Chloroquine interferes with dengue-2 virus replication in U937 cells

Kleber Juvenal Silva Farias, Paula Renata Lima Machado, Renato Ferreira de Almeida Junior, Ana Alice de Aquino, Benedito Antônio Lopes da Fonseca, Kleber Juvenal Silva Farias, Paula Renata Lima Machado, Renato Ferreira de Almeida Junior, Ana Alice de Aquino, Benedito Antônio Lopes da Fonseca

Abstract

The objective of this study was to investigate the use of chloroquine (CLQ) as an antiviral agent against dengue. Chloroquine, an amine acidotropic drug known to affect intracellular exocytic pathways by increasing endosomal pH, was used in the in vitro treatment of U937 cells infected with dengue virus type 2 (DENV-2). Viral replication was assessed by quantification of virus produced through detection of copy numbers of DENV-2 RNA, plaque assay and indirect immunofluorescence. qRT-PCR and plaque assays were used to quantify the DENV-2 load in infected U937 cells after CLQ treatment. It was found that a dose of 50 μg/mL of CLQ was not toxic to the cells and resulted in significantly less virus production in infected U937 cells than occurred in untreated cells. In the present work, CLQ was effective against DENV-2 replication in U937 cells, and also caused a statistically significant reduction in expression of proinflammatory cytokines. The present study indicates that CLQ may be used to reduce viral yield in U937 cells.

Keywords: DENV-2; chloroquine; plaque assay; quantitative reverse transcription polymerase chain reaction.

© 2014 The Societies and Wiley Publishing Asia Pty Ltd.

Figures

Figure 1
Figure 1
The effect of CLQ on the cytotoxicity in U937 cells. Concentrations equal or higher than 500 μg/mL (CLQ) were highly cytotoxic to U937 cells, while concentrations equal or lower than 50 μg/mL (CLQ) did not induce significant cytotoxicity.
Figure 2
Figure 2
Action of CLQ on DENV‐2 replication in U937 cells. (a) The viral RNA present in the culture supernatants of U937cells infected with DENV‐2, treated and untreated CLQ post‐infection, was extracted and analyzed by qRT‐PCR. (b) Culture supernatants of U937cells infected with DENV‐2, treated and untreated CLQ post‐infection, were analyzed by plaque assay. The results represent the average values of the viral RNA copy number (P  < 0.001).
Figure 3
Figure 3
Action of CLQ added at 24‐hr intervals on DENV‐2 replication in U937 cells. (a) The viral RNA present in the culture supernatants of U937 cells infected with DENV‐2, treated and untreated CLQ (every 24 hr post‐infection), was extracted and analyzed by qRT‐PCR. (b) Culture supernatants of U937 cells infected with DENV‐2, treated and untreated CLQ (every 24 hr post‐infection), was analyzed by plaque assay. The results represent the average values of the viral RNA copy number (P  < 0.05).
Figure 4
Figure 4
Action of CLQ added at 12‐hr intervals on DENV‐2 replication in U937 cells. The viral RNA present in the culture supernatants of U937 cells infected with DENV‐2, treated and untreated CLQ (every 12 hr post‐infection), was extracted and analyzed by qRT‐PCR. The results represent the average values of the viral RNA copy number (P  < 0.05).
Figure 5
Figure 5
Indirect immunofluorescence assay. Detection of DENV‐2 virus in U937 cells infected or uninfected, treated and untreated CLQ (every 24 hr post‐infection). (Original magnification 200–1,000×).
Figure 6
Figure 6
Relative quantification of cytokines by real‐time PCR in U937 cells infected or not with DENV‐2 in the presence or absence of CLQ added at 24‐hr intervals. IFN‐α (a), IFN‐β (b), IFN‐γ (c), TNF‐α (d), IL‐6 (e), IL‐12 (f), and IL‐10 (g) mRNA expression in DENV‐2 infected or uninfected U937 cells, treated and untreated CLQ were analyzed by quantitative RT‐PCR, and normalized to mouse beta actin gene. Values were submitted to t test in which *P < 0.05 vs. U937 cells infected with DENV‐2s.

References

    1. Karabatsos N.( 1985) International Catalog of Arboviruses Including Certain Other Viruses of Vertebrates. 3rd edn. Fort Collins, CO: American Society of Tropical Medicine & Hygiene, p. 1147.
    1. Gibbons R.V., Vaughn D.W. ( 2002) Dengue: An escalating problem. Br Med J 324: 1563–6.
    1. Rigau‐pérez J.G., Clark G.G., Gubler J.D., Reiter P., Sanders J.E., Vorndam V. ( 1998) Dengue and dengue haemorrhagic fever. Lancet 353: 971–7.
    1. Monath T.P., Burke D.S.( 2001) Flaviviruses. Fields Virology. Vol. 33, 4th edn. Philadelphia: Lippincott Willians & Wilkins, pp. 1043–1126.
    1. Rey F.A. ( 2003) Dengue virus envelope glycoprotein structure: New insight into its interactions during viral entry. Proc Natl Acad Sci U S A 100: 6899–901.
    1. Chambers T.C., Hahn C.S., Galler R., Rice C.M. ( 1990) Flavivirus genome organization, expression, and replication. Annu Rev Microbiol 44: 649–88.
    1. Rice C.M. ( 1996) Flaviviridae: The viruses and their replication. Fields Virology. In: Fields BN, Knipe DM, Howley PMed. 3rd edn. New York: Lippincott‐Raven, pp. 931–959.
    1. Tassaneetrithep B., Burgess T.H., Granelli‐piperno A., Trumpfheller C., Finke J., Sun W., Eller M.A., Pattanapanyasat K., Sarasombath S., Birx D.L., Steinman R.M., Schlesinger S., Marovich M.A. ( 2003) DC‐SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med 197: 823–9.
    1. Lozach P.Y., Burleigh L., Staropoli I., Navarro‐sanchez E., Harriague J., Virelizier J.L., Rey F.A., Despres P., Arenzana‐seisdedos F., Amara A. ( 2005) Dendritic cell‐specific intercellular adhesion molecule 3‐grabbing non‐integrin (DCSIGN)‐mediated enhancement of dengue virus infection is independent of DC‐SIGN internalization signals. J Biol Chem 280: 23698–708.
    1. Miller J.L., Dewet B.J., Martinez‐pomares L., Radcliffe C.M., Dwek R.A., Rudd P.M., Gordon S. ( 2008) The mannose receptor mediates dengue virus infection of macrophages. PLoS Pathog 4: 1–11.
    1. Marianneau P., Steffan A.M., Royer C., Drouet M.T., Kirn A., Deubel V. ( 1998) Differing infection patterns of dengue and yellow fever viruses in a human hepatoma cell line. J Infect Dis 178: 1270–8.
    1. de Duve C. ( 1983) Lysosomes revisited. Eur J Biochem 137: 391–7.
    1. Ruiz A.C.G., Nascimento R.T., de Paula S.O., da Fonseca B.A.L. ( 2006) SYBR green and TaqMan real‐time PCR assays are equivalent for the diagnosis of dengue virus type 3 infections. J Med Virol 78: 760–3.
    1. Houng H.S.H., Chen R.C., Vaughn D.W., Kanesathasan N. ( 2001) Development of a fluorogenic RT‐PCR system for quantitative identification of dengue virus serotypes 1‐4 using conserved and serotype‐specific 3(noncoding sequences. J Virol Methods 95: 19–32.
    1. Savarino A., Boelaert J.R., Cassone A., Majori G., Cauda R. ( 2003) Effects of chloroquine on viral infections: An old drug against today's diseases?. Lancet Infect Dis 3: 722–7.
    1. Bruce‐chwatt L.J. ed. ( 1981) Chemotherapy of Malaria. 2nd edn. WHO Monograph Series 27. Geneva, Switzerland: World Health Organization.
    1. Wellems T.E., Plowe C.V. ( 2001) Chloroquine‐resistant malaria. J Infect Dis 184: 770–6.
    1. Rolain J.M., Colson P., Raoult D. ( 2007) Recycling of chloroquine and its hydroxyl analogue to face bacterial, fungal and viral infections in the 21st century. Int J Antimicrob Agents 30: 297–308.
    1. Farias K.J.S., Machado P.R.L., Fonseca B.A.L. ( 2013) Chloroquine inhibits dengue virus type 2 replication in Vero cells but not in C6/36 cells. Sci World J 2013: 1–5.
    1. Civitico G., Wang Y.Y., Luscombe C., Bishop N., Tachedjian G., Gust I., Locarnini S. ( 1990) Antiviral strategies in chronic hepatitis B virus infection: II. Inhibition of duck hepatitis B virus in vitro using conventional antiviral agents and supercoiled‐DNA active compounds. J Med Virol 31: 90–7.
    1. Offensperger W.B., Offensperger S., Walter E., Blum H.E., Gerok W. ( 1991) Inhibition of duck hepatitis B virus infection by lysosomotropic agents. Virology 183: 415–8.
    1. Koyama A.H., Uchida T. ( 1984) Inhibition of multiplication of herpes simplex virus type 1 by ammonium chloride and chloroquine. Virology 138: 332–5.
    1. Ooi E.E., Chew J.S., Loh J.P., Chua R.C. ( 2006) In vitro inhibition of human influenza A virus replication by chloroquine. Virol J 3: 39.
    1. Sperber K., Kalb T.H., Stecher V.J., Banerjee R., Mayer L. ( 1993) Inhibition of human immunodeficiency virus type 1 replication by hydroxychloroquine in T cells and monocytes. AIDS Res Hum Retroviruses 9: 91–8.
    1. Pardridge W.M., Yang J., Diagne A. ( 1998) Chloroquine inhibits HIV‐1 replication in human peripheral blood lymphocytes. Immunol Lett 64: 45–47.
    1. Savarino A., Lucia M.B., Rastrelli E., Rutella S., Golotta C., Morra E., Tamburrini E., Perno C.F., Boelaert J.R., Sperber K., Cauda R. ( 2004) Anti‐HIV effects of chloroquine: Inhibition of viral particle glycosylation and synergism with protease inhibitors. J Acquir Immune Defic Syndr 35: 223–32.
    1. Keyaerts E., Vijgen L., Maes P., Neyts J., Ranst M.V. ( 2004) In vitro inhibition of severe acute respiratory syndrome coronavirus by chloroquine. Biochem Biophys Res Commun 323: 264–8.
    1. Vincent M.J., Bergeron E., Benjannet S., Erickson B.R., Rollin P.E., Ksiazek T.G., Seidah N.G., Nichol S.T. ( 2005) Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol J 2: 1–10.
    1. de Clercq E. ( 2006) Potential antivirals and antiviral strategies against SARS coronavirus infections. Expert Rev Anti Infect Ther 4: 291–302.
    1. Miller N., Hutt‐fletcher L.M. ( 1992) Epstein‐Barr virus enters B cells and epithelial cells by different routes. J Virol 66: 3409–14.
    1. Gandini M., Gras C., Azeredo E.L., Pinto L.M., Smith N., Despres P., da Cunha R.V., de Souza L.J., Kubelka C.F., Herbeuval J.P. ( 2013) Dengue virus activates membrane TRAIL relocalization and IFN‐α production by human plasmacytoid dendritic cells in vitro and in vivo. PLoS Negl Trop Dis 7: e2257.
    1. Borges M.C., Castro L.A., da Fonseca B.A.L. ( 2013) Chloroquine use improves dengue‐related symptoms. Mem Inst Oswaldo Cruz. Mem Inst Oswaldo Cruz, Rio de Janeiro 108 (5): 596–9.
    1. Tricou V., Minh N.N., Van T.P., Lee S.J., Farrar J., Wills B., Tran H.T., Simmons C.P. ( 2010) A randomized controlled trial of chloroquine for the treatment of dengue in Vietnamese adults. PLoS Negl Trop Dis 4: e785.
    1. Van den borne B.E., Dijkmans B.A., de Rooij H.H., le Cessie S., Verweij C.L. ( 1997) Chloroquine and hydroxychloroquine equally affect tumor necrosis factor‐alpha, interleukin 6, and interferon‐gamma production by peripheral blood mononuclear cells. J Rheumatol 24: 55–60.
    1. Weber S.M., Levitz S.M. ( 2000) Chloroquine interferes with lipopolysaccharide‐induced TNF‐alpha gene expression by a nonlysosomotropic mechanism. J Immunol 165: 1534–40.
    1. Weber S.M., Chen J.M., Levitz S.M. ( 2002) Inhibition of mitogen‐activated protein kinase signaling by chloroquine. J Immunol 168: 5303–9.
    1. Jang C.H., Choi J.H., Byun M.S., Jue D.M.( 2006) Chloroquine inhibits production of TNF‐alpha, IL‐1beta and IL‐6 from lipopolysaccharide‐stimulated human monocytes/ macrophages by different modes. Rheumatology (Oxford) 45: 703–10.
    1. Jeong J.Y., Choi J.W., Jeon K.I., Jue D.M. ( 2002) Chloroquine decreases cell‐ surface expression of tumour necrosis factor receptors in human histiocytic U‐937 cells. Immunology 105: 83–91.
    1. Silva M.C., Guerrero‐Plata A., Gilfoy F.D., Garofalo R.P., Mason P.W. ( 2007) Differential activation of human monocyte‐derived and plasmacytoid dendritic cells by West Nile virus generated in different host cells. J Virol 81: 13640–8.
    1. Jeong J.Y., Jue D.M. ( 1997) Chloroquine inhibits processing of tumor necrosis factor in lipopolysaccharide‐stimulated RAW 264. 7 macrophages. J Immunol 158: 4901–7.

Source: PubMed

3
Předplatit