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:

Chikungunya virus cell-to-cell transmission is mediated by intercellular extensions in vitro and in vivo

Abstract

Chikungunya virus (CHIKV) has recently emerged to cause millions of human infections worldwide. Infection can induce the formation of long intercellular extensions that project from infected cells and form stable non-continuous membrane bridges with neighbouring cells. The mechanistic role of these intercellular extensions in CHIKV infection was unclear. Here we developed a co-culture system and flow cytometry methods to quantitatively evaluate transmission of CHIKV from infected to uninfected cells in the presence of neutralizing antibody. Endocytosis and endosomal acidification were critical for virus cell-to-cell transmission, while the CHIKV receptor MXRA8 was not. By using distinct antibodies to block formation of extensions and by evaluation of transmission in HeLa cells that did not form extensions, we showed that intercellular extensions mediate CHIKV cell-to-cell transmission. In vivo, pre-treatment of mice with a neutralizing antibody blocked infection by direct virus inoculation, while adoptive transfer of infected cells produced antibody-resistant host infection. Together our data suggest a model in which the contact sites of intercellular extensions on target cells shield CHIKV from neutralizing antibodies and promote efficient intercellular virus transmission both in vitro and in vivo.

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: Cell-to-cell transmission promotes CHIKV infection in cell culture.
Fig. 2: Role of MXRA8 and the endocytic pathway in cell-to-cell transmission.
Fig. 3: Tetherin expression inhibits CHIKV cell-to-cell transmission.
Fig. 4: Properties of CHIKV infection in HeLa cells.
Fig. 5: Inhibition of ILE blocks cell-to-cell transmission.
Fig. 6: CHIKV cell-to-cell transmission in mice.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are available within the paper, its Extended data or Source data files. Representative microscopy images are included in the main or extended data figures. Additional microscopy image files are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Kuhn, R. J. in Fields Virology: Emerging Viruses-Volume 1 Vol. 1 (eds Howley, P. M. & Knipe, D. M.) Ch. 5, 170–193 (Lippincott Williams & Wilkins, 2021).

  2. Weaver, S. C., Winegar, R., Manger, I. D. & Forrester, N. L. Alphaviruses: population genetics and determinants of emergence. Antivir. Res 94, 242–257 (2012).

    CAS  PubMed  Google Scholar 

  3. Silva, L. A. & Dermody, T. S. Chikungunya virus: epidemiology, replication, disease mechanisms, and prospective intervention strategies. J. Clin. Invest. 127, 737–749 (2017).

    PubMed  PubMed Central  Google Scholar 

  4. Baxter, V. K. & Heise, M. T. Immunopathogenesis of alphaviruses. Adv. Virus Res. 107, 315–382 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. McCarthy, M. K., Davenport, B. J. J. & Morrison, T. E. Chronic chikungunya virus disease. Curr. Top. Microbiol. Immunol. 435, 55–80 (2022).

    CAS  PubMed  Google Scholar 

  6. Robinson, M. C. An epidemic of virus disease in Southern Province, Tanganyika Territory, in 1952–53. I. Clinical features. Trans. R. Soc. Trop. Med. Hyg. 49, 28–32 (1955).

    CAS  PubMed  Google Scholar 

  7. Morrison, T. E. Reemergence of chikungunya virus. J. Virol. 88, 11644–11647 (2014).

    PubMed  PubMed Central  Google Scholar 

  8. Levi, L. I. & Vignuzzi, M. Arthritogenic alphaviruses: a worldwide emerging threat? Microorganisms 7, 133 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Brown, R. S., Wan, J. J. & Kielian, M. The alphavirus exit pathway: what we know and what we wish we knew. Viruses 10, 89 (2018).

    PubMed  PubMed Central  Google Scholar 

  10. Holmes, A. C., Basore, K., Fremont, D. H. & Diamond, M. S. A molecular understanding of alphavirus entry. PLoS Pathog. 16, e1008876 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Kielian, M., Chanel-Vos, C. & Liao, M. Alphavirus entry and membrane fusion. Viruses 2, 796–825 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Cifuentes-Munoz, N., El Najjar, F. & Dutch, R. E. Viral cell-to-cell spread: conventional and non-conventional ways. Adv. Virus Res 108, 85–125 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhong, P., Agosto, L. M., Munro, J. B. & Mothes, W. Cell-to-cell transmission of viruses. Curr. Opin. Virol. 3, 44–50 (2013).

    CAS  PubMed  Google Scholar 

  14. McDonald, D. et al. Recruitment of HIV and its receptors to dendritic cell–T cell junctions. Science 300, 1295–1297 (2003).

    CAS  PubMed  Google Scholar 

  15. Sowinski, S. et al. Membrane nanotubes physically connect T cells over long distances presenting a novel route for HIV-1 transmission. Nat. Cell Biol. 10, 211–219 (2008).

    CAS  PubMed  Google Scholar 

  16. Sherer, N. M. et al. Retroviruses can establish filopodial bridges for efficient cell-to-cell transmission. Nat. Cell Biol. 9, 310–315 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Favoreel, H. W., Van Minnebruggen, G., Adriaensen, D. & Nauwynck, H. J. Cytoskeletal rearrangements and cell extensions induced by the US3 kinase of an alphaherpesvirus are associated with enhanced spread. Proc. Natl Acad. Sci. USA 102, 8990–8995 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Hahon, N. & Zimmerman, W. D. Chikungunya virus infection of cell monolayers by cell-to-cell and extracellular transmission. Appl. Microbiol. 19, 389–391 (1970).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Lee, C. Y. et al. Chikungunya virus neutralization antigens and direct cell-to-cell transmission are revealed by human antibody-escape mutants. PLoS Pathog. 7, e1002390 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Martinez, M. G. & Kielian, M. Intercellular extensions are induced by the alphavirus structural proteins and mediate virus transmission. PLoS Pathog. 12, e1006061 (2016).

    PubMed  PubMed Central  Google Scholar 

  21. Jose, J., Taylor, A. B. & Kuhn, R. J. Spatial and temporal analysis of alphavirus replication and assembly in mammalian and mosquito cells. mBio 8, e02294–02216 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Meshram, C. D. et al. Multiple host factors interact with the hypervariable domain of chikungunya virus nsP3 and determine viral replication in cell-specific mode. J. Virol. 92, e00838–18 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Pal, P. et al. Development of a highly protective combination monoclonal antibody therapy against chikungunya virus. PLoS Pathog. 9, e1003312 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Sun, S. et al. Structural analyses at pseudo atomic resolution of chikungunya virus and antibodies show mechanisms of neutralization. eLife 2, e00435 (2013).

    PubMed  PubMed Central  Google Scholar 

  25. Jolly, C. & Sattentau, Q. J. Retroviral spread by induction of virological synapses. Traffic 5, 643–650 (2004).

    CAS  PubMed  Google Scholar 

  26. Zhang, R. et al. Mxra8 is a receptor for multiple arthritogenic alphaviruses. Nature 557, 570–574 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang, R. et al. Expression of the Mxra8 receptor promotes alphavirus infection and pathogenesis in mice and Drosophila. Cell Rep. 28, 2647–2658 e2645 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. McCluskey, A. et al. Building a better dynasore: the dyngo compounds potently inhibit dynamin and endocytosis. Traffic 14, 1272–1289 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Stenmark, H. et al. Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis. EMBO J. 13, 1287–1296 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Hoornweg, T. E. et al. Dynamics of chikungunya virus cell entry unraveled by single-virus tracking in living cells. J. Virol. 90, 4745–4756 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Glomb-Reinmund, S. & Kielian, M. The role of low pH and disulfide shuffling in the entry and fusion of Semliki Forest virus and Sindbis virus. Virology 248, 372–381 (1998).

    CAS  PubMed  Google Scholar 

  32. Neil, S. J. The antiviral activities of tetherin. Curr. Top. Microbiol. Immunol. 371, 67–104 (2013).

    CAS  PubMed  Google Scholar 

  33. Ooi, Y. S., Dube, M. & Kielian, M. BST2/tetherin inhibition of alphavirus exit. Viruses 7, 2147–2167 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Wan, J. J., Ooi, Y. S. & Kielian, M. Mechanism of tetherin inhibition of alphavirus release. J. Virol. 93, e02165–18 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Pal, P. et al. Chikungunya viruses that escape monoclonal antibody therapy are clinically attenuated, stable, and not purified in mosquitoes. J. Virol. 88, 8213–8226 (2014).

    PubMed  PubMed Central  Google Scholar 

  36. Gorchakov, R. et al. Attenuation of chikungunya virus vaccine strain 181/clone 25 is determined by two amino acid substitutions in the E2 envelope glycoprotein. J. Virol. 86, 6084–6096 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Fox, J. M. et al. Optimal therapeutic activity of monoclonal antibodies against chikungunya virus requires Fc–FcγR interaction on monocytes. Sci. Immunol. 4, eaav5062 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhong, P. et al. Cell-to-cell transmission can overcome multiple donor and target cell barriers imposed on cell-free HIV. PLoS ONE 8, e53138 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Jolly, C., Booth, N. J. & Neil, S. J. Cell-cell spread of human immunodeficiency virus type 1 overcomes tetherin/BST-2-mediated restriction in T cells. J. Virol. 84, 12185–12199 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Young, A. R. et al. Dermal and muscle fibroblasts and skeletal myofibers survive chikungunya virus infection and harbor persistent RNA. PLoS Pathog. 15, e1007993 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Hoarau, J. J. et al. Persistent chronic inflammation and infection by chikungunya arthritogenic alphavirus in spite of a robust host immune response. J. Immunol. 184, 5914–5927 (2010).

    CAS  PubMed  Google Scholar 

  42. Hawman, D. W. et al. Pathogenic chikungunya virus evades B cell responses to establish persistence. Cell Rep. 16, 1326–1338 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Ashbrook, A. W. et al. Residue 82 of the chikungunya virus E2 attachment protein modulates viral dissemination and arthritis in mice. J. Virol. 88, 12180–12192 (2014).

    PubMed  PubMed Central  Google Scholar 

  44. Liljeström, P., Lusa, S., Huylebroeck, D. & Garoff, H. In vitro mutagenesis of a full-length cDNA clone of Semliki Forest virus: the small 6,000-molecular-weight membrane protein modulates virus release. J. Virol. 65, 4107–4113 (1991).

    PubMed  PubMed Central  Google Scholar 

  45. Hardwick, J. M. & Levine, B. Sindbis virus vector system for functional analysis of apoptosis regulators. Methods Enzymol. 322, 492–508 (2000).

    CAS  PubMed  Google Scholar 

  46. Dube, M., Etienne, L., Fels, M. & Kielian, M. Calcium-dependent rubella virus fusion occurs in early endosomes. J. Virol. 90, 6303–6313 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Poddar, S., Hyde, J. L., Gorman, M. J., Farzan, M. & Diamond, M. S. The interferon-stimulated gene IFITM3 restricts infection and pathogenesis of arthritogenic and encephalitic alphaviruses. J. Virol. 90, 8780–8794 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Crill, W. D. & Chang, G. J. Localization and characterization of flavivirus envelope glycoprotein cross-reactive epitopes. J. Virol. 78, 13975–13986 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Quiroz, J. A. et al. Human monoclonal antibodies against chikungunya virus target multiple distinct epitopes in the E1 and E2 glycoproteins. PLoS Pathog. 15, e1008061 (2019).

    PubMed  PubMed Central  Google Scholar 

  50. Voss, J. E. et al. Glycoprotein organization of chikungunya virus particles revealed by X-ray crystallography. Nature 468, 709–712 (2010).

    CAS  PubMed  Google Scholar 

  51. Kielian, M., Jungerwirth, S., Sayad, K. U. & DeCandido, S. Biosynthesis, maturation, and acid-activation of the Semliki Forest virus fusion protein. J. Virol. 64, 4614–4624 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Meyer, W. J. & Johnston, R. E. Structural rearrangement of infecting Sindbis virions at the cell surface: mapping of newly accessible epitopes. J. Virol. 67, 5117–5125 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all the members of our laboratory for their helpful discussions, C. Martin for comments on the paper, and L. Kim and A. Fayed for technical support. We thank the Einstein Analytical Imaging Facility and the Flow Cytometry Core Facility for use of their instruments, and A. Briceno for training on the SP5 confocal microscope and J. Zhang for training on the BD LSR-II analyser. We thank E. Frolova (University of Alabama) for providing the CHIKV-GFP (181/25) infectious clone, Z. Bornholdt (Mapp Biopharmaceutial) for providing mAb chCHK-152, and F. Rey (Institut Pasteur) for helpful discussions on CHIKV antibodies. This work was supported by National Institutes of Health grants to M.K. from NIGMS (R01GM057454), to T.E.M. from NIAID (R01AI141436), to J.R.L. from NIAID (R01AI125462), to M.S.D. from NIAID (R01AI143673) and by NCI Cancer Center Support Grant P30CA013330. The development of mAb E10-18 was funded by the Investissement d’Avenir programme, Laboratoire d’Excellence ‘Integrative Biology of Emerging Infectious Diseases’ (ANR-10-LABX-62-IBEID), the ‘Investissements d’Avenir’ programme (ANR-10-IHUB-0002, ANR-15-CE15-00029 ZIKAHOST and the INCEPTION programme ANR-16-CONV-0005), Institut Pasteur, Inserm. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. The content of this paper is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

P.Y. and M.K. conceived the project; P.Y. performed the experimental work with B.J.D. performing the mouse experiments; J.J.W. provided important experimental background on ILE, antibody blocking of ILE and ELISA, M.K. and T.E.M. supervised the research; A.K., M.S.D., B.C.W., K.T., T.C., M.L. and J.R. L. developed and contributed key reagents; P.Y. and M.K. wrote and edited the paper. All authors reviewed and revised the paper and agreed to the published version of the paper. T.C., now deceased, reviewed and approved an earlier version of this paper.

Corresponding author

Correspondence to Margaret Kielian.

Ethics declarations

Competing interests

P.Y., B.J.D., J.J.W., A.S.K., B.C.W., K.T., T. C., M. L., T.E.M. and M.K. report no competing interests. J.R.L. is a paid consultant for Celdara Medical, LLC. M.S.D. is a consultant for Inbios, Vir Biotechnology, Senda Biosciences, Ocugen, Moderna and Immunome. The Diamond laboratory has received unrelated funding support in sponsored research agreements from Vir Biotechnology, Moderna, Generate Biomedicine and Emergent BioSolutions.

Peer review

Peer review information

Nature Microbiology thanks Young Ki Choi, Rebecca Dutch and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Growth properties of CHIKV and CHIKV-GFP in MEF cells.

a, Schematic representation of CHIKV 181/25 and CHIKV 181/25 GFP infectious clone constructs. b, Growth kinetics of CHIKV and CHIKV-GFP in MEF cells. MEF cells were inoculated with CHIKV or CHIKV-GFP (MOI = 10) for 2 h at 37 °C, then washed to remove unbound virus. Virus production at the indicated times was quantitated by ICA. The graphs represent the means and range for 2 independent experiments. c, Schematic of experiments to test CHIKV cell-to-cell transmission. After infection of producer cells, dye-labeled target cells are added, co-culture is performed under various conditions, and virus in the medium and infection of target cells are quantitated. d, Representative gating scheme to identify uninfected and CHIKV-GFP-infected producer and target cells.

Source data

Extended Data Fig. 2 Infection of MEF or U-2 OS target cells in the presence of mAb DEN-4G2 or chCHK-152.

a, MEF producer cells were inoculated with CHIKV-GFP (MOI = 10) for 2 h, washed, and then co-cultured for 12 h with MEF target cells in the presence of the indicated concentrations of mAb chCHK-152 or DEN-4G2, a control mAb against an unrelated virus. The media were collected, and cells were analyzed by flow cytometry. Data are representative of 3 independent experiments. b, Quantitation of target cell infection in samples prepared as in Extended Data 2a. The bar graph represents the mean ± S.D. of 3 independent experiments (shown as points). Statistical significance was calculated by unpaired two-tailed t-tests. c, U-2 OS producer cells were infected with CHIKV-GFP (MOI = 10), washed, and incubated at 37 °C for 2 h. U-2 OS target cells stained with CMRA dye were then plated onto the producer cells in the presence of the indicated concentrations of chCHK-152 and co-cultured for 12 h. Cells were then fixed and analyzed by flow cytometry. d, Quantitation of target cell infection in samples from Extended Data 2c. The bar graph represents the mean ± S.D. of 3 independent experiments (shown as points). Statistical significance was calculated by unpaired two-tailed t-tests.

Source data

Extended Data Fig. 3 Effect of Bafilomycin or Dyngo-4a on ILE formation.

a, Examples of ILE induced by CHIKV-GFP infection in U-2 OS treated with Bafilomycin A1 or Dyngo-4a. U-2 OS cells were infected with CHIKV-GFP (MOI = 0.5) for 1 h, treated with Bafilomycin A1 (100 nM), Dyngo-4a (60 μM) or DMSO vehicle for 1 h, then cultured for 9 h. ILE visualized as in Fig. 1b. White arrowheads indicate ILE. Bar = 20 μm. b, ILE were quantitated as described in Fig. 1c. Data shown represent the mean of 2 independent experiments, with points showing the results from each experiment.

Source data

Extended Data Fig. 4 Characterization of inducible Rab5 U-2 OS cell lines.

a, Clonal U-2 OS cell lines inducibly expressing GFP-tagged Rab5-WT or Rab5-DN were cultured in the presence or absence of doxycycline (dox) for 16 h and analyzed by flow cytometry. b, The indicated U-2 OS cell lines were incubated in the presence or absence of doxycycline for 16 h and infected with SFV at an MOI of 1 for 12 h. Cells were stained with antibody to detect E2/E1 and quantitated by flow cytometry. The bar graph represents the mean ± S.D of 3 independent experiments (shown as points). Statistical significance was calculated using unpaired two-tailed multiple t-tests. c, Clonal U-2 OS cell lines were cultured in the presence of doxycycline (dox) for 16 h and infected with CHIKV for 11 h. ILE were visualized as in Fig. 1b. White arrowheads indicate ILE. Bar = 20 μm. d, Cells were treated and infected as in panel c, and ILE in infected cells were quantitated as in Fig. 1c. Data shown represent the mean of 2 independent experiments (shown as points).

Source data

Extended Data Fig. 5 ILE formation in U-2 OS-tetherin cell lines.

Examples of ILE induced by CHIKV-GFP infection in U-2 OS expressing WT-tetherin and L-tetherin vs. the parental cells. The indicated U-2 OS cells were incubated with or without doxycycline for 16 h and infected with CHIKV-GFP (MOI = 0.5) for 11 h. Cells were permeabilized and stained with antibodies against the E2/E1 proteins (red, pseudo color) and tubulin (green, pseudo color). The GFP reporter channel is not shown. White arrowheads indicate ILE. Bar = 20 μm.

Extended Data Fig. 6 MAb E10-18 does not inhibit free virus infection of target cells.

MEF producer cells were infected with CHIKV-GFP (MOI = 10) for 2 h, washed, and then co-cultured for 12 h with MEF target cells in the presence of the indicated concentrations of mAbs chCHK-152 or E10-18. Infection of target cells was determined by flow cytometry. The bar graph represents the mean ± S.D. of 3 independent experiments (shown as points). Statistical significance was calculated by unpaired two-tailed t-tests.

Source data

Extended Data Fig. 7 E1-K61T-G64S escapes chCHK-166 inhibition of ILE formation.

Examples of ILE from experiment in Fig. 5h. MEF cells were infected with CHIKV-GFP or CHKV-GFP E1-K61T-G64S for 2 h, then cultured in presence or absence of 20 μg/mL mAb chCHK-166 for 9 h. ILE were visualized as in Fig. 1b. White arrowheads indicate ILE. Bar = 20 μm.

Extended Data Fig. 8 CHK-152 N297Q pre-treatment can block CHIKV infection in ipsilateral ankle.

a, WT C57BL/6 mice were treated with 100 μg of chCHK-152 N297Q i.p. (n = 2) or with an equal volume of PBS (n = 1) as a negative control. At 6 h following antibody or PBS treatment, mice were inoculated with 103 PFU CHIKV-Venus s.c. in the left footpad. At 24 h post-virus inoculation, single cell suspensions were prepared from collagenase/DNase digested ipsilateral ankle tissue, incubated with fixable viability dye (Vi-421), and stained with CD45 BUV395. Figure created with Biorender.com. b, Representative dot plots showing the gating scheme to identify host cells that are productively infected with CHIKV-Venus. c, Flow plots from PBS and chCHK-152 N297Q pretreated mice. All plots are gated on viable singlet CD45 cells. d, Frequency of Venus+ cells in the CD45 fraction of ipsilateral ankle tissue.

Extended Data Fig. 9 FACS gating strategies for Fig. 6.

a, Representative gating scheme to identify donor Violet+ MEF cells, and endogenous host Violet- CD45 cells isolated from the ipsilateral ankle. b, In all MEF adoptive transfer experiments, a group of control mice was PBS-treated and adoptively transferred with 106 mock-infected Cell-Trace Violet loaded MEF cells. Flow plots show that the presence of Violet+ MEF cells does not interfere with detection of Venus+ cells.

Extended Data Fig. 10 ILE formation in primary joint cells infected with CHIKV ex vivo.

Examples of ILE quantitated in Fig. 6f. Ankle tissue from C57BL/6 mice was collected, subjected to collagenase/Dnase digestion, and cultured for 24 h. The cells were infected with WT CHIKV strain AF15561 or CHIKV strain 181/25 for 16 h, fixed and stained with antibodies to E1/E2(red) and tubulin (green). White arrowheads indicate ILE. Bar = 20 μm.

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yin, P., Davenport, B.J., Wan, J.J. et al. Chikungunya virus cell-to-cell transmission is mediated by intercellular extensions in vitro and in vivo. Nat Microbiol 8, 1653–1667 (2023). https://doi.org/10.1038/s41564-023-01449-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-023-01449-0

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