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.

  • Protocol Extension
  • Published:

Infection of zebrafish larvae with human norovirus and evaluation of the in vivo efficacy of small-molecule inhibitors

Abstract

We have recently established that human norovirus (HuNoV) replicates efficiently in zebrafish larvae after inoculation of a clinical sample into the yolk, providing a simple and robust in vivo system in which to study HuNoV. In this Protocol Extension, we present a detailed description of virus inoculation by microinjection, subsequent daily monitoring and harvesting of larvae, followed by viral RNA quantification. This protocol can be used to study viral replication of genogroup (G)I and GII HuNoVs in vivo within 3–4 d. Additionally, we describe how to evaluate the in vivo antiviral effect and toxicity of small molecules using HuNoV-infected zebrafish larvae, in multi-well plates and without the need for specific formulations. This constitutes a great advantage for drug discovery efforts, as no specific antivirals or vaccines currently exist to treat or prevent norovirus gastroenteritis.

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

Fig. 1: HuNoV GII.P4-GII.4 replicates in WT AB, nacre and PTU-treated zebrafish larvae.
Fig. 2: HuNoV replication in WT AB zebrafish larvae is reproducible across labs.
Fig. 3: Microinjection setup in a biosafety cabinet in a BSL-2 facility.
Fig. 4: Unfertilized and non-properly developed zebrafish embryos.
Fig. 5: Correct orientation of zebrafish larvae for yolk injection.
Fig. 6: HuNoV GII.P7-GII.6 replication at 3 dpi at different temperatures.
Fig. 7: Development of edema or postural aberrations in zebrafish larvae.
Fig. 8: HuNoV GII.P7-GII.6 replication can be detected in a single zebrafish larva.
Fig. 9: Distribution of rhodamine B after an injection in the pericardial cavity of zebrafish larvae.

Similar content being viewed by others

Data availability

The data presented in Figs. 1, 2, 4, 6, 8 and 9 were generated for this protocol and are available from the corresponding author upon reasonable request.

References

  1. Bartsch, S. M., Lopman, B. A., Ozawa, S., Hall, A. J. & Lee, B. Y. Global economic burden of norovirus gastroenteritis. PloS ONE 11, e0151219 (2016).

    Article  Google Scholar 

  2. Jones, M. K. et al. Enteric bacteria promote human and mouse norovirus infection of B cells. Science 346, 755–759 (2014).

    Article  CAS  Google Scholar 

  3. Ettayebi, K. et al. Replication of human noroviruses in stem cell-derived human enteroids. Science 353, 1387–1393 (2016).

    Article  Google Scholar 

  4. Van Dycke, J. et al. A robust human norovirus replication model in zebrafish larvae. PLoS Pathog. 15, e1008009 (2019).

    Article  Google Scholar 

  5. Cheetham, S. et al. Pathogenesis of a genogroup II human norovirus in gnotobiotic pigs. J. Virol. 80, 10372–10381 (2006).

    Article  CAS  Google Scholar 

  6. Souza, M., Azevedo, M. S., Jung, K., Cheetham, S. & Saif, L. J. Pathogenesis and immune responses in gnotobiotic calves after infection with the genogroup II.4-HS66 strain of human norovirus. J. Virol. 82, 1777–1786 (2008).

    Article  CAS  Google Scholar 

  7. Taube, S. et al. A mouse model for human norovirus. mBio https://doi.org/10.1128/mBio.00450-13 (2013).

  8. Sullivan, C. et al. Using zebrafish models of human influenza A virus infections to screen antiviral drugs and characterize host immune cell responses. J. Vis. Exp. https://doi.org/10.3791/55235 (2017).

  9. Harvie, E. A. & Huttenlocher, A. Non-invasive imaging of the innate immune response in a zebrafish larval model of Streptococcus iniae infection. J. Vis. Exp. https://doi.org/10.3791/52788 (2015).

  10. Mazon Moya, M. J., Colucci-Guyon, E. & Mostowy, S. Use of Shigella flexneri to study autophagy-cytoskeleton interactions. J. Vis. Exp. https://doi.org/10.3791/51601 (2014).

  11. Howe, K. et al. The zebrafish reference genome sequence and its relationship to the human genome. Nature 496, 498–503 (2013).

    Article  CAS  Google Scholar 

  12. Aleström, P. et al. Zebrafish: housing and husbandry recommendations. Lab Anim. https://doi.org/10.1177/0023677219869037 (2019).

  13. Costantini, V. et al. Human norovirus replication in human intestinal enteroids as model to evaluate virus inactivation. Emerg. Infect. Dis. 24, 1453–1464 (2018).

    Article  CAS  Google Scholar 

  14. Takaki, K., Davis, J. M., Winglee, K. & Ramakrishnan, L. Evaluation of the pathogenesis and treatment of Mycobacterium marinum infection in zebrafish. Nat. Protoc. 8, 1114–1124 (2013).

    Article  Google Scholar 

  15. Saydmohammed, M. & Tsang, M. High-throughput automated chemical screens in zebrafish. Methods Mol. Biol. (Clifton, N.J.) 1683, 383–393 (2018).

    Article  CAS  Google Scholar 

  16. Gray, C. et al. Simultaneous intravital imaging of macrophage and neutrophil behaviour during inflammation using a novel transgenic zebrafish. Thromb. Haemost. 105, 811–819 (2011).

    Article  CAS  Google Scholar 

  17. Willis, A. R. et al. Shigella-induced emergency granulopoiesis protects zebrafish larvae from secondary infection. mBio 9, e00933–00918 (2018).

    Article  CAS  Google Scholar 

  18. Miskolci, V. et al. Distinct inflammatory and wound healing responses to complex caudal fin injuries of larval zebrafish. eLife 8, e45976 (2019).

    Article  Google Scholar 

  19. Burgos, J. S., Ripoll-Gomez, J., Alfaro, J. M., Sastre, I. & Valdivieso, F. Zebrafish as a new model for herpes simplex virus type 1 infection. Zebrafish 5, 323–333 (2008).

    Article  CAS  Google Scholar 

  20. Gabor, K. A. et al. Influenza A virus infection in zebrafish recapitulates mammalian infection and sensitivity to anti-influenza drug treatment. Dis. Model. Mech. 7, 1227–1237 (2014).

    Article  CAS  Google Scholar 

  21. Palha, N. et al. Real-time whole-body visualization of Chikungunya virus infection and host interferon response in zebrafish. PLoS Pathog. 9, e1003619 (2013).

    Article  CAS  Google Scholar 

  22. Wilson, C. Aspects of larval rearing. ILAR J. 53, 169–178 (2012).

    Article  CAS  Google Scholar 

  23. Gore, A. V., Pillay, L. M., Venero Galanternik, M. & Weinstein, B. M. The zebrafish: a fintastic model for hematopoietic development and disease. Wiley Interdiscip. Rev. Dev. Biol. 7, e312–e312 (2018).

    Article  Google Scholar 

  24. Goody, M. F., Sullivan, C. & Kim, C. H. Studying the immune response to human viral infections using zebrafish. Dev. Comp. Immunol. 46, 84–95 (2014).

    Article  CAS  Google Scholar 

  25. Kanther, M. & Rawls, J. F. Host-microbe interactions in the developing zebrafish. Curr. Opin.Immunol. 22, 10–19 (2010).

    Article  CAS  Google Scholar 

  26. Lewis, K. L., Del Cid, N. & Traver, D. Perspectives on antigen presenting cells in zebrafish. Dev. Comp. Immunol. 46, 63–73 (2014).

    Article  CAS  Google Scholar 

  27. Lister, J. A., Robertson, C. P., Lepage, T., Johnson, S. L. & Raible, D. W. nacre encodes a zebrafish microphthalmia-related protein that regulates neural-crest-derived pigment cell fate. Development 126, 3757–3767 (1999).

    CAS  PubMed  Google Scholar 

  28. Karlsson, J., von Hofsten, J. & Olsson, P. E. Generating transparent zebrafish: a refined method to improve detection of gene expression during embryonic development. Mar. Biotechnol. 3, 522–527 (2001).

    Article  CAS  Google Scholar 

  29. Stals, A. et al. Multiplex real-time RT-PCR for simultaneous detection of GI/GII noroviruses and murine norovirus 1. J. Virol. Methods 161, 247–253 (2009).

    Article  CAS  Google Scholar 

  30. Kageyama, T. et al. Broadly reactive and highly sensitive assay for Norwalk-like viruses based on real-time quantitative reverse transcription-PCR. J. Clin. Microbiol. 41, 1548–1557 (2003).

    Article  CAS  Google Scholar 

  31. Balla, K. M., Rice, M. C., Gagnon, J. A. & Elde, N. C. Linking virus discovery to immune responses visualized during zebrafish infections. Curr. Biol. 30, 2092–2103.e2095 (2020).

    Article  CAS  Google Scholar 

  32. Giusti, A. et al. Safety assessment of compounds after in vitro metabolic conversion using zebrafish eleuthero embryos. Int. J. Mol. Sci. https://doi.org/10.3390/ijms20071712 (2019).

Download references

Acknowledgements

We very much appreciate the expert technical assistance and dedication of J. Rymenants, L. Bervoets and J. Maes. We thank the KU Leuven aquatic facility for breeding the zebrafish larvae. We thank the pediatrics department and the CEMOL Molecular Diagnostic department of the University Hospital of Leuven for the collaboration. J.V.D. and the research leading to these results has received funding from the Scientific Fund for Research of Flanders (FWO) as an SB doctoral fellow. A.C. is supported by internal funding from the KU Leuven.

Author information

Authors and Affiliations

Authors

Contributions

Designed experiments: J.V.D., A.C., S.R., S.T., J.R.-P. Performed experiments: J.V.D., A.C., J.K. Analyzed data: J.V.D., A.C., J.K., S.T., J.R.-P. Supervised the experiments: A.N., P.d.W., S.T., J.N., J.R.-P. Wrote manuscript: J.V.D., A.C., S.T., J.R.-P. Graphs: J.V.D., A.C., J.K., S.T., J.R.-P. Figures and videos: A.C., J.V.D. All authors reviewed and edited the protocol.

Corresponding author

Correspondence to Joana Rocha-Pereira.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Jean-Pieere Levraud and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Van Dycke, J. et al. PLoS Pathog. 15, e1008009 (2019): https://doi.org/10.1371/journal.ppat.1008009

Takaki, K. et al. Nat. Prot. 8, 1114–1124 (2013): https://doi.org/10.1038/nprot.2013.068

This protocol is an extension to: Nat. Protoc. 8, 1114–1124 (2013): https://doi.org/10.1038/nprot.2013.068

Supplementary information

Reporting Summary

Supplementary Video 1

Clipping of the microinjection needle

Supplementary Video 2

Correct size bolus

Supplementary Video 3

Microinjection in the yolk of 3 dpf zebrafish larvae

Supplementary Video 4

Dechorionation of 2 dpf zebrafish larvae

Supplementary Video 5

Microinjection in the pericardial cavity of 3 dpf zebrafish larvae

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Van Dycke, J., Cuvry, A., Knickmann, J. et al. Infection of zebrafish larvae with human norovirus and evaluation of the in vivo efficacy of small-molecule inhibitors. Nat Protoc 16, 1830–1849 (2021). https://doi.org/10.1038/s41596-021-00499-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-021-00499-0

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research