1887

Abstract

The fidelity of flaviviruses is thought to be tightly regulated for optimal fitness within and between hosts. West Nile virus (WNV) high-fidelity (HiFi) mutations V793I and G806R within the RNA-dependent RNA polymerase, and low-fidelity (LoFi) mutation T248I within the methyltransferase, were previously shown to attenuate infectivity and replicative fitness in mosquitoes and (CXT) cells but not in mammalian cells. We hypothesized that fidelity alterations would modify adaptation and maintenance in a host-specific manner. To test this hypothesis, wild-type (WT), HiFi (V793I/G806R) and LoFi (T248I) variants were sequentially passaged eight times in avian (PDE) or mosquito cells, or alternately between the two. Initial characterization confirmed that fidelity mutants are attenuated in mosquito, but not avian, cells. Deep sequencing revealed mutations unique to both cell lines and fidelity mutants, including ENV G1378A, a mutation associated with avian cell adaptation. To characterize maintenance and adaptation, viral outputs were monitored throughout passaging and viral fitness was assessed. The results indicate that fidelity mutants can at times recover fitness during mosquito cell passage, but remain attenuated relative to WT. Despite similar initial fitness, LoFi mutants were impaired during sequential passage in avian cells. Conversely, HiFi mutants passaged in avian cells showed increased adaptation, suggesting that increased fidelity may be advantageous in avian hosts. Although some adaptation occurred with individual mutants, the output titres of fidelity mutants were on average lower and were often lost during host switching. These data confirm that arbovirus fidelity is likely fine-tuned to maximize survival in disparate hosts.

Funding
This study was supported by the:
  • National Institutes of Health (Award AI146856)
    • Principle Award Recipient: Alexander T. Ciota
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001393
2020-02-18
2024-04-24
Loading full text...

Full text loading...

/deliver/fulltext/jgv/101/4/410.html?itemId=/content/journal/jgv/10.1099/jgv.0.001393&mimeType=html&fmt=ahah

References

  1. Bäck AT, Lundkvist A. Dengue viruses - an overview. Infect Ecol Epidemiol 2013; 3:
    [Google Scholar]
  2. McMullen AR, May FJ, Li L, Guzman H, Bueno R et al. Evolution of new genotype of West Nile virus in North America. Emerg Infect Dis 2011; 17:785–793 [View Article][PubMed]
    [Google Scholar]
  3. Ciota A, Kramer L. Vector-virus interactions and transmission dynamics of West Nile virus. Viruses 2013; 5:3021–3047 [View Article]
    [Google Scholar]
  4. Hayes EB, Gubler DJ. West Nile virus: epidemiology and clinical features of an emerging epidemic in the United States. Annu Rev Med 2006; 57:181–194 [View Article]
    [Google Scholar]
  5. Musso D, Gubler DJ, Virus Z. Zika virus. Clin Microbiol Rev 2016; 29:487–524 [View Article]
    [Google Scholar]
  6. Kilpatrick AM. Globalization, land use, and the invasion of West Nile virus. Science 2011; 334:323–327 [View Article][PubMed]
    [Google Scholar]
  7. Dengue GDJ. Urbanization and Globalization: The Unholy Trinity of the 21(st) Century. Trop Med Health 2011; 39:3–11
    [Google Scholar]
  8. Baharuddin A, Hassan A, Sheng G, Nasir S, Othman S et al. Current approaches in antiviral drug discovery against the Flaviviridae family. Curr Pharm Des 2014; 20:3428–3444 [View Article]
    [Google Scholar]
  9. Añez G, Grinev A, Chancey C, Ball C, Akolkar N et al. Evolutionary dynamics of West Nile virus in the United States, 1999–2011: phylogeny, selection pressure and evolutionary time-scale analysis. PLoS Negl Trop Dis 2013; 7:e2245 [View Article]
    [Google Scholar]
  10. Kramer LD, Styer LM, Ebel GD. A global perspective on the epidemiology of West Nile virus. Annu Rev Entomol 2008; 53:61–81 [View Article]
    [Google Scholar]
  11. Klema V, Padmanabhan R, Choi K. Flaviviral Replication Complex: Coordination between RNA Synthesis and 5’-RNA Capping. Viruses 2015; 7:4640–4656 [View Article]
    [Google Scholar]
  12. Lauring AS, Andino R. Quasispecies theory and the behavior of RNA viruses. PLoS Pathog 2010; 6:e1001005 [View Article]
    [Google Scholar]
  13. Ciota AT, Ehrbar DJ, Van Slyke GA, Willsey GG, Kramer LD. Cooperative interactions in the West Nile virus mutant Swarm. BMC Evol Biol 2012; 12:58 [View Article]
    [Google Scholar]
  14. Ciota AT, Ngo KA, Lovelace AO, Payne AF, Zhou Y et al. Role of the mutant spectrum in adaptation and replication of West Nile virus. Journal of General Virology 2007; 88:865–874 [View Article]
    [Google Scholar]
  15. Brackney DE, Schirtzinger EE, Harrison TD, Ebel GD, Hanley KA. Modulation of flavivirus population diversity by RNA interference. J Virol 2015; 89:4035–4039 [View Article]
    [Google Scholar]
  16. Vignuzzi M, Stone JK, Arnold JJ, Cameron CE, Andino R. Quasispecies diversity determines pathogenesis through cooperative interactions in a viral population. Nature 2006; 439:344–348 [View Article]
    [Google Scholar]
  17. Van Slyke GA, Ciota AT, Willsey GG, Jaeger J, Shi P-Y et al. Point mutations in the West Nile virus (Flaviviridae; flavivirus) RNA-dependent RNA polymerase alter viral fitness in a host-dependent manner in vitro and in vivo. Virology 2012; 427:18–24 [View Article]
    [Google Scholar]
  18. Jerzak GVS, Brown I, Shi P-Y, Kramer LD, Ebel GD. Genetic diversity and purifying selection in West Nile virus populations are maintained during host switching. Virology 2008; 374:256–260 [View Article]
    [Google Scholar]
  19. Van Slyke GA, Arnold JJ, Lugo AJ, Griesemer SB, Moustafa IM et al. Sequence-Specific fidelity alterations associated with West Nile virus attenuation in mosquitoes. PLoS Pathog 2015; 11:e1005009 [View Article]
    [Google Scholar]
  20. Griesemer SB, Kramer LD, Van Slyke GA, Pata JD, Gohara DW et al. Mutagen resistance and mutation restriction of St. Louis encephalitis virus. J Gen Virol 2017; 98:201–211 [View Article]
    [Google Scholar]
  21. Warmbrod KL, Patterson EI, Kautz TF, Stanton A, Rockx-Brouwer D et al. Viral RNA-dependent RNA polymerase mutants display an altered mutation spectrum resulting in attenuation in both mosquito and vertebrate hosts. PLoS Pathog 2019; 15:e1007610-e [View Article]
    [Google Scholar]
  22. Riemersma KK, Steiner C, Singapuri A, Coffey LL. Chikungunya virus fidelity variants exhibit differential attenuation and population diversity in cell culture and adult mice. Journal of Virology 2019; 93:e01606–01618
    [Google Scholar]
  23. Shi P-Y, Tilgner M, Lo MK. Construction and characterization of subgenomic replicons of new York strain of West Nile virus. Virology 2002; 296:219–233 [View Article][PubMed]
    [Google Scholar]
  24. Ebel GD, Dupuis AP II, Ngo KA, Nicholas DC, Kauffman EB et al. Partial genetic characterization of West Nile virus strains, New York state. EmergInfectDis 2001; 7:650–653
    [Google Scholar]
  25. Ehrbar DJ, Ngo KA, Campbell SR, Kramer LD, Ciota AT. High levels of local inter- and intra-host genetic variation of West Nile virus and evidence of fine-scale evolutionary pressures. Infection, Genetics and Evolution 2017; 51:219–226 [View Article]
    [Google Scholar]
  26. Payne AF, Binduga-Gajewska I, Kauffman EB, Kramer LD. Quantitation of flaviviruses by fluorescent focus assay. J Virol Methods 2006; 134:183–189 [View Article]
    [Google Scholar]
  27. Kanai R, Kar K, Anthony K, Gould LH, Ledizet M et al. Crystal structure of West Nile virus envelope glycoprotein reveals viral surface epitopes. J Virol 2006; 80:11000–11008 [View Article]
    [Google Scholar]
  28. Ciota AT, Jia Y, Payne AF, Jerzak G, Davis LJ et al. Experimental passage of St. Louis encephalitis virus in vivo in mosquitoes and chickens reveals evolutionarily significant virus characteristics. PLoS One 2009; 4:e7876 [View Article]
    [Google Scholar]
  29. Fitzpatrick KA, Deardorff ER, Pesko K, Brackney DE, Zhang B et al. Population variation of West Nile virus confers a host-specific fitness benefit in mosquitoes. Virology 2010; 404:89–95 [View Article]
    [Google Scholar]
  30. Göertz G, Miesen P, Overheul G, van Rij R, van Oers M et al. Mosquito small RNA responses to West Nile and insect-specific virus infections in Aedes and Culex mosquito cells. Viruses 2019; 11:271 [View Article]
    [Google Scholar]
  31. Vogel E, Santos D, Mingels L, Verdonckt T-W, Broeck JV. RNA interference in insects: protecting Beneficials and controlling pests. Front Physiol 2019; 9:1912 [View Article]
    [Google Scholar]
  32. Brackney DE, Beane JE, Ebel GD. Rnai targeting of West Nile virus in mosquito midguts promotes virus diversification. PLoS Pathog 2009; 5:e1000502 [View Article]
    [Google Scholar]
  33. Brackney DE, Pesko KN, Brown IK, Deardorff ER, Kawatachi J et al. West Nile virus genetic diversity is maintained during transmission by Culex pipiens quinquefasciatus mosquitoes. PLoS One 2011; 6:e24466 [View Article]
    [Google Scholar]
  34. Eigen M. Selforganization of matter and the evolution of biological macromolecules. Naturwissenschaften 1971; 58:465–523 [View Article]
    [Google Scholar]
  35. Loeb LA, Essigmann JM, Kazazi F, Zhang J, Rose KD et al. Lethal mutagenesis of HIV with mutagenic nucleoside analogs. Proc Natl Acad Sci U S A 1999; 96:1492–1497 [View Article]
    [Google Scholar]
  36. Anderson JP, Daifuku R, Loeb LA. Viral error catastrophe by mutagenic nucleosides. Annu Rev Microbiol 2004; 58:183–205 [View Article]
    [Google Scholar]
  37. Bull JJ, Sanjuan R, Wilke CO. Theory of lethal mutagenesis for viruses. J Virol 2007; 81:2930–2939 [View Article]
    [Google Scholar]
  38. Jerzak G, Bernard KA, Kramer LD, Ebel GD. Genetic variation in West Nile virus from naturally infected mosquitoes and birds suggests quasispecies structure and strong purifying selection. J GenVirol 2005; 86:2175–2183 [View Article]
    [Google Scholar]
  39. Jerzak GVS, Bernard K, Kramer LD, Shi P-Y, Ebel GD. The West Nile virus mutant spectrum is host-dependant and a determinant of mortality in mice. Virology 2007; 360:469–476 [View Article]
    [Google Scholar]
  40. Nelson CW, Sibley SD, Kolokotronis S-O, Hamer GL, Newman CM et al. Selective constraint and adaptive potential of West Nile virus within and among naturally infected avian hosts and mosquito vectors. Virus Evolution 2018; 4:vey013 [View Article]
    [Google Scholar]
  41. Deardorff ER, Fitzpatrick KA, Jerzak GVS, Shi P-Y, Kramer LD et al. West Nile virus experimental evolution in vivo and the trade-off hypothesis. PLoS Pathog 2011; 7:e1002335 [View Article]
    [Google Scholar]
  42. Novella IS, Hershey CL, Escarmis C, Domingo E, Holland JJ. Lack of evolutionary stasis during alternating replication of an arbovirus in insect and mammalian cells. J Mol Biol 1999; 287:459–465 [View Article]
    [Google Scholar]
  43. Escarmís C, Lázaro E, Arias A, Domingo E. Repeated bottleneck transfers can lead to Non-cytocidal forms of a cytopathic virus: implications for viral extinction. J Mol Biol 2008; 376:367–379 [View Article]
    [Google Scholar]
  44. Novella IS, Ebendick-Corpus BE. Molecular basis of fitness loss and fitness recovery in vesicular stomatitis virus. J Mol Biol 2004; 342:1423–1430 [View Article]
    [Google Scholar]
  45. Weaver SC, Brault AC, Kang W, Holland JJ. Genetic and fitness changes accompanying adaptation of an arbovirus to vertebrate and invertebrate cells. J Virol 1999; 73:4316–4326 [View Article]
    [Google Scholar]
  46. Novella IS, Presloid JB, Smith SD, Wilke CO. Specific and nonspecific host adaptation during arboviral experimental evolution. J Mol Microbiol Biotechnol 2011; 21:71–81 [View Article]
    [Google Scholar]
  47. Grubaugh ND, Smith DR, Brackney DE, Bosco-Lauth AM, Fauver JR et al. Experimental evolution of an RNA virus in wild birds: evidence for host-dependent impacts on population structure and competitive fitness. PLoS Pathog 2015; 11:e1004874 [View Article]
    [Google Scholar]
  48. Kimura M. Diffusion models in population genetics. J Appl Probab 1964; 1:177–232 [View Article]
    [Google Scholar]
  49. Selisko B, Papageorgiou N, Ferron F, Canard B. Structural and functional basis of the fidelity of nucleotide selection by flavivirus RNA-dependent RNA polymerases. Viruses 2018; 10:59 [View Article]
    [Google Scholar]
  50. Zeng J, Wang H, Xie X, Li C, Zhou G et al. Ribavirin-resistant variants of foot-and-mouth disease virus: the effect of restricted quasispecies diversity on viral virulence. J Virol 2014; 88:4008–4020 [View Article]
    [Google Scholar]
  51. Kaufmann B, Vogt MR, Goudsmit J, Holdaway HA, Aksyuk AA et al. Neutralization of West Nile virus by cross-linking of its surface proteins with Fab fragments of the human monoclonal antibody CR4354. Proc Natl Acad Sci U S A 2010; 107:18950–18955 [View Article]
    [Google Scholar]
  52. Modis Y, Ogata S, Clements D, Harrison SC. A ligand-binding pocket in the dengue virus envelope glycoprotein. Proc Natl Acad Sci U S A 2003; 100:6986–6991 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001393
Loading
/content/journal/jgv/10.1099/jgv.0.001393
Loading

Data & Media loading...

Supplements

Supplementary material 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