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:

LRP8 is a receptor for tick-borne encephalitis virus

Abstract

Tick-borne encephalitis virus (TBEV) causes tick-borne encephalitis (TBE), a severe and sometimes life-threatening disease characterized by viral invasion of the central nervous system with symptoms of neuroinflammation1,2. As with other orthoflaviviruses—enveloped, arthropod-borne RNA viruses—host factors required for TBEV entry remain poorly defined. Here we used a genome-scale CRISPR–Cas9-based screen to identify LRP8, an apolipoprotein E and reelin receptor with high expression in the brain, as a TBEV receptor. LRP8 downregulation reduced TBEV infection in human cells, and its overexpression enhanced infection. LRP8 bound directly to the TBEV E glycoprotein and mediated viral attachment and internalization into cells. An LRP8-based soluble decoy blocked infection of human cell lines and neuronal cells and protected mice from lethal TBEV challenge. LRP8’s role as a TBEV receptor has implications for TBEV neuropathogenesis and the development of antiviral countermeasures.

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: Genome-scale CRISPR–Cas9 screen identifies LRP8 as an essential TBEV host factor.
Fig. 2: Mapping binding sites of LRP8 and TBEV E interaction.
Fig. 3: LRP8 mediates TBEV attachment and internalization into cells.
Fig. 4: LRP8 is required for TBEV pathogenesis in mice.

Similar content being viewed by others

Data availability

All data associated with this study are provided in the Article and its Supplementary Information. FASTQ files for CRISPR screens are available at the SRA BioProject under accession number PRJNA1226671: ‘Genome-wide CRISPR screen for tick-borne encephalitis virus host dependency factors’. The following sequences available at NCBI GenBank were used in this study: flavivirus genome sequences (KC806252, DQ401140.3, NC_003687, EF571853.1, OP037819.1, U27495); open reading frames encoding flavivirus C/prM/E (NC_001672, NC_003687.1, AGI15884, KF667312.1, KF769016.1); LRP8 (NM_004631, NM_053073.3); and PCDH1 (NM_002587). Source data are provided with this paper.

Code availability

Custom bash and MATLAB scripts for NGS data processing and CellProfiler pipelines are available at GitHub (https://github.com/chandranlab/mittler_2024).

References

  1. Lindquist, L. & Vapalahti, O. Tick-borne encephalitis. Lancet 371, 1861–1871 (2008).

    Article  PubMed  Google Scholar 

  2. Süss, J. Tick-borne encephalitis 2010: epidemiology, risk areas, and virus strains in Europe and Asia-an overview. Ticks Tick Borne Dis. 2, 2–15 (2011).

    Article  PubMed  Google Scholar 

  3. Van Heuverswyn, J. et al. Spatiotemporal spread of tick-borne encephalitis in the EU/EEA, 2012 to 2020. Euro Surveill. 28, 2200543 (2023).

    PubMed  PubMed Central  Google Scholar 

  4. Albinsson, B. et al. Seroprevalence of tick-borne encephalitis virus and vaccination coverage of tick-borne encephalitis, Sweden, 2018 to 2019. Euro Surveill. 29, 2300221 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Heinz, F. X. et al. Vaccination and tick-borne encephalitis, central Europe. Emerg. Infect. Dis. 19, 69–76 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Erber, W. & Schmitt, H.-J. Self-reported tick-borne encephalitis (TBE) vaccination coverage in Europe: results from a cross-sectional study. Ticks Tick Borne Dis. 9, 768–777 (2018).

    Article  PubMed  Google Scholar 

  7. Kubinski, M. et al. Tick-borne encephalitis virus: a quest for better vaccines against a virus on the rise. Vaccines 8, 451 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ruzek, D. et al. Tick-borne encephalitis in Europe and Russia: review of pathogenesis, clinical features, therapy, and vaccines. Antiviral Res. 164, 23–51 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Hasan, S. S., Sevvana, M., Kuhn, R. J. & Rossmann, M. G. Structural biology of Zika virus and other flaviviruses. Nat. Struct. Mol. Biol. 25, 13–20 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Pierson, T. C. & Diamond, M. S. The continued threat of emerging flaviviruses. Nat. Microbiol. 5, 796–812 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kanojia, A., Sharma, M., Shiraz, R. & Tripathi, S. Flavivirus-host interaction landscape visualized through genome-wide CRISPR screens. Viruses 14, 2164 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. See, W. R., Yousefi, M. & Ooi, Y. S. A review of virus host factor discovery using CRISPR screening. mBio 15, e0320523 (2024).

    Article  PubMed  Google Scholar 

  13. Yousefi, M. et al. GeneRaMeN enables integration, comparison, and meta-analysis of multiple ranked gene lists to identify consensus, unique, and correlated genes. Brief. Bioinform. 25, bbae452 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Han, J. et al. Genome-wide CRISPR/Cas9 screen identifies host factors essential for influenza virus replication. Cell Rep. 23, 596–607 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kim, D. H. et al. Human apolipoprotein E receptor 2. A novel lipoprotein receptor of the low density lipoprotein receptor family predominantly expressed in brain. J. Biol. Chem. 271, 8373–8380 (1996).

    Article  CAS  PubMed  Google Scholar 

  17. Clark, L. E. et al. VLDLR and ApoER2 are receptors for multiple alphaviruses. Nature 602, 475–480 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

  18. Li, W. et al. Shifts in receptors during submergence of an encephalitic arbovirus. Nature 632, 614–621 (2024).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  19. Rosendal, E. et al. Influence of the pre-membrane and envelope proteins on structure, pathogenicity, and tropism of tick-borne encephalitis virus. J. Virol. https://doi.org/10.1128/jvi.00870-25 (2025).

  20. Rey, F. A., Stiasny, K. & Heinz, F. X. Flavivirus structural heterogeneity: implications for cell entry. Curr. Opin. Virol. 24, 132–139 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Anwar, M. N. et al. The interactions of flaviviruses with cellular receptors: implications for virus entry. Virology 568, 77–85 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Palakurty, S. et al. The VLDLR entry receptor is required for the pathogenesis of multiple encephalitic alphaviruses. Cell Rep. 43, 114809 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Monteil, V. M. et al. Crimean-Congo haemorrhagic fever virus uses LDLR to bind and enter host cells. Nat. Microbiol. 9, 1499–1512 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Finkelshtein, D., Werman, A., Novick, D., Barak, S. & Rubinstein, M. LDL receptor and its family members serve as the cellular receptors for vesicular stomatitis virus. Proc. Natl Acad. Sci. USA 110, 7306–7311 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xu, Z.-S. et al. LDLR is an entry receptor for Crimean-Congo hemorrhagic fever virus. Cell Res. 34, 140–150 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ganaie, S. S. et al. Lrp1 is a host entry factor for Rift Valley fever virus. Cell 184, 5163–5178.e24 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schwarz, M. M. et al. Oropouche orthobunyavirus infection is mediated by the cellular host factor Lrp1. Proc. Natl Acad. Sci. USA 119, e2204706119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cosset, F.-L. & Denolly, S. Lipoprotein receptors: a little grease for enveloped viruses to open the lock? J. Biol. Chem. 300, 107849 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zimmerman, O., Holmes, A. C., Kafai, N. M., Adams, L. J. & Diamond, M. S. Entry receptors—the gateway to alphavirus infection. J. Clin. Invest. 133, e165307 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nikolic, J. et al. Structural basis for the recognition of LDL-receptor family members by VSV glycoprotein. Nat. Commun. 9, 1029 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  31. Jangra, R. K. et al. Protocadherin-1 is essential for cell entry by New World hantaviruses. Nature 563, 559–563 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cao, D., Ma, B., Cao, Z., Zhang, X. & Xiang, Y. Structure of Semliki Forest virus in complex with its receptor VLDLR. Cell 186, 2208–2218.e15 (2023).

    Article  CAS  PubMed  Google Scholar 

  33. Adams, L. J. et al. Structural and functional basis of VLDLR usage by eastern equine encephalitis virus. Cell 187, 360–374 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fan, X. et al. Molecular basis for shifted receptor recognition by an encephalitic arbovirus. Cell 188, 2957–2973.e28 (2025).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Song, H. et al. Molecular basis of arthritogenic alphavirus receptor MXRA8 binding to chikungunya virus envelope protein. Cell 177, 1714–1724.e12 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Basore, K. et al. Cryo-EM structure of chikungunya virus in complex with the Mxra8 receptor. Cell 177, 1725–1737 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sevvana, M. & Kuhn, R. J. Mapping the diverse structural landscape of the flavivirus antibody repertoire. Curr. Opin. Virol. 45, 51–64 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mandl, C. W., Allison, S. L., Holzmann, H., Meixner, T. & Heinz, F. X. Attenuation of tick-borne encephalitis virus by structure-based site-specific mutagenesis of a putative flavivirus receptor binding site. J. Virol. 74, 9601–9609 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Vaney, M.-C. et al. Evolution and activation mechanism of the flavivirus class II membrane-fusion machinery. Nat. Commun. 13, 3718 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Malashkevich, V. N., Kammerer, R. A., Efimov, V. P., Schulthess, T. & Engel, J. The crystal structure of a five-stranded coiled coil in COMP: a prototype ion channel? Science 274, 761–765 (1996).

    Article  ADS  CAS  PubMed  Google Scholar 

  41. Jerabek-Willemsen, M., Wienken, C. J., Braun, D., Baaske, P. & Duhr, S. Molecular interaction studies using microscale thermophoresis. Assay Drug Dev. Technol. 9, 342–353 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Fan, Q. W., Iosbe, I., Asou, H., Yanagisawa, K. & Michikawa, M. Expression and regulation of apolipoprotein E receptors in the cells of the central nervous system in culture: a review. J. Am. Aging Assoc. 24, 1–10 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bílý, T. et al. Electron tomography analysis of tick-borne encephalitis virus infection in human neurons. Sci. Rep. 5, 10745 (2015).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  44. Velay, A. et al. Tick-borne encephalitis virus: molecular determinants of neuropathogenesis of an emerging pathogen. Crit. Rev. Microbiol. 45, 472–493 (2019).

    Article  PubMed  Google Scholar 

  45. Fares, M. et al. Pathological modeling of TBEV infection reveals differential innate immune responses in human neurons and astrocytes that correlate with their susceptibility to infection. J. Neuroinflamm. 17, 76 (2020).

    Article  CAS  Google Scholar 

  46. Salát, J. et al. Development and testing of a new tick-borne encephalitis virus vaccine candidate for veterinary use. Vaccine 36, 7257–7261 (2018).

    Article  PubMed  Google Scholar 

  47. Zhang, X. et al. T-cell immunoglobulin and mucin domain 1 (TIM-1) is a functional entry factor for tick-borne encephalitis virus. mBio 13, e0286021 (2022).

    Article  PubMed  Google Scholar 

  48. He, L. et al. Single-cell RNA sequencing of mouse brain and lung vascular and vessel-associated cell types. Sci. Data 5, 180160 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Daneman, R. et al. The mouse blood-brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS ONE 5, e13741 (2010).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  50. Moreno, H., Möller, R., Fedeli, C., Gerold, G. & Kunz, S. Comparison of the innate immune responses to pathogenic and nonpathogenic clade B new world arenaviruses. J. Virol. 93, e00148–19 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Calvo-Garrido, J. et al. Protocol for the derivation, culturing, and differentiation of human iPS-cell-derived neuroepithelial stem cells to study neural differentiation in vitro. STAR Protoc. 2, 100528 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Stirling, D. R. et al. CellProfiler 4: improvements in speed, utility and usability. BMC Bioinform. 22, 433 (2021).

    Article  Google Scholar 

  53. Spuul, P., Balistreri, G., Kääriäinen, L. & Ahola, T. Phosphatidylinositol 3-kinase-, actin-, and microtubule-dependent transport of Semliki Forest Virus replication complexes from the plasma membrane to modified lysosomes. J. Virol. 84, 7543–7557 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Thomas, J. M., Klimstra, W. B., Ryman, K. D. & Heidner, H. W. Sindbis virus vectors designed to express a foreign protein as a cleavable component of the viral structural polyprotein. J. Virol. 77, 5598–5606 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Brown, R. S., Anastasakis, D. G., Hafner, M. & Kielian, M. Multiple capsid protein binding sites mediate selective packaging of the alphavirus genomic RNA. Nat. Commun. 11, 4693 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Vratskikh, O. et al. Dissection of antibody specificities induced by yellow fever vaccination. PLoS Pathog. 9, e1003458 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Stiasny, K., Brandler, S., Kössl, C. & Heinz, F. X. Probing the flavivirus membrane fusion mechanism by using monoclonal antibodies. J. Virol. 81, 11526–11531 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Shao, W., Sharma, R., Clausen, M. H. & Scheller, H. V. Microscale thermophoresis as a powerful tool for screening glycosyltransferases involved in cell wall biosynthesis. Plant Methods 16, 99 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Paez, E. Valencia and M. Ramirez, K. Dempsey, C. O’Brien, A. I. Kuehne and N. M. Josleyn for laboratory management and technical assistance; K. Cogliano for programmatic and administrative support; K. Stiasny for providing TBEV monoclonal antibodies B2 and B4; the members of the Einstein Epigenomics Core and Computational Genomics Core for NGS analysis and data analysis support; the staff at the Einstein Flow Cytometry Core Facility (supported by the Einstein National Cancer Institute’s cancer center support grant P30CA013330) for surface flow cytometry sorting of cell lines; the members of the Einstein Macromolecular Therapeutics Development Facility for protein characterization by size-exclusion chromatography; and the staff at the Karolinska Institutet ANA Futura BSL3 Core Facility (supported by the Infrastructure Board at Karolinska Institutet) for enabling virus studies under high-containment. US National Institutes of Health grant R01AI132633 (K.C.); Marianne and Marcus Wallenberg Foundation (S.G.-R.); Region Stockholm (Clinical Research Appointment) (S.G.-R.); Swedish Research Council (Dnr 2020-06249 and 2021-06602) (S.G.-R.); Swedish Research Council (Dnr 2020-06224) (A.K.Ö.); US National Institute of Allergy and Infectious Diseases grants R01AI165932 and R01AI174584 (B.M.); Congressionally Directed Medical Research Programs grant HT9425-24-1-0873 (C.F.); US National Institute of Allergy and Infectious Diseases grant R21AI182834 (C.K.M.); and French National Research Agency grants ANR-18-CE92-0006, ANR-22-CE35-0004 and ANR-10-LABX-62-10 IBEID (F.A.R.). Opinions, conclusions, interpretations and recommendations are those of the authors and are not necessarily endorsed by the US Department of the Army, the US Department of Defense or the US Department of Health and Human Services.

Author information

Authors and Affiliations

Authors

Contributions

B.M. and J.H. generated and validated the CRISPR–Cas9 KO library in A549 cells. R.V. performed the CRISPR–Cas9 genetic screen and NGS, the latter with support from E.M.; K.C. and E.M. analysed NGS data. D.H. and A.L.T. established the single-cycle RVP system. A.L.T., J.J. and E.M. generated cell lines, RVPs and recombinant proteins and performed infectivity studies with RVPs. P.-T.-H.T., C.F., J.J., E.M., M.L., E.C., E.R.W., W.C. and C.K.M. performed infectivity studies with authentic viruses. E.K. generated cell lines lacking expression of LDLR homologues with assistance of J.B. R.M.O., G.B.-S., F.A.R., E.R., A.K.Ö. and M.M.S. provided critical reagents. A.L.T. and E.M. designed and executed immunoprecipitation, ELISA and BLI experiments. V.K.M., A.L.T. and E.M. designed and executed MST experiments. P.-T.-H.T. performed virus attachment and internalization experiments, including analysis of confocal microscopy data. C.F. performed in vivo challenge studies with assistance from R.R.B., as well as ISH studies with support from X.Z., J.L.R. and S.K.; P.-T.-H.T. generated and characterized iPS-cell-derived neurons with critical support by M.S. and A.F.; A.K.Ö., A.S.H., M.S., C.F., M.K., A.F., E.M., F.A.R., E.G., G.B.-S., J.K., K.C. and S.G.-R. provided project supervision and participated in study conceptualization. K.C. and S.G.-R. acquired funding. C.F., E.M., A.L.T., P.-T.-H.T. and K.C. wrote the original draft of the manuscript and all of the authors participated in reviewing and editing.

Corresponding authors

Correspondence to Eva Mittler, Andrew S. Herbert, Kartik Chandran or Sara Gredmark-Russ.

Ethics declarations

Competing interests

K.C. and A.S.H. are scientific advisors to and hold equity in Integrum Scientific. K.C. holds equity in Eitr Biologics. A.F. is the chief scientific officer of CCRM Nordic. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Amy Hartman, Richard Kuhn and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Analysis of rank uniqueness of top hits in CRISPR/Cas9 screen.

The rank uniqueness of the top 100 gene hits in our screen relative to the top 100 hits in each of 25 previously published flavivirus screen datasets was analysed on the GeneRaMeN webserver13. Rank uniqueness was determined using the Robust Rank Aggregation (RRA) method with a significance cutoff of p = 0.05. A heatmap of the top 50 unique ranks in each screen is shown. The colour scale indicates the rank of each hit in its original screen. The gray boxes indicate genes that were not present in the gene list for the screen. Both hit genes and screens are clustered according to similarity. The top 10 hits of our screens are indicated in red.

Extended Data Fig. 2 Establishment and characterization of cell lines with reduced or increased LRP8 expression.

a, A549 single-cell LRP8-knockout (KO) clones were generated by CRISPR/Cas9 engineering. Sequences of LRP8-KO alleles in clones 1 and 2 are shown. The sgRNA target sequence is highlighted in blue. b, A549 LRP8-KO and wild-type (WT) cell lines were immunostained with an anti-LRP8 antibody and visualized by fluorescence microscopy. Experiments were performed three times with similar results. Scale bar, 20 μm. c (left panel), WT and A549 LRP8-KO cell lines as well as A549 LRP8-KO cells overexpressing Flag-tagged LRP8 were immunostained with an anti-Flag mAb and visualized by fluorescence microscopy. The experiment was performed twice and representative images from one experiment are shown. Scale bar, 20 μm. (Right panel), LRP8 expression in indicated cells was determined by western blotting using LRP8-specific antibodies. The housekeeping protein β-actin was included as a loading control. Data from one experiment representative of two independent experiments are shown. Mr, relative molecular weight (K denotes ×1,000). d, A549 cells were treated with siRNAs targeting LRP8, VLDLR, and LRP1 or non-targeting (NT) siRNAs, respectively. Reduction of LRP8, VLDLR and LRP1 expression was determined by western blotting. The housekeeping protein GAPDH was included as a loading control. Experiments were performed two times with similar results. Mr, relative molecular weight (K denotes ×1,000). e (left panel), A549 clones overexpressing Flag-tagged LRP8 and WT cell lines were either subjected to immunostaining with an anti-LRP8 antibody to detect cell surface-localized LRP8 (‘Surface’ staining) or alternatively permeabilized and immunostained with an anti-Flag mAb to detect LRP8 throughout the cell (‘Intracellular’ staining). Images from one experiment representative of three independent experiments with similar results are shown. Scale bar, 20 μm. (Right panel), LRP8 expression in WT A549 cells and cells overexpressing Flag-tagged LRP8 was determined by western blotting using LRP8-specific antibodies. The housekeeping protein β-actin was included as a loading control. The experiment was performed twice and representative results from one experiment are shown. Mr, relative molecular weight (K denotes ×1,000). For gel source data, see Supplementary Information Fig. 2.

Extended Data Fig. 3 LRP8 does not support entry by divergent flaviviruses.

a, A549 WT, LRP8-KO and LRP8-overexpressing cell lines were exposed to TBEVToro and TBEVHypr. Infection levels were determined at 6 h.p.i. (TBEVHypr) and 24 h.p.i. (TBEVToro). Averages ± s.d. from three independent experiments are shown (n = 6). n indicates the number of biologically independent replicates. Comparisons are presented between WT and LRP8-KO or LRP8-overexpressing cells, respectively; data was analysed by one-way ANOVA with Dunnett’s test: ****, P < 0.0001; ***, P = 0.0001. b, A549 WT, LRP8-KO and LRP8-KO cells complemented with LRP8 cDNA were exposed to TBEVHypr and infection levels were determined at 6 h.p.i. Averages ± s.d. from two experiments are shown (n = 4), in which n indicates the number of biologically independent samples. Comparisons are presented between WT and LRP8-KO or LRP8-overexpressing cells, respectively; data was analysed by one-way ANOVA with Dunnett’s test: ****, P < 0.0001. c, A549 WT, LRP8-KO and LRP8-overexpressing cell lines were exposed to Semliki Forest virus (SFV), Sindbis virus (SINV), Langat virus (LGTV), West Nile virus (WNV), Zika virus (ZIKV), Dengue virus type-2 (DENV-2), Japanese Encephalitis virus (JEV), and Powassan virus (POWV). Infection levels were determined at 16 h.p.i. (SFV, SINV), 24 h.p.i. (LGTV, WNV, JEV), 30 h.p.i. (ZIKV), 48 h.p.i. (POWV), and 65 h.p.i. (DENV-2). Averages ± s.d. from three [n = 12 (SFV, SINV), n = 6 (LGTV, WNV, JEV, POWV)], four (n = 7; ZIKV), or six (n = 18; DENV-2) independent experiments are shown. Comparisons are shown between WT and LRP8-KO or LRP8-overexpressing cells, respectively; data was analysed by one-way ANOVA with Dunnett’s test: ****, P < 0.0001; **, P = 0.0026; *, P = 0.0107. d (left panel), Human Huh-7.5.1-SpCas9 cells were transduced to express LRP8-targeting sgRNA and reduction in LRP8 expression was determined by western blotting using an LRP8-specific antibody. The housekeeping protein β-actin was included as a loading control. Mr, relative molecular weight (K denotes ×1,000). For gel source data, see Supplementary Information Fig. 3. (Right panel), sgRNA-treated A549 cells were exposed to TBEVNeudörfl, POWV, JEV, WNV and yellow fever virus (YFV) RVPs. Cells were scored for infection at 24 h.p.i. Averages ± s.d. are shown, n = 18 from six experiments (TBEVNeudörfl), n = 9 from three experiments (POWV), n = 6 from two experiments (YFV, JEV, WNV). Comparisons are shown between control and sgRNA-treated cells and were analysed by one-way ANOVA with Dunnett’s test: ****, P < 0.0001.

Source Data

Extended Data Fig. 4 Expression of LRP8 variants in A549 cells and expression, purification and characterization of soluble LRP8 and PCDH1-EC1 proteins.

a, A549 cells expressing endogenous levels of LRP8 (WT) or cells overexpressing the indicated Flag-tagged LRP8 variants were immunostained with an anti-Flag mAb and visualized by fluorescence microscopy. Experiments were performed two times with similar results. Scale bar, 20 µm. b, A549 cells expressing endogenous levels of LRP8 (WT) or cells overexpressing the indicated Flag-tagged LRP8 variants were lysed and LRP8 expression levels were visualized by western blotting. The housekeeping protein β-actin was included as a loading control. Western blot images from one experiment representative of three independent experiments are shown. Mr, relative molecular weight (K denotes ×1,000). For gel source data, see Supplementary Information Fig. 3. c, Purified soluble LRP8-LA1-2, LRP8-LA1 and PCDH1-EC1 C-terminally fused to human IgG1 Fc tags were resolved on an SDS-polyacrylamide gel under non-reducing (- dithiothreitol, DTT) or reducing (+ DTT) conditions and visualized by Coomassie blue staining. Experiments were performed three times with similar results. Mr, relative molecular weight (K denotes ×1,000). For gel source data, see Supplementary Information Fig. 3. d, Analytical size-exclusion chromatography (SEC) of proteins from c. Absorbance (milli-absorbance units, mAu) was monitored at 280 nm.

Source Data

Extended Data Fig. 5 Mapping of LRP8:E binding.

a, TBEV RVPs were added to mAb 4G2-coated plates and capture of the indicated human IgG1 proteins was measured by ELISA. A450nm, absorbance at 450 nm. Averages ± s.d. are shown from three independent experiments, n = 6, where n indicates the number of biologically independent samples. Comparisons are between EC1-Fc and LRP8-LA1-Fc or LRP8-LA1-2-Fc, respectively and were analysed by two-way ANOVA with Tukey’s correction: ****, P < 0.0001; ***, P = 0.006; *, P = 0.0427. b, Sensorgrams of binding of TBEV RVPs (quantified based on TBEV E [nM]) to sensors coated with EC1-Fc as measured by BLI. Data presented are from one representative measurement of two independent experiments (n = 2). c, Sensorgrams of binding of JEV RVPs (quantified based on JEV E [nM]) to sensors coated with LRP8-LA1-2-Fc as measured by BLI. Data presented are from one representative measurement of two independent experiments (n = 2). d, Monomeric purified TBEVNeudörfl E domain III (DIII) bearing a C-terminal hexa-histidine tag was resolved on an SDS-polyacrylamide gel (under nonreducing and reducing conditions [-/+ DTT]) and visualized by Coomassie blue staining. Experiment was performed three times with similar results. Mr, relative molecular weight (K denotes ×1,000). e, TBEVNeudörfl E DIII was captured onto anti-His mAb-coated plates and the binding of decreasing amounts of LRP8-LA1-2-Fc and -LA1-Fc was determined by ELISA. A450nm, absorbance at 450 nm. Averages ± s.d. are shown from three independent experiments, n = 6, where n indicates the number of biologically independent samples. f, LRP8-LA-1-2-Fc was coated onto plates and capture of TBEV soluble E protein (sE) was measured by ELISA. A450nm, absorbance at 450 nm. Averages ± s.d. are shown from two independent experiments, n = 4. g, TBEV sE was incubated with LRP8-LA1-2-Fc and subsequently captured with Protein G magnetic beads. Co-immunoprecipitated protein complexes and a fraction of the input material were analysed by western blot. Data from one experiment representative of two independent experiments (n = 2) are shown. Mr, relative molecular weight (K denotes ×1,000). h, TBEV and POWV-I E DIII bearing a C-terminal COMP pentamerization domain followed by a hexa-histidine tag were resolved on an SDS-polyacrylamide gel under nonreducing and reducing conditions (-/+ DTT) and visualized by Coomassie blue staining. Experiment was performed three times with similar results. Mr, relative molecular weight (K denotes ×1,000). i, Analytical SEC of COMP-tagged TBEV E DIII. Absorbance (mAu) was monitored at 280 nm. j, TBEV and POWV-I E DIII C-terminally fused to a COMP domain were captured onto anti-His mAb-coated plates and the binding of decreasing amounts of LRP8-LA1-2-Fc was determined by ELISA. A450nm, absorbance at 450 nm. Averages ± s.d. are shown from two independent experiments, n = 5. k, Binding of EC1-Fc to TBEV E DIII-COMP as measured by microscale thermophoresis (MST). Results from one experiment (shown as mean ± s.e.m. of three technical replicates) representative of two independent experiments are shown (n = 2). For gel source data, see Supplementary Information Fig. 3.

Source Data

Extended Data Fig. 6 LRP8 boosts TBEV E-mediated attachment to host cells.

a, A549 WT, LRP8-KO and cells overexpressing LRP8 variants (LA1-7, LA1-3, LA1-2, and LA1) were exposed to TBEVSofjin (MOI, 10 IU) at 4 °C. Cells were immunostained for TBEV E, plasma membranes were labelled with wheat germ agglutinin, and cells were imaged by fluorescent confocal microscopy. Images from one experiment representative of >10 randomly captured images of two independent experiments (n = 2) are shown. Scale bar, 50 μm. b (left panel), A549 WT, LRP8-KO and LRP8-overexpressing cells were exposed to rLGTVTBEV-93/783 (MOI, 0.5 IU) at 4 °C and the amounts of bound viral particles were determined by western blotting. GAPDH was included as a loading control. Data from one experiment representative of four independent experiments (n = 4) is shown. Mr, relative molecular weight (K denotes ×1,000). For gel source data, see Supplementary Information Fig. 4. (Right panel), E signal was quantified on a ChemiDoc XRS+ Imaging System using the onboard software. Averages ± s.d. are shown from four experiments, n = 4. WT versus LRP8-KO or LRP8-overexpressing cells, one-way ANOVA with Dunnett’s test: ****, P < 0.0001; ***, P = 0.0003; *, P = 0.0140. c, A549 LRP8-overexpressing cells were exposed to TBEVSofjin (MOI, 10 IU) at 37 °C. Cells were immunostained for TBEV E and the early endosomal marker EEA1 and imaged by fluorescent confocal microscopy. Arrowheads indicate colocalization of TBEV E and EEA1. Images from one experiment representative of >20 randomly captured images of two independent experiments (n = 2) are shown. Scale bar, 50 μm.

Source Data

Extended Data Fig. 7 LRP8 selectively inhibits TBEV E-mediated infection.

a, Infection of Huh-7.5.1 cells with single-cycle JEV RVPs after pre-incubation with increasing amounts of LRP8-LA1-2-Fc, LRP8-LA1-Fc, or human IgG isotype control. Averages ± s.d. are shown from three independent experiments, n = 9, where n indicates the number of biologically independent samples. b, Infection of Huh-7.5.1 cells with TBEVHypr after pre-incubation with increasing amounts of LRP8-LA1-2-Fc. Averages ± s.d. are shown from three experiments, n = 6, where n indicates the number of biologically independent replicates. c, Infection of U-87 MG glioblastoma cells with TBEVSofjin after pre-incubation with decreasing amounts of LRP8-LA1-2-Fc. Averages ± s.d. are shown from three experiments, n = 6. n indicates the number of biologically independent samples. Comparisons are shown between cells infected with untreated and LRP8-incubated virus and were analysed by one-way ANOVA with Dunnett’s test: ****, P < 0.0001.

Source Data

Extended Data Fig. 8 Characterization of neural progenitor cells and TBEV infection of neuronal cell cultures differentiated for 60 days.

a, Neural progenitor cells (NPCs) were immunostained for SOX2, NESTIN, GFAP, OLIG2, and PDGFRα and their cellular distribution was visualized by fluorescence microscopy. The number of NPCs expressing the indicated marker proteins was quantified manually or using ImageJ and a custom analysis pipeline in CellProfiler. Pooled data from one experiment is shown (n  ≥  400 individual cells per marker protein). Arrowheads indicate GFAP+ cells. Scale bar, 100 μm. b, Neural cells were differentiated in culture for 60 days, immunostained for the neuronal markers NeuN, MAP2, PSD-95, βIII-tubulin, LRP8 and GFAP, and their cellular distribution was visualized by fluorescence microscopy. Arrowheads indicate GFAP+ cells. Representative images of one experiment per condition are shown. Scale bar, 100 μm. c, The number of neural cell bodies (6.1 μm extension from the perimeter of the nuclei) from cells characterized in b and co-expressing LRP8 and βIII-tubulin or LRP8 and GFAP, respectively was quantified using ImageJ and a custom analysis pipeline in CellProfiler. Pooled data from one experiment is shown (n  =  6000 individual cells per protein combination). d (left panel), Infection of neuronal cells characterized in b with TBEVSofjin (MOI, 0.02 IU) after pre-incubation with LRP8-LA1-2-Fc or human IgG (400 nM). Cells were subjected to immunostaining followed by immunofluorescence microscopy. Images from one experiment representative of two independent experiments are shown. Scale bar, 100 μm. (Right panel), TBEV-infected neural cell bodies in fluorescent images were enumerated using ImageJ and a custom analysis pipeline in CellProfiler. Pooled data from two independent experiments is shown as averages ± s.d. [n = 29 (human IgG) and n = 23 (LRP8-LA1-2-Fc), in which n represents fluorescent images]. Comparisons are presented between cells infected with human IgG- or LRP8-LA1-2-Fc-treated TBEV, respectively; data was analysed by two-tailed unpaired t-test: ****, P < 0.0001.

Source Data

Extended Data Fig. 9 Murine LRP8 ortholog boosts TBEV infection.

a, Amino-acid sequence alignment of LRP8’s ligand-binding domain (LBD) of human (HsLRP8) and murine (MmLRP8) orthologs is shown. Signal sequences (HsLRP8: aa 1–32; MmLRP8: aa 1–26) are not shown, and residue numbering reflects the mature sequences. The LA repeats are marked. Orange shading, divergent residues. b, Overexpression of murine and human LRP8 orthologs in A549 WT cells was determined via western blotting. The housekeeping protein β-actin was included as a loading control. Experiment was performed two times with similar results. Mr, relative molecular weight (K denotes ×1,000). For gel source data, see Supplementary Information Fig. 4. c, A549 WT and LRP8-overexpressing cell lines were exposed to TBEV RVPs and infection levels were determined at 24 hpi. Averages ± s.d. from four independent experiments are shown (n = 12; n indicates the number of biologically independent samples). Comparisons are presented between WT and LRP8-overexpressing cells, respectively; data was analysed by one-way ANOVA with Dunnett’s test: ****, P < 0.0001.

Source Data

Extended Data Fig. 10 Weight changes, clinical scores and in situ hybridization scores of the TBEV-challenged mice study cohorts.

a, C57BL/6 mice were challenged with TBEVHypr (1,000 PFU, i.p.) pre-complexed with LRP8-LA1-2-Fc, EC1-Fc or vehicle (PBS) and monitored daily for weight loss for 28 days. Averages from two independent experiments are shown and data are represented as mean ± s.d., n = 20 mice per group. b, Clinical scores of challenged animals in a were monitored for 28 days. Averages from two experiments are shown, n = 20 mice per group. c, C57BL/6 mice were administered LRP8-LA1-2-Fc, EC1-Fc or vehicle 6 h prior to TBEVHypr challenge (1,000 PFU, i.p.) and were subsequently monitored daily for weight loss for 28 days. Averages from two independent experiments are shown and data are represented as mean ± s.d., n = 19 mice (EC1-Fc) and n = 20 mice (vehicle, LRP8-LA1-2-Fc). d, Clinical scores of challenged animals in c were monitored for 28 days. Averages from two experiments are shown, n = 19 mice (EC1-Fc) and n = 20 mice (vehicle, LRP8-LA1-2-Fc). e, In situ hybridization (ISH) scores from challenged animals in a. On day 6 post TBEVHypr challenge, 5 animals per treatment group were sacrificed, brain tissues harvested and subjected to ISH. TBEV RNA level scoring: +++, RNA-positive cells frequently detected; ++, RNA-positive cells occasionally detected; +, RNA-positive cells rarely detected; -, no RNA-positive cells detected. f, ISH scores from challenged animals in c. Mice brain tissues were harvested, prepared for ISH and scored as described in e.

Source Data

Supplementary information

Supplementary Figs. 1–4

Supplementary Figs. 1–4.

Reporting Summary

Supplementary Table 1

CRISPR–Cas9 screen: MAGeCK data analysis from Fig. 1a.

Supplementary Table 2

CRISPR–Cas9 screen: sgRNA summary from Fig. 1a.

Supplementary Table 3

Analysis of rank uniqueness of top hits in CRISPR–Cas9 screen from Extended Data Fig. 1.

Peer Review File

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

Mittler, E., Tse, A.L., Tran, PTH. et al. LRP8 is a receptor for tick-borne encephalitis virus. Nature (2025). https://doi.org/10.1038/s41586-025-09500-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41586-025-09500-2

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