Hantavirus Infection Is Inhibited by Griffithsin in Cell Culture

Punya Shrivastava-Ranjan, Michael K Lo, Payel Chatterjee, Mike Flint, Stuart T Nichol, Joel M Montgomery, Barry R O'Keefe, Christina F Spiropoulou, Punya Shrivastava-Ranjan, Michael K Lo, Payel Chatterjee, Mike Flint, Stuart T Nichol, Joel M Montgomery, Barry R O'Keefe, Christina F Spiropoulou

Abstract

Andes virus (ANDV) and Sin Nombre virus (SNV), highly pathogenic hantaviruses, cause hantavirus pulmonary syndrome in the Americas. Currently no therapeutics are approved for use against these infections. Griffithsin (GRFT) is a high-mannose oligosaccharide-binding lectin currently being evaluated in phase I clinical trials as a topical microbicide for the prevention of human immunodeficiency virus (HIV-1) infection (ClinicalTrials.gov Identifiers: NCT04032717, NCT02875119) and has shown broad-spectrum in vivo activity against other viruses, including severe acute respiratory syndrome coronavirus, hepatitis C virus, Japanese encephalitis virus, and Nipah virus. In this study, we evaluated the in vitro antiviral activity of GRFT and its synthetic trimeric tandemer 3mGRFT against ANDV and SNV. Our results demonstrate that GRFT is a potent inhibitor of ANDV infection. GRFT inhibited entry of pseudo-particles typed with ANDV envelope glycoprotein into host cells, suggesting that it inhibits viral envelope protein function during entry. 3mGRFT is more potent than GRFT against ANDV and SNV infection. Our results warrant the testing of GRFT and 3mGRFT against ANDV infection in animal models.

Keywords: antiviral; griffithsin; haemorrhagic fever; hantaviridae; hantavirus.

Copyright © 2020 Shrivastava-Ranjan, Lo, Chatterjee, Flint, Nichol, Montgomery, O'Keefe and Spiropoulou.

Figures

Figure 1
Figure 1
Griffithsin inhibits Andes virus replication. Griffithsin (GRFT) inhibited Andes virus (ANDV) replication in a concentration-dependent manner. (A) Vero-E6 cells were treated for 1 h with varying concentrations of GRFT before infection with ANDV at a multiplicity of infection (MOI) of 0.1. At 72 h post infection, the cells were fixed, permeabilized, and stained with an antibody against Puumala virus nucleoprotein that is cross-reactive with ANDV nucleoprotein. Green, ANDV nucleoprotein; blue, cell nuclei; red, cell cytoplasm. (B,C) Dose-response curve showing the quantitation of ANDV-infected cells after GRFT (B) or 3mGRFT (C) treatment (% normalized to the vehicle-only control). (D,E) Inhibition (neutralization) of ANDV by GRFT. Increasing concentrations of GRFT were pre-incubated with 2,000 TCID50 of ANDV at 37°C for 1 h, and virus-GRFT (D) or virus-3mGRFT (E) mixtures were added to Vero-E6 cells. After 2 h of incubation at 37°C, inoculum was removed, and cells were replenished with fresh medium. After 3 days, cells were fixed, imaged, and analyzed as in (B). (F,G) GRFT inhibited titers of infectious ANDV. 2000 TCID50 of ANDV were pre-incubated with varying concentration of GRFT (F) or 3mGRFT (G) as in E. At 72 h, supernatants were harvested, and viral yield was determined by TCID50 assays on Vero-E6 cells. Graphs represent the mean ± SD and are representative of three independent experiments, performed in quadruplicate.
Figure 2
Figure 2
GRFT and 3mGRFT minimally affect post viral entry steps. Vero-E6 cells were infected with ANDV at MOI of 0.2 for 2 h at 37°C. Cells were then treated with varying concentrations of GRFT (A) or 3mGRFT (B). At 72 h post infection, cells were fixed, stained, and analyzed as in Figure 1B. Dose-response curve shows quantitation of ANDV-infected cells treated with GRFT (A) or 3mGRFT (B) (% normalized to vehicle-only control). Graphs represent the mean ± SD and are representative of three independent experiments, performed in quadruplicate.
Figure 3
Figure 3
GRFT and 3mGRFT affect viral entry. HIV particles bearing glycoproteins of ANDV or vesicular stomatitis virus (VSV) were prepared and used as previously described. Viral glycoprotein-dependent entry assays were performed using HT1080 cells pre-treated with serial dilutions of GRFT and inoculated with either ANDV pseudo-particles (pp) or control VSVpp in the presence of GRFT (A) or 3mGRFT (B) at increasing concentrations. Intracellular firefly luciferase signal was measured 72 h post transduction and normalized to signal in untreated, untransduced cells. Graphs represent the mean ± SD and are representative of three independent experiments, performed in quadruplicate. (C) Vero-E6 cells were treated for 1 h with either 1 or 5 μg/mL of GRFT or 3mGRFT before infection with ANDV at MOI of either 1or 3. At 24 h post infection, the cells were fixed, permeabilized, and stained with an antibody against Puumala virus nucleoprotein as described in Figure 1. Column chart showing the quantitation of ANDV-infected cells treated with GRFT or 3mGRFT (% normalized to vehicle-only controls). Graphs represent the mean ± SD and are representative of 3 independent experiments, performed in quadruplicate. P* < 0.001, P*** < 0.0001.
Figure 4
Figure 4
Antiviral effects of GRFT and 3mGRFT on Sin Nombre virus replication. Vero-E6 cells were treated for 1 h with either 1 or 5 μg/mL of GRFT or 3mGRFT before infection with Sin Nombre virus (SNV) at MOI of 0.2. At 72 h post infection, the cells were fixed, permeabilized, and stained with an antibody against Puumala virus nucleoprotein that cross-reacts with SNV nucleoprotein. Dose-response curve showing the quantitation of SNV-infected cells treated with GRFT or 3mGRFT (% normalized to vehicle-only controls). Graphs represent the mean ± SD and are representative of three independent experiments, performed in quadruplicate. P* < 0.05, P*** < 0.001.

References

    1. Alonso D. O., Perez-Sautu U., Bellomo C. M., Prieto K., Iglesias A., Coelho R., et al. . (2020). Person-to-person transmission of andes virus in hantavirus pulmonary syndrome, argentina, 2014. Emerg. Infect. Dis. 26, 756–759. 10.3201/eid2604.190799
    1. Bird B. H., Shrivastava-Ranjan P., Dodd K. A., Erickson B. R., Spiropoulou F. C. (2016). Effect of Vandetanib on Andes virus survival in the hamster model of Hantavirus pulmonary syndrome. Antiviral Res. 132, 66–69. 10.1016/j.antiviral.2016.05.014
    1. Brocato R. L., Hooper W. J. (2019). Progress on the prevention and treatment of hantavirus disease. Viruses 11:610. 10.3390/v11070610
    1. Chapman L. E., Mertz G. J., Peters C. J., Jolson H. M., Khan A. S., Ksiazek T. G., et al. . (1999). Intravenous ribavirin for hantavirus pulmonary syndrome: safety and tolerance during 1 year of open-label experience. Ribavirin study group. Antivir. Ther. 4, 211–219
    1. Emau P., Tian B., O'Keefe B. R., Mori T., McMahon J. B., Palmer K. E., et al. . (2007). Griffithsin, a potent HIV entry inhibitor, is an excellent candidate for anti-HIV microbicide. J. Med. Primatol. 36, 244–253. 10.1111/j.1600-0684.2007.00242.x
    1. Ethier M., Krokhin O., Ens W., Standing K. G., Wilkins J. A., Perreault H. (2005). Global and site-specific detection of human integrin alpha 5 beta 1 glycosylation using tandem mass spectrometry and the StrOligo algorithm. Rapid Commun. Mass Spectrom. 19, 721–727. 10.1002/rcm.1844
    1. Hepojoki J., Strandin T., Vaheri A., Lankinen H. (2010). Interactions and oligomerization of hantavirus glycoproteins. J. Virol. 84, 227–242. 10.1128/JVI.00481-09
    1. Hooper J. W., Larsen T., Custer D. M., Schmaljohn S. C. (2001). A lethal disease model for hantavirus pulmonary syndrome. Virology 289, 6–14. 10.1006/viro.2001.1133
    1. Huiskonen J. T., Hepojoki J., Laurinmaki P., Vaheri A., Lankinen H., Butcher S. J., et al. . (2010). Electron cryotomography of Tula hantavirus suggests a unique assembly paradigm for enveloped viruses. J. Virol. 84, 4889–4897. 10.1128/JVI.00057-10
    1. Ishag H. Z., Li C., Huang L., Sun M. X., Wang F., Ni B., et al. . (2013). Griffithsin inhibits Japanese encephalitis virus infection in vitro in vitro and in vivoin vivo. Arch. Virol. 158, 349–358. 10.1007/s00705-012-1489-2
    1. Ishag H. Z., Li C., Wang F., Mao X. (2016). Griffithsin binds to the glycosylated proteins (E and prM) of Japanese encephalitis virus and inhibit its infection. Virus Res. 215, 50–54. 10.1016/j.virusres.2016.01.016
    1. Jonsson C. B., Figueiredo L. T., Vapalahti O. (2010). A global perspective on hantavirus ecology, epidemiology, and disease. Clin. Microbiol. Rev. 23, 412–441. 10.1128/CMR.00062-09
    1. Jonsson C. B., Hooper J., Mertz G. (2008). Treatment of hantavirus pulmonary syndrome. Antiviral Res. 78, 162–169. 10.1016/j.antiviral.2007.10.012
    1. Lo M. K., Spengler J. R., Krumpe L. R. H., Welch S. R., Chattopadhyay A., Harmon J. R., et al. . (2020). Griffithsin inhibits nipah virus entry and fusion and can protect Syrian golden hamsters from lethal nipah virus challenge. J. Infect. Dis. 221 (Suppl. 4), S480–S492. 10.1093/infdis/jiz630
    1. Lusvarghi S., Bewley A. C. (2016). Griffithsin: an antiviral lectin with outstanding therapeutic potential. Viruses 8:296. 10.3390/v8100296
    1. Martinez V. P., Bellomo C., San Juan J., Pinna D., Forlenza R., Elder M., et al. . (2005). Person-to-person transmission of Andes virus. Emerg. Infect. Dis. 11, 1848–1853. 10.3201/eid1112.050501
    1. McNulty S., Flint M., Nichol S. T., Spiropoulou F. C. (2013). Host mTORC1 signaling regulates andes virus replication. J. Virol. 87, 912–922. 10.1128/JVI.02415-12
    1. Mertz G. J., Miedzinski L., Goade D., Pavia A. T., Hjelle B., Hansbarger C. O., et al. (2004). Placebo-controlled, double-blind trial of intravenous ribavirin for the treatment of hantavirus cardiopulmonary syndrome in North America. Clin. Infect. Dis. 39, 1307–1313. 10.1086/425007
    1. Meuleman P., Albecka A., Belouzard S., Vercauteren K., Verhoye L., Wychowski C., et al. . (2011). Griffithsin has antiviral activity against hepatitis C virus. Antimicrob. Agents Chemother. 55, 5159–5167. 10.1128/AAC.00633-11
    1. Millet J. K., Seron K., Labitt R. N., Danneels A., Palmer K. E., Whittaker G. R., et al. . (2016). Middle East respiratory syndrome coronavirus infection is inhibited by griffithsin. Antiviral Res. 133, 1–8. 10.1016/j.antiviral.2016.07.011
    1. Mittler E., Dieterle M. E., Kleinfelter L. M., Slough M. M., Chandran K., Jangra K. R. (2019). Hantavirus entry perspectives and recent advances. Adv. Virus Res. 104, 185–224. 10.1016/bs.aivir.2019.07.002
    1. Mohr E. L., McMullan L. K., Lo M. K., Spengler J. R., Bergeron E., Albarino C. G., et al. . (2013). Inhibitors of cellular kinases with broad-spectrum antiviral activity for hemorrhagic fever viruses. Antiviral Res. 120, 40–47. 10.1016/j.antiviral.2015.05.003
    1. Moulaei T., Alexandre K. B., Shenoy S. R., Meyerson J. R., Krumpe L. R., Constantine B., et al. . (2015). Griffithsin tandemers: flexible and potent lectin inhibitors of the human immunodeficiency virus. Retrovirology 12:6. 10.1186/s12977-014-0127-3
    1. O'Keefe B. R., Giomarelli B., Barnard D. L., Shenoy S. R., Chan P. K., McMahon J. B., et al. . (2010). Broad-spectrum in vitro in vitro activity and in vivoin vivo efficacy of the antiviral protein griffithsin against emerging viruses of the family Coronaviridae. J. Virol. 84, 2511–2521. 10.1128/JVI.02322-09
    1. O'Keefe B. R., Vojdani F., Buffa V., Shattock R. J., Montefiori D. C., Bakke J., et al. . (2009). Scaleable manufacture of HIV-1 entry inhibitor griffithsin and validation of its safety and efficacy as a topical microbicide component. Proc. Natl. Acad. Sci. U.S.A. 106, 6099–6104. 10.1073/pnas.0901506106
    1. Safronetz D., Falzarano D., Scott D. P., Furuta Y., Feldmann H., Gowen B. B. (2013). Antiviral efficacy of favipiravir against two prominent etiological agents of hantavirus pulmonary syndrome. Antimicrob. Agents Chemother. 57, 4673–4680. 10.1128/AAC.00886-13
    1. Schmaljohn C., Hjelle B. (1997). Hantaviruses: a global disease problem. Emerg. Infect. Dis. 3, 95–104. 10.3201/eid0302.970202
    1. Shi X., McCaughey C., Elliott M. R. (2003). Genetic characterisation of a Hantavirus isolated from a laboratory-acquired infection. J. Med. Virol. 71, 105–109. 10.1002/jmv.10446
    1. Shrivastava-Ranjan P., Bergeron E., Chakrabarti A. K., Albarino C. G., Flint M., Nichol S. T., et al. . (2016). 25-Hydroxycholesterol inhibition of Lassa virus infection through aberrant GP1 glycosylation. mBio 7:e01808–16. 10.1128/mBio.01808-16
    1. Shrivastava-Ranjan P., Flint M., Bergeron E., McElroy A. K., Chatterjee P., Albarino C. G., et al. . (2018). Statins suppress Ebola virus infectivity by interfering with glycoprotein processing. mBio 9:e00660–18. 10.1128/mBio.00660-18
    1. Shrivastava-Ranjan P., Rollin P. E., Spiropoulou F. C. (2010). Andes virus disrupts the endothelial cell barrier by induction of vascular endothelial growth factor and downregulation of VE-cadherin. J. Virol. 84, 11227–11234. 10.1128/JVI.01405-10
    1. Warner B. M., Stein D. R., Jangra R. K., Slough M. M., Sroga P., Sloan A., et al. . (2019). Vesicular stomatitis virus-based vaccines provide cross-protection against andes and sin nombre viruses. Viruses 11:645. 10.3390/v11070645
    1. Zeitlin L., Pauly M., Whaley J. K. (2009). Second-generation HIV microbicides: continued development of griffithsin. Proc. Natl. Acad. Sci. U.S A. 106, 6029–6030. 10.1073/pnas.0902239106

Source: PubMed

3
Tilaa