Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Zika viral dynamics and shedding in rhesus and cynomolgus macaques

A Corrigendum to this article was published on 07 February 2017

This article has been updated

Abstract

Infection with Zika virus has been associated with serious neurological complications and fetal abnormalities. However, the dynamics of viral infection, replication and shedding are poorly understood. Here we show that both rhesus and cynomolgus macaques are highly susceptible to infection by lineages of Zika virus that are closely related to, or are currently circulating in, the Americas. After subcutaneous viral inoculation, viral RNA was detected in blood plasma as early as 1 d after infection. Viral RNA was also detected in saliva, urine, cerebrospinal fluid (CSF) and semen, but transiently in vaginal secretions. Although viral RNA during primary infection was cleared from blood plasma and urine within 10 d, viral RNA was detectable in saliva and seminal fluids until the end of the study, 3 weeks after the resolution of viremia in the blood. The control of primary Zika virus infection in the blood was correlated with rapid innate and adaptive immune responses. We also identified Zika RNA in tissues, including the brain and male and female reproductive tissues, during early and late stages of infection. Re-infection of six animals 45 d after primary infection with a heterologous strain resulted in complete protection, which suggests that primary Zika virus infection elicits protective immunity. Early invasion of Zika virus into the nervous system of healthy animals and the extent and duration of shedding in saliva and semen underscore possible concern for additional neurologic complications and nonarthropod-mediated transmission in humans.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Blood plasma ZIKV RNA kinetics during primary infection.
Figure 2: Timing and magnitude of ZIKV shedding in CSF and mucosal fluids during primary infection.
Figure 3: Infection of rhesus macaques with a Puerto Rican ZIKV isolate.
Figure 4: Detection of ZIKV RNA in lymphoid, neurologic and reproductive tissues.
Figure 5: Protection against heterologous challenge with a Puerto Rican ZIKV isolate.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

Change history

  • 19 October 2016

    In the version of this article initially published online, Robert Were Omange's name was misspelled in the author list. The original version listed Robert Omage. The error has been corrected in the print, PDF and HTML versions of this article.

References

  1. Dick, G.W., Kitchen, S.F. & Haddow, A.J. Zika virus. I. Isolations and serological specificity. Trans. R. Soc. Trop. Med. Hyg. 46, 509–520 (1952).

    CAS  PubMed  Google Scholar 

  2. Brasil, P. et al. Zika virus outbreak in Rio de Janeiro, Brazil: clinical characterization, epidemiological and virological Aspects. PLoS Negl. Trop. Dis. 10, e0004636 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Cerbino-Neto, J. et al. Clinical manifestations of Zika virus infection, Rio de Janeiro, Brazil, 2015. Emerg. Infect. Dis. 22, 1318–1320 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Duffy, M.R. et al. Zika virus outbreak on Yap Island, Federated States of Micronesia. N. Engl. J. Med. 360, 2536–2543 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Campos, G.S., Bandeira, A.C. & Sardi, S.I. Zika virus outbreak, Bahia, Brazil. Emerg. Infect. Dis. 21, 1885–1886 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Cao-Lormeau, V.M. & Musso, D. Emerging arboviruses in the Pacific. Lancet 384, 1571–1572 (2014).

    Article  PubMed  Google Scholar 

  7. Cardoso, C.W. et al. Outbreak of exanthematous illness associated with Zika, chikungunya, and dengue viruses, Salvador, Brazil. Emerg. Infect. Dis. 21, 2274–2276 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Oehler, E. et al. Zika virus infection complicated by Guillain-Barre syndrome—case report, French Polynesia, December 2013. Euro Surveill. 19, 20720 (2014).

    Article  PubMed  Google Scholar 

  9. Millon, P. Epidémiologie des syndromes de Guillain-Barré en Nouvelle-Calédonie entre 2011 et 2014: influence des arboviroses, U.J. Fourier, Editor. 2015.

  10. Cao-Lormeau, V.M. et al. Guillain-Barré Syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study. Lancet 387, 1531–1539 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Brasil, P. et al. Guillain-Barré syndrome associated with Zika virus infection. Lancet 387, 1482 (2016).

    Article  PubMed  Google Scholar 

  12. Schuler-Faccini, L. et al. Possible association between Zika virus infection and microcephaly—Brazil, 2015. MMWR Morb. Mortal. Wkly. Rep. 65, 59–62 (2016).

    Article  PubMed  Google Scholar 

  13. European Centre for Disease Prevention and Control. Microcephaly in Brazil potentially linked to the Zika virus epidemic 2015: Stockholm.

  14. Mlakar, J. et al. Zika virus associated with microcephaly. N. Engl. J. Med. 374, 951–958 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Calvet, G. et al. Detection and sequencing of Zika virus from amniotic fluid of fetuses with microcephaly in Brazil: a case study. Lancet Infect. Dis. 16, 653–660 (2016).

    Article  PubMed  Google Scholar 

  16. Peterson, A.T., Osorio, J., Qiao, H. & Escobar, L.E. Zika virus, elevation, and transmission risk. PLoS Curr. http://currents.plos.org/outbreaks/article/zika-virus-elevation-and-transmission-risk/ (2016).

  17. Foy, B.D. et al. Probable non-vector-borne transmission of Zika virus, Colorado, USA. Emerg. Infect. Dis. 17, 880–882 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Musso, D. et al. Potential sexual transmission of Zika virus. Emerg. Infect. Dis. 21, 359–361 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bearcroft, W.G. Zika virus infection experimentally induced in a human volunteer. Trans. R. Soc. Trop. Med. Hyg. 50, 442–448 (1956).

    Article  CAS  PubMed  Google Scholar 

  20. Bingham, A.M. et al. Comparison of test results for Zika virus RNA in urine, serum, and saliva specimens from persons with travel-associated Zika virus disease—Florida, 2016. MMWR Morb. Mortal. Wkly. Rep. 65, 475–478 (2016).

    Article  PubMed  Google Scholar 

  21. Musso, D. et al. Detection of Zika virus in saliva. J. Clin. Virol. 68, 53–55 (2015).

    Article  PubMed  Google Scholar 

  22. Gourinat, A.C., O'Connor, O., Calvez, E., Goarant, C. & Dupont-Rouzeyrol, M. Detection of Zika virus in urine. Emerg. Infect. Dis. 21, 84–86 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hamel, R. et al. Biology of Zika virus infection in human skin cells. J. Virol. 89, 8880–8896 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Tabata, T. et al. Zika virus targets different primary human placental cells, suggesting two routes for vertical transmission. Cell Host Microbe 20, 155–166 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dudley, D.M. et al. A rhesus macaque model of Asian-lineage Zika virus infection. Nat. Commun. 7, 12204 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Styer, L.M. et al. Mosquitoes inoculate high doses of West Nile virus as they probe and feed on live hosts. PLoS Pathog. 3, 1262–1270 (2007).

    Article  CAS  PubMed  Google Scholar 

  27. Gubler, D.J. & Rosen, L. A simple technique for demonstrating transmission of dengue virus by mosquitoes without the use of vertebrate hosts. Am. J. Trop. Med. Hyg. 25, 146–150 (1976).

    Article  CAS  PubMed  Google Scholar 

  28. Dutra, H.L. et al. Wolbachia blocks currently circulating Zika virus isolates in Brazilian Aedes aegypti mosquitoes. Cell Host Microbe 19, 771–774 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Rathakrishnan, A. et al. Cytokine expression profile of dengue patients at different phases of illness. PLoS One 7, e52215 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Pandey, N. et al. Serum levels of IL-8, IFNγ, IL-10, and TGF β and their gene expression levels in severe and non-severe cases of dengue virus infection. Arch. Virol. 160, 1463–1475 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Kumar, Y. et al. Serum proteome and cytokine analysis in a longitudinal cohort of adults with primary dengue infection reveals predictive markers of DHF. PLoS Negl. Trop. Dis. 6, e1887 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Whitney, J.B. et al. Monitoring HIV vaccine trial participants for primary infection: studies in the SIV/macaque model. AIDS 23, 1453–1460 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Bai, F. et al. A paradoxical role for neutrophils in the pathogenesis of West Nile virus. J. Infect. Dis. 202, 1804–1812 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Soares, C.N. et al. Fatal encephalitis associated with Zika virus infection in an adult. J. Clin. Virol. 83, 63–65 (2016).

    Article  PubMed  Google Scholar 

  35. Ventura, C.V., Maia, M., Bravo-Filho, V., Góis, A.L. & Belfort, R. Jr. Zika virus in Brazil and macular atrophy in a child with microcephaly. Lancet 387, 228 (2016).

    Article  PubMed  Google Scholar 

  36. Mlakar, J. et al. Zika virus associated with microcephaly. N. Engl. J. Med. 374, 951–958 (2016).

    Article  CAS  PubMed  Google Scholar 

  37. Barcellos, C. et al. Increased hospitalizations for neuropathies as indicators of Zika virus infection, according to Health Information System Data, Brazil. Emerg. Infect. Dis. http://dx.doi.org/10.3201/eid2211.160901 (2016).

  38. Mécharles, S. et al. Acute myelitis due to Zika virus infection. Lancet 387, 1481 (2016).

    Article  PubMed  Google Scholar 

  39. Turmel, J.M. et al. Late sexual transmission of Zika virus related to persistence in the semen. Lancet 387, 2501 (2016).

    Article  PubMed  Google Scholar 

  40. D'Ortenzio, E. et al. Evidence of sexual transmission of Zika virus. N. Engl. J. Med. 374, 2195–2198 (2016).

    Article  PubMed  Google Scholar 

  41. Deckard, D.T. et al. Male-to-male sexual transmission of Zika virus—Texas, January 2016. MMWR Morb. Mortal. Wkly. Rep. 65, 372–374 (2016).

    Article  PubMed  Google Scholar 

  42. Prisant, N. et al. Zika virus in the female genital tract. Lancet Infect. Dis. http://dx.doi.org/10.1016/S1473-3099(16)30193-1 (2016).

  43. Roederer, M., Nozzi, J.L. & Nason, M.C. SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A 79, 167–174 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Harrison, R.M. Semen parameters in Macaca mulatta: ejaculates from random and selected monkeys. J. Med. Primatol. 9, 265–273 (1980).

    Article  CAS  PubMed  Google Scholar 

  45. Deleage, C. et al. Defining HIV and SIV reservoirs in lymphoid tissues. Pathog. Immun. 1, 68–106 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Kuhrt, D. et al. Naïve and memory B cells in the rhesus macaque can be differentiated by surface expression of CD27 and have differential responses to CD40 ligation. J. Immunol. Methods 363, 166–176 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Webster, R.L. & Johnson, R.P. Delineation of multiple subpopulations of natural killer cells in rhesus macaques. Immunology 115, 206–214 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Reeves, R.K. et al. CD16 natural killer cells: enrichment in mucosal and secondary lymphoid tissues and altered function during chronic SIV infection. Blood 115, 4439–4446 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pitcher, C.J. et al. Development and homeostasis of T cell memory in rhesus macaque. J. Immunol. 168, 29–43 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Donaldson, M.M. et al. Optimization and qualification of an 8-color intracellular cytokine staining assay for quantifying T cell responses in rhesus macaques for pre-clinical vaccine studies. J. Immunol. Methods 386, 10–21 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Baccam, P., Beauchemin, C., Macken, C.A., Hayden, F.G. & Perelson, A.S. Kinetics of influenza A virus infection in humans. J. Virol. 80, 7590–7599 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Banerjee, S., Guedj, J., Ribeiro, R.M., Moses, M. & Perelson, A.S. Estimating biologically relevant parameters under uncertainty for experimental within-host murine West Nile virus infection. J. R. Soc. Interface 13, 20160130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Fukuhara, M. et al. Quantification of the dynamics of enterovirus 71 infection by experimental-mathematical investigation. J. Virol. 87, 701–705 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Guedj, J. et al. Modeling shows that the NS5A inhibitor daclatasvir has two modes of action and yields a shorter estimate of the hepatitis C virus half-life. Proc. Natl. Acad. Sci. USA 110, 3991–3996 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Ramratnam, B. et al. Rapid production and clearance of HIV-1 and hepatitis C virus assessed by large volume plasma apheresis. Lancet 354, 1782–1785 (1999).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank B. Finneyfrock, Z. Pippin, A. Dodson and A. Cook for their expert animal husbandry and care, and J. Guedj for suggestions about the model simulations. CD38 antibodies were obtained from the NIH Nonhuman Primate Reagent Resource supported by HHSN272200900037C and OD010976. The data presented in this study are tabulated in the main paper and in the supplementary materials. This work was supported in part by federal funds from the National Cancer Institute (NIH Contract HHSN261200800001E). The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does the mention of trade names, commercial products or organizations imply endorsement by the US Government. A.S.P. acknowledges support from NIH grants AI028433 and OD0110995. D.S. acknowledges support from the Public Health Agency of Canada.

Author information

Authors and Affiliations

Authors

Contributions

J.B.W. designed the studies. C.E.O., H.A.-E. and S.-Y.L. led the virologic assays. S.-Y.L. and C.E.O. led the immunologic assays. C.D. and J.D.E. led all tissue analysis. L.T.S., R.W.O. and M.L. conducted the cytokine assays. P.T.H, M.L., C.E.O. and S.-Y.L. led the cytokine and chemokine analysis. S.-Y.L. and P.T.H. led the peptide design analysis. B.D.G., D. Stein and D. Safronetz led the antibody assays. D.L.V., S. Huang, S. Higgs and D. Safronetz produced the ZIKV stocks K.B., E.F.C.O. and A.S.P. led the mathematical modeling. H.A.-E. and M.G.L. led the clinical care of the macaques. J.B.W. led the studies and wrote the paper with all co-authors.

Corresponding author

Correspondence to James B Whitney.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–2 and Supplementary Figures 1–10 (PDF 4077 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Osuna, C., Lim, SY., Deleage, C. et al. Zika viral dynamics and shedding in rhesus and cynomolgus macaques. Nat Med 22, 1448–1455 (2016). https://doi.org/10.1038/nm.4206

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4206

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing