manfroni G, Paeshuyse J, Massari S, Zanoli S, Gatto B et al. Inhibition of subgenomic hepatitis C virus RNA replication by acridone derivatives: identification of an NS3 helicase inhibitor. J Med Chem 2009; 52:3354–3365 [View Article][PubMed]
    [Google Scholar]
  45. Kovalenko AL, Kazakov VI, Slita AV, Zarubaev VV, Sukhinin VP. Intracellular localization of cycloferon, its binding with DNA and stimulation of cytokines expression after exposure to cycloferon. Tsitologiia 2000; 42:659–664[PubMed]
    [Google Scholar]
  46. Storch E, Kirchner H, Brehm G, Hüller K, Marcucci F. Production of interferon-β by murine T-cell lines induced by 10-carboxymethyl-9-acridanone. Scand J Immunol 1986; 23:195–199 [View Article][PubMed]
    [Google Scholar]
  47. Herath HMTB, Muller K, Diyabalanage HVK. Synthesis of acrimarins from 1,3,5-trioxygenated-9-acridone derivatives. J Heterocycl Chem 2004; 41:23–28 [View Article]
    [Google Scholar]
  48. Wyles DL, Kaihara KA, Korba BE, Schooley RT, Beadle JR et al. The octadecyloxyethyl ester of (S)-9-[3-hydroxy-2-(phosphonomethoxy) propyl]adenine is a potent and selective inhibitor of hepatitis C virus replication in genotype 1A, 1B, and 2A replicons. Antimicrob Agents Chemother 2009; 53:2660–2662 [View Article][PubMed]
    [Google Scholar]
  49. Saeed M, Scheel TK, Gottwein JM, Marukian S, Dustin LB et al. Efficient replication of genotype 3a and 4a hepatitis C virus replicons in human hepatoma cells. Antimicrob Agents Chemother 2012; 56:5365–5373 [View Article][PubMed]
    [Google Scholar]
  50. Wakita T, Pietschmann T, Kato T, Date T, Miyamoto M et al. Production of infectious hepatitis C virus in tissue culture from a cloned viral genome. Nat Med 2005; 11:791–796 [View Article][PubMed]
    [Google Scholar]
  51. Macdonald A, Crowder K, Street A, McCormick C, Saksela K et al. The hepatitis C virus non-structural NS5A protein inhibits activating protein-1 function by perturbing ras-ERK pathway signaling. J Biol Chem 2003; 278:17775–17784 [View Article][PubMed]
    [Google Scholar]
  52. Nahmias Y, Goldwasser J, Casali M, van Poll D, Wakita T et al. Apolipoprotein B-dependent hepatitis C virus secretion is inhibited by the grapefruit flavonoid naringenin. Hepatology 2008; 47:1437–1445 [View Article][PubMed]
    [Google Scholar]
  53. Krawczyk M, Wasowska-Lukawska M, Oszczapowicz I, Boguszewska-Chachulska AM. Amidinoanthracyclines – a new group of potential anti-hepatitis C virus compounds. Biol Chem 2009; 390:351–360 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.000808
Loading
/content/journal/jgv/10.1099/jgv.0.000808
Loading

Data & Media loading...

Supplements

Supplementary File 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error