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.

  • Letter
  • Published:

Exosome mimicry by a HAVCR1–NPC1 pathway of endosomal fusion mediates hepatitis A virus infection

Abstract

Cell-to-cell communication by exosomes controls normal and pathogenic processes1,2. Viruses can spread in exosomes and thereby avoid immune recognition3. While biogenesis, binding and uptake of exosomes are well characterized4,5, delivery of exosome cargo into the cytoplasm is poorly understood3. We report that the phosphatidylserine receptor HAVCR1 (refs. 6,7) and the cholesterol transporter NPC1 (ref. 8) participate in cargo delivery from exosomes of hepatitis A virus (HAV)-infected cells (exo-HAV) by clathrin-mediated endocytosis. Using CRISPR–Cas9 knockout technology, we show that these two lipid receptors, which interact in the late endosome9, are necessary for the membrane fusion and delivery of RNA from exo-HAV into the cytoplasm. The HAVCR1–NPC1 pathway, which Ebola virus exploits to infect cells9, mediates HAV infection by exo-HAV, which indicates that viral infection via this exosome mimicry mechanism does not require an envelope glycoprotein. The capsid-free viral RNA in the exosome lumen, but not the endosomal uncoating of HAV particles contained in the exosomes, is mainly responsible for exo-HAV infectivity as assessed by methylene blue inactivation of non-encapsidated RNA. In contrast to exo-HAV, infectivity of HAV particles is pH-independent and requires HAVCR1 or another as yet unidentified receptor(s) but not NPC1. Our findings show that envelope-glycoprotein-independent fusion mechanisms are shared by exosomes and viruses, and call for a reassessment of the role of envelope glycoproteins in infection.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Exosomes require HAVCR1 for cargo delivery.
Fig. 2: Cargo delivery of free HAV RNA from the lumen of exo-HAV RNA into the cytoplasm.
Fig. 3: NPC1 is required for exosome cargo delivery.
Fig. 4: Exosome cell entry by CME and cargo delivery via the HAVCR1–NPC1 pathway.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study are available from the corresponding author upon reasonable request. Numerical and statistical source data that underlie the graphs in figures, extended data and supplemental data are provided with the paper.

References

  1. Raposo, G. & Stoorvogel, W. Extracellular vesicles: exosomes, microvesicles, and friends. J. Cell Biol. 200, 373–383 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Pitt, J. M., Kroemer, G. & Zitvogel, L. Extracellular vesicles: masters of intercellular communication and potential clinical interventions. J. Clin. Invest. 126, 1139–1143 (2016).

    PubMed  PubMed Central  Google Scholar 

  3. van Dongen, H. M., Masoumi, N., Witwer, K. W. & Pegtel, D. M. Extracellular vesicles exploit viral entry routes for cargo delivery. Microbiol. Mol. Biol. Rev. 80, 369–386 (2016).

    PubMed  PubMed Central  Google Scholar 

  4. French, K. C., Antonyak, M. A. & Cerione, R. A. Extracellular vesicle docking at the cellular port: extracellular vesicle binding and uptake. Semin. Cell Dev. Biol. 67, 48–55 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Hessvik, N. P. & Llorente, A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 75, 193–208 (2018).

    CAS  PubMed  Google Scholar 

  6. Kaplan, G. et al. Identification of a surface glycoprotein on African green monkey kidney cells as a receptor for hepatitis A virus. EMBO J. 15, 4282–4296 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Kobayashi, N. et al. TIM-1 and TIM-4 glycoproteins bind phosphatidylserine and mediate uptake of apoptotic cells. Immunity 27, 927–940 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Carstea, E. D. et al. Niemann–Pick C1 disease gene: homology to mediators of cholesterol homeostasis. Science 277, 228–231 (1997).

    CAS  PubMed  Google Scholar 

  9. Kuroda, M. et al. Interaction between TIM-1 and NPC1 is important for cellular entry of Ebola virus. J. Virol. 89, 6481–6493 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. van Niel, G., D’Angelo, G. & Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 19, 213–228 (2018).

    PubMed  Google Scholar 

  11. Kaplan, G. G. et al. in Viral Hepatitis (eds Thomas, H. C. et al.) 29–42 (Wiley–Blackwell, 2013).

  12. Feng, Z. et al. A pathogenic picornavirus acquires an envelope by hijacking cellular membranes. Nature 496, 367–371 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. McKnight, K. L. et al. Protein composition of the hepatitis A virus quasi-envelope. Proc. Natl Acad. Sci. USA 114, 6587–6592 (2017).

    CAS  PubMed  Google Scholar 

  14. Feng, D. et al. Cellular internalization of exosomes occurs through phagocytosis. Traffic 11, 675–687 (2010).

    CAS  PubMed  Google Scholar 

  15. Tian, T. et al. Exosome uptake through clathrin-mediated endocytosis and macropinocytosis and mediating miR-21 delivery. J. Biol. Chem. 289, 22258–22267 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. White, J. M. & Whittaker, G. R. Fusion of enveloped viruses in endosomes. Traffic 17, 593–614 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Feigelstock, D., Thompson, P., Mattoo, P., Zhang, Y. & Kaplan, G. G. The human homolog of HAVcr-1 codes for a hepatitis A virus cellular receptor. J. Virol. 72, 6621–6628 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Wei, X. et al. Surface phosphatidylserine is responsible for the internalization on microvesicles derived from hypoxia-induced human bone marrow mesenchymal stem cells into human endothelial cells. PLoS ONE 11, e0147360 (2016).

    PubMed  PubMed Central  Google Scholar 

  19. Feigelstock, D., Thompson, P., Mattoo, P. & Kaplan, G. G. Polymorphisms of the hepatitis A virus cellular receptor 1 in African green monkey kidney cells result in antigenic variants that do not react with protective monoclonal antibody 190/4. J. Virol. 72, 6218–6222 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Costafreda, M. I. & Kaplan, G. HAVCR1 (CD365) and its mouse ortholog are functional hepatitis A virus (HAV) cellular receptors that mediate HAV infection. J. Virol. 92, e02065-17 (2018).

    PubMed  PubMed Central  Google Scholar 

  21. Kachko, A. et al. Determinants in the Ig variable domain of human HAVCR1 (TIM-1) are required to enhance hepatitis C virus entry. J. Virol. 92, e01742-17 (2018).

    PubMed  PubMed Central  Google Scholar 

  22. Konduru, K. & Kaplan, G. G. Stable growth of wild-type hepatitis A virus in cell culture. J. Virol. 80, 1352–1360 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Lötvall, J. et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 3, 26913 (2014).

    PubMed  Google Scholar 

  24. Manangeeswaran, M. et al. Binding of hepatitis A virus to its cellular receptor 1 inhibits T-regulatory cell functions in humans. Gastroenterology 142, 1516–1525 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Quijada, N. M., Fongaro, G., Barardi, C. R. M., Hernández, M. & Rodríguez-Lázaro, D. Propidium monoazide integrated with qPCR enables the detection and enumeration of infectious enteric RNA and DNA viruses in clam and fermented sausages. Front. Microbiol. 7, 2008 (2016).

    PubMed  PubMed Central  Google Scholar 

  26. Squillace, D. M., Zhao, Z., Call, G. M., Gao, J. & Yao, J. Q. Viral inactivation of human osteochondral grafts with methylene blue and light. Cartilage 5, 28–36 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Higgins, M. E., Davies, J. P., Chen, F. W. & Ioannou, Y. A. Niemann–Pick C1 is a late endosome-resident protein that transiently associates with lysosomes and the trans-Golgi network. Mol. Genet. Metab. 68, 1–13 (1999).

    CAS  PubMed  Google Scholar 

  28. Carette, J. E. et al. Ebola virus entry requires the cholesterol transporter Niemann–Pick C1. Nature 477, 340–343 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Cote, M. et al. Small molecule inhibitors reveal Niemann–Pick C1 is essential for Ebola virus infection. Nature 477, 344–348 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Kwon, H. J. et al. Structure of N-terminal domain of NPC1 reveals distinct subdomains for binding and transfer of cholesterol. Cell 137, 1213–1224 (2009).

    PubMed  PubMed Central  Google Scholar 

  31. Ndungo, E. et al. A single residue in Ebola virus receptor NPC1 influences cellular host range in reptiles. mSphere 1, e00007-16 (2016).

    PubMed  PubMed Central  Google Scholar 

  32. Dahl, N. K., Reed, K. L., Daunais, M. A., Faust, J. R. & Liscum, L. Isolation and characterization of Chinese hamster ovary cells defective in the intracellular metabolism of low density lipoprotein-derived cholesterol. J. Biol. Chem. 267, 4889–4896 (1992).

    CAS  PubMed  Google Scholar 

  33. Millard, E. E. et al. The sterol-sensing domain of the Niemann–Pick C1 (NPC1) protein regulates trafficking of low density lipoprotein cholesterol. J. Biol. Chem. 280, 28581–28590 (2005).

    CAS  PubMed  Google Scholar 

  34. Ivanov, A. I. Pharmacological inhibition of endocytic pathways: is it specific enough to be useful? Methods Mol. Biol. 440, 15–33 (2008).

    CAS  PubMed  Google Scholar 

  35. Mesaki, K., Tanabe, K., Obayashi, M., Oe, N. & Takei, K. Fission of tubular endosomes triggers endosomal acidification and movement. PLoS ONE 6, e19764 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Gillespie, E. J. et al. Selective inhibitor of endosomal trafficking pathways exploited by multiple toxins and viruses. Proc. Natl Acad. Sci. USA 110, E4904–E4912 (2013).

    CAS  PubMed  Google Scholar 

  37. Dreux, M. et al. Short-range exosomal transfer of viral RNA from infected cells to plasmacytoid dendritic cells triggers innate immunity. Cell Host Microbe 12, 558–570 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Baglio, S. R. et al. Sensing of latent EBV infection through exosomal transfer of 5′pppRNA. Proc. Natl Acad. Sci. USA 113, E587–E596 (2016).

    CAS  PubMed  Google Scholar 

  39. Feng, Z. et al. Human pDCs preferentially sense enveloped hepatitis A virions. J. Clin. Invest. 125, 169–176 (2015).

    PubMed  Google Scholar 

  40. Pinto, R., Bosch, A. & Kaplan, G. in Liver Immunology: Principles and Practice (eds Gershwin, M. E. et al.) 173–189 (Springer, 2013).

  41. Sainz, B. Jr. et al. Identification of the Niemann–Pick C1-like 1 cholesterol absorption receptor as a new hepatitis C virus entry factor. Nat. Med. 18, 281–285 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Das, A. et al. TIM1 (HAVCR1) is not essential for cellular entry of either quasi-enveloped or naked hepatitis A virions. mBio 8, e00969-17 (2017).

    PubMed  PubMed Central  Google Scholar 

  43. Dveksler, G. S., Gagneten, S. E., Scanga, C. A., Cardellichio, C. B. & Holmes, K. V. Expression of the recombinant anchorless N-terminal domain of mouse hepatitis virus (MHV) receptor makes hamster of human cells susceptible to MHV infection. J. Virol. 70, 4142–4145 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Yang, S. T., Kreutzberger, A. J. B., Lee, J., Kiessling, V. & Tamm, L. K. The role of cholesterol in membrane fusion. Chem. Phys. Lipids 199, 136–143 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Totsuka, A. & Moritsugu, Y. in Viral Hepatitis and Liver Disease. Proceedings of the International Symposium on Viral Hepatitis and Liver Disease: Molecules Today, More Cures Tomorrow (eds Nishioka, K. et al.) 509–513 (Spring-Verlag Tokyo, 1994).

  46. Uphoff, C. C., Gignac, S. M. & Drexler, H. G. Mycoplasma contamination in human leukemia cell lines. I. Comparison of various detection methods. J. Immunol. Methods 149, 43–53 (1992).

    CAS  PubMed  Google Scholar 

  47. Cohen, J. I., Ticehurst, J. R., Feinstone, S. M., Rosenblum, B. & Purcell, R. H. Hepatitis A virus cDNA and its RNA transcripts are infectious in cell culture. J. Virol. 61, 3035–3039 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Konduru, K. & Kaplan, G. G. Determinants in 3Dpol modulate the rate of growth of hepatitis A virus. J. Virol. 84, 8342–8347 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Konduru, K., Virata-Theimer, M. L., Yu, M. Y. & Kaplan, G. G. A simple and rapid hepatitis A virus (HAV) titration assay based on antibiotic resistance of infected cells: evaluation of the HAV neutralization potency of human immune globulin preparations. Virol. J. 5, 155 (2008).

    PubMed  PubMed Central  Google Scholar 

  50. Whitt, M. A. Generation of VSV pseudotypes using recombinant DeltaG-VSV for studies on virus entry, identification of entry inhibitors, and immune responses to vaccines. J. Virol. Methods 169, 365–374 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Konduru, K. et al. Ebola virus glycoprotein Fc fusion protein confers protection against lethal challenge in vaccinated mice. Vaccine 29, 2968–2977 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Spouge, J. L. Statistical analysis of sparse infection data and its implications for retroviral treatment trials in primates. Proc. Natl Acad. Sci. USA 89, 7581–7585 (1992).

    CAS  PubMed  Google Scholar 

  53. Théry, C., Amigorena, S., Raposo, G. & Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. 30, 3.22.21–3.22.29 (2006).

    Google Scholar 

  54. Nunes-Correia, I. et al. Fluorescent probes for monitoring virus fusion kinetics: comparative evaluation of reliability. Biochim. Biophy. Acta 1561, 65–75 (2002).

    CAS  Google Scholar 

  55. Costafreda, M. I., Bosch, A. & Pinto, R. M. Development, evaluation, and standardization of a real-time TaqMan reverse transcription–PCR assay for quantification of hepatitis A virus in clinical and shellfish samples. Appl. Environ. Microbiol. 72, 3846–3855 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Cantin, R., Diou, J., Bélanger, D., Tremblay, A. M. & Gilbert, C. Discrimination between exosomes and HIV-1: purification of both vesicles from cell-free supernatants. J. Immunol. Methods 338, 21–30 (2008).

    CAS  PubMed  Google Scholar 

  57. Wang, X., Campos, B., Kaetzel, M. A. & Dedman, J. R. Secretion of annexin V from cultured cells requires a signal peptide. Placenta 22, 837–845 (2001).

    CAS  PubMed  Google Scholar 

  58. Ueyama, T. et al. Sequential binding of cytosolic Phox complex to phagosomes through regulated adaptor proteins: evaluation using the novel monomeric Kusabira–Green System and live imaging of phagocytosis. J. Immunol. 181, 629–640 (2008).

    CAS  PubMed  Google Scholar 

  59. Vercauteren, D. et al. The use of inhibitors to study endocytic pathways of gene carriers: optimization and pitfalls. Mol. Ther. 18, 561–569 (2010).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by funding from the US Food and Drug Administration Intramural Program to G.K. and a Medical Countermeasures Initiative to G.K. This project was supported in part by appointments to the Research Fellowship Program at the Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, administered by the Oak Ridge Institute for Science and Education through an interagency agreement between the US Department of Energy and the FDA (to M.I.C., A.A. and H.L.).

Author information

Authors and Affiliations

Authors

Contributions

M.I.C. and G.K. were responsible for the overall design of the study. M.I.C. carried out most of the experiments, and A.A. and H.L. performed the confocal microscopy studies. G.K. performed the studies with NPC1 mutants, block of infectivity by liposomes and characterization of exosomes by western blot analysis and flow cytometry. M.I.C., A.A. and G.K. analysed the data. G.K. wrote the manuscript and M.I.C helped with the editing. All authors reviewed and commented on the manuscript.

Corresponding author

Correspondence to Gerardo Kaplan.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Fusion of R18-labeled liposomes is temperature dependent and requires phosphatidylserine and cholesterol.

a, Huh7 cells were treated with PS:PC:Chl-R18 liposomes for 30 min at 37 °C, 15 °C, or 4 °C to test for liposome fusion (red). Nuclei were stained with DAPI (blue). Cells were analyzed in a LSM 700 confocal microscope. Micrographs were taken using a 40x oil objective. b, Quantitative analysis of the fusion of R18-labeled liposomes in endosomes from (a). The R18 fluorescence intensity (red) of 20 cells from (a) was measured using ImagJ software and the experiment was repeated 3 times (n = 60). c, Huh7 cells were treated with PS:PC:Chl-R18, PS:PC-R18, PC:Chl-R18, or PC-R18 during 30 min at 37 °C. Nuclei were stained with DAPI (blue). Cells were analyzed in a LSM 700 confocal microscope. Micrographs were taken with a 40x oil objective. d, Quantitative analysis of the fusion of R18-labeled liposomes in endosomes from (c). R18 fluorescence intensity (red) of 20 cells from (c) was measured using ImagJ software, and the experiment was repeated 3 times (n=60). Scale bars in (a) and (c) represent 50 mm. In (b) and (d), box and whiskers plots were done using the Tukey method. Box limits are upper and lower quartiles, center lines are the medians, whiskers are 1.5x interquartile range, and points represent outliers. P values were determined by two-sided Mann-Whitney test.

Source data

Extended Data Fig. 2 Intracellular fusion of R18-labeled liposomes is pH-dependent.

a, Liposome fusion (red) in Huh7 cells treated with 2 µM Monensin, 30 µM Chloroquine diphosphate,or 5 mM Amonium chloride (NH4Cl) prior to the addition of PS:PC:Chl-R18 liposomes. Nuclei were stained with DAPI (blue). Cells were analyzed in a LSM 700 confocal microscope. Micrographs were taken using a 40x oil objective. Scale bars represent 50 µm. b, Quantitative analysis of the fusion of R18-labeled liposomes in endosomes from (a). The R18 fluorescence intensity (red) of 20 cells from (a) was measured using ImagJ software and the experiment was repeated 3 times (n=60). Box and whiskers plot was done using the Tukey method. Box limits, upper and lower quartiles; Center line, median; whiskers, 1.5x interquartile range; points, outliers; P values between untreated and treated cells were determined by two-sided Mann-Whitney test. c, Lack of cytotoxicity of the compounds was confirmed by flow cytometry using a caspase-3 apoptosis assay based on the irreversible binding of cell-permeable inhibitor DEVD-fmk conjugated to sulfo-rhodamine (Red-DEVD-fmk) to activated caspase-3. Stained cells were analyzed with a FACSCanto II (BD Biosciences) using FlowJo v8.5 software. Apoptotic/dead Huh7 control cells were prepared by heat treatment at 55 °C for 7 min. d, Liposome fusion with intracellular membranes. Huh7 cells were treated with PS:PC:Chl-R18 liposomes (R18 liposome fusion events, red) for 15 min at 37 °C, the plasma membrane was stained with CellMask (green) for 15 min at 37 °C, and nuclei were stained with DRAQ5 (blue). Cells were analyzed in a LSM 700 confocal microscope. Micrographs were taken with 40x oil objective. Scale Bars represent 25 µm. Liposome fusion events detected by R18 fluorescence (red) that did not co-localize with plasma membrane stained with Cell Mask (green). Results are representative of 3 independent experiments.

Source data

Extended Data Fig. 3 Characterization of exosomes purified from HAV-infected cells.

a, Presence of PS at the surface of exosomes from HAV-infected cells. Purified exo-HAV/Bsd treated with (+) or without (-) biotinylated ANX5 (ANX5 –Biotin) and anti-biotin mAbs was bound to magnetic beads, washed extensively, and extracted HAV RNA was quantitated by RT-qPCR. Data are mean ± sem, n=3. P values between complete and incomplete immunomagnetic sandwiches were analyzed by one-way ANOVA with Dunnett’s post-test. b, Purification by isopycnic ultracentrifugation of supernatants of HAV.8Y- Bsd-infected Huh7 cells. Gradient collected in 25 fractions from the top. H AV RNA in fractions 5–24 was quantified by RT-qPCR showing peaks of exo-HAV (fractions 11–13, 1.08–1.10 g cm-3) and vpHAV (fraction 21–23, 1.23–1.33 g cm-3). Data are mean ± sd of RT-qPCR duplicates. c, Enrichment of exosomal markers HSP70, FLOT-1, and TSG101 but not Golgi marker GOLGA1 in exo-HAV assessed by Western blot analysis of gradient fractions 5–25 from (b). Fractions aligned with (b). Cell extracts from uninfected Huh7 cells were used as controls. Migration of molecular weight markers is shown in kDa. d, Flow cytometry analysis of cell surface expression of CD9, CD63, and CD81 on Huh7 cells using PE-labeled anti-human CD9, CD63, or CD81 mAs compared to a PE-labeled isotype control. Gating strategy in left panel. e, Flow cytometry analysis of gradient fractions 2–24 from (b) absorbed to latex-beads and stained with mAbs as in (d). f, Content of CD63 and CD81 on exo-HAV (fraction 11 from b) compared to control vpHAV (fraction 22 from b) by flow cytometry as described in (e). Gating strategy in left panel. g, Sizing of exo-HAV (fractions 11–13 from (b) by DLS analysis in a Zetasizer nano ZS90 instrument performed in 3 runs (red, blue, and green lines) of 10 measurements each. Data are representative of 3 independent experiments.

Source data

Extended Data Fig. 4 HAVCR1 is an exo-HAV cellular receptor.

a, Specific infectivity of purified exo-HAV and vpHAV from Fig. 1c in Huh7 cells was determined as the ratio between HAV RNA genome equivalents (GE) assessed by RT-qPCR and infectious particles assessed by ARTA. Data are mean ± sem, n=5 from 5 different titrations. b, Expression of HAVCR1 but not HAVCR1 N94A restored exo-HAV cell entry in Huh7 HAVCR1 KO cells. HAVCR1, HAVCR1 N94A, or vector transfectants were infected with exo-HAV or vpHAV from Fig. 1c, and tested for Bsd-resistant CFU. Data are mean ± sem, n=4 from two independent experiments with biological duplicates. c, Flow cytometry analysis of the cell surface expression of HAVCR1 (red dots) or HAVCR1DIgV/ANX5 (blue dots) compared to vector-transfected cells (grey dots) on Huh7 HAVCR1 KO cell transfectants stained with PE-labeled anti-HAVCR1 mucin mAb1750. Gates contain cells expressing the HAVCR1 mucin 1750 epitope. Data representative of 4 independent experiments. d, Huh7 HAVCR1 KO cell transfectants expressing HAVCR1 or HAVCR1DIgV/ANX5 bind apoptotic cells. Monolayers of vector, HAVCR1, or HAVCR1DIgV/ANX5 cell transfectants or control Huh7 parental cells were incubated with CMFDA-labeled apoptotic Jurkat cells and washed extensively. Phase contrast (DIC) and green fluorescence (CMFDA) micrographs were taken with an Axiovert 200 fluorescence microscope using a 20x objective. Scale bar represents 50 mm. Data representative of 3 independent experiments. e, Expression of an annexin V fusion protein at the cell surface restores infectivity of exo-HAV. Huh7 HAVCR1 KO cell transfectants from (c) were infected with purified exo-HAV from Fig. 1c using a m.o.i of 0.1–0.5 for 48 h at 37oC. Total RNA was extracted from the cells and HAV RNA was quantitated by HAV RT-qPCR. Data are mean±sem, n=4 from 2 independent experiments with 2 biological replicates. P values were determined by two sided Mann-Whitney test.

Source data

Extended Data Fig. 5 Knock out of NPC1 in Huh7 cells blocks EBOV cell entry and endosomal liposome fusion.

a, Growth of rVSV-EBOVgp-GFP in Huh7 parental and NPC1 KO cells. Huh7 parental and NPC1 KO cells were infected with rVSV-EBOVgp-GFP for 24 h. At different times p.i., GFP fluorescence was assessed using a fluorescence plate reader. Data are mean ± sem, n=4 from 4 independent experiments. P values between Huh7 parental and N PC1 KO cells were determined by two-sided Mann-Whitney test. b, Quan titative analysis of the fusion of R18-labeled liposomes in endosomes. Huh7 parental, HAVCR1 KO and NPC1 KO cells were treated with PS:PC:Chl-R18 liposomes 10 min at 15 °C and incubated or not for 30 min at 37 °C to test for liposome fusion (red). Nuclei were stained with DAPI (blue). Cells were analyzed using a LSM 700 confocal microscope (Carl Zeiss), and micrographs were taken using a 40x oil objective. Scale bar represents 50 mm. c, Quantitative analysis of the fusion of R18-labeled liposomes in endosomes from (b). The R18 fluorescence intensity (red) of 25 cells was measured using ImagJ software, and the experiment was repeated 3 times (n=75). Box and whiskers plots were done using the Tukey method. Box limits, upper and lower quartiles; Center line, median; whiskers, 1.5x interquartile range; points, outliers; P values between mean fluorescence intensity (MFI) of Huh7 HAVCR1 KO or Huh7 NPC1 KO cells compared to Huh7 parental cells were determined by two-sided Mann-Whitney test.

Source data

Extended Data Fig. 6 Interaction of NPC1 with HAVCR1 in Huh7 NPC1 KO cell transfectants.

a, Schematic representation of bimolecular fluorescence complementation (BiFC) assay based on the complementation of the monomeric Kusabira green (mKG) protein57 168 amino acid (aa) N-terminus fragment (mKG(N)) fused to the C-terminus of NPC1 (NPC1-mKG(N)) and 51 aa C-terminus fragment (mKG(C)) fused to the C-terminus of HAVCR1 (HAVCR1-mKG(C)). The interaction of NPC1 and HAVCR1 results in BiFC of mKG9 that emits peak fluorescence at 507 nm upon excitation at 492 nm. b, Huh7 NPC1 KO cells transfected with vector, plasmid coding for NPC1-mKG(N) wild type or NPC1 mutants L175A/L176A (double mutation in NT D cholesterol binding pocket prevents function of NPC130), P202A/F203A (double mutation in NTD in the rim of the cholesterol binding pocket prevents function of NPC130), F503W (mutation in domain C increases binding of GP31), F503Y (mutation in domain C reduces infectivity of rVSV-ZEBOV GP by 2 logs31), L656F (mutation in sterol-sensing domain increases cholesterol binding, Millard 200533), G660R (Mutation transmembrane helix 3 prevents function of NPC130,32, or P691S (mutation in transmembrane domain results in defect in cholesterol uptake and trafficking33) and co-transfected or not with a plasmid coding for HAVCR1-mKG(C) for 48 h to determine HAVCR1-NPC1 interaction (green). Nuclei were stained with Hoechst 33342 (blue). Cells were analyzed in an Axiovert 200 fluorescence microscope, and micrographs were taken using a 40x objective. Scale bars represent 50 mm. Results are representative of 3 independent experiments.

Extended Data Fig. 7 Effect of NPC1 mutations and cell entry inhibitors in exo-HAV infection.

a, Huh7 NPC1 KO cells were transfected with cDNA of NPC1 WT, NPC1 mutants L173A/L176A, P202A/F203A, F503W, F503Y, L656F, G660R, or P691S, or vector and infected at 48 h p.t. with purified exo-HAV from Fig. 1c. Total RNA was extracted 4 days p.i. and analyzed by HAV RT-qPCR. Data are mean ± sem, n=3 from 3 independent experiments. P values between NPC1 WT and NPC1 mutants or vector were determined by one-way ANOVA with Dunnett’s post-test. b, Cell transfectants as in (a) were infected with rVSV-EBOVgp-GFP for 24 h, and virus in the supernatant was titrated by a fluorescence endpoint dilution assay in 96-well plates containing Huh7 monolayers. Viral titers were determined at 72 h p.i. using the ID50 program. Data are log10 TCID50/ml ± s.d., n=3. P values were determined as in (a). c, Viability of Huh7 cells treated with cell entry inhibitors for 72 h by dual-fluorescence Acridine Orange / Propidium Iodide assay. Data are mean ± sem, n=4 from two independent experiments with biological duplicates. d, Huh7 cells were treated with inhibitors for 1 h, infected with rVSV-EBOVgp-GFP, and GFP fluorescence was quantified at 16–20 h p.i. in a fluorescence plate reader. Data are mean ± sem, n=4 from two independent experiments with biological duplicates. e, Huh7 cells were pretreated with inhibitors for 1 h, infected with purified vpHAV from Fig. 1c. HAV replication was quantitated at 72 h p.i. by RT-qPCR. Data are mean ± sem, from left to right n=5, 5, 5, 3 3, and 3 biological replicates. P values between DMSO and inhibitors were determined as in (a). In (c) and (d), P values between DMSO and inhibitors were determined by two-sided Mann-Whitney test.

Source data

Extended Data Fig. 8 Chlorpromazine and Dynasore inhibit clathrin-mediated endocytosis.

a, Huh7 cells were treated with inhibitors of endocytic pathways (5 µg/ml CPZ, 80 µM Dynasore, 10 µM EGA, 40 µM EIPA, 10 µM LY294002) or DMSO prior to the addition of tetramethylrhodamine conjugate-transferrin (Tritc-Transferrin), a marker of clathrin-mediated endocytosis. Tritc-Transferrin (red fluorescence) uptake in the presence of inhibitors was analyzed using an LSM 700 confocal fluorescence microscope. Nuclei were stained with DAPI (blue fluorescence). Micrographs were taken with a 40x oil objective. Scale bars represent 50µm. b, Quantitative analysis of the endocytosis of Tritc-Transferrin from (a). The Tritc-Transferrin fluorescence intensity (red) of 16 cells was measured using ImagJ software, and the experiment was repeated 3 times (n=48). Box and whiskers plot was done using the Tukey method. Box limits, upper and lower quartiles; Center line, median; whiskers, 1.5x interquartile range; points, outliers; P values between DMSO and inhibitors were determined by two-sided Mann-Whitney test.

Source data

Extended Data Fig. 9 Dynasore but not chlorpromazine inhibit caveolae-mediated endocytosis.

a, Huh7 cells were treated with BODIPY FL C5-Lactoceramide complexed to BSA (LAC-cer), a marker of caveolae-mediated endocytosis, and treated with inhibitors of endocytosis (5 µg/ml CPZ, 80 µM Dynasore, 10 µM EGA, 40 µM EIPA, 10 µM LY294002) or DMSO as control. LAC-cer (green) uptake was analyzed using a LSM 700 confocal microscope. Nuclei were stained with DRAQ5 (blue). Micrographs were taken with a 40x oil objective. Scale bars represent 50µm. b, Quantitative analysis of the endocytosis of LAC-cer from (a). The LAC-cer fluorescence intensity (green) of 20 cells was measured using ImagJ software, and the experiment was repeated 3 times (n=60). Box and whiskers plot was done using the Tukey method. Box limits, upper and lower quartiles; Center line, median; whiskers, 1.5x interquartile range; points, outliers; P values between DMSO and inhibitors were determined by two-sided Mann-Whitney test.

Source data

Extended Data Fig. 10 Effect of endocytosis inhibitors in different cellular compartments.

Huh7 cells in 8-wells chamber slides were infected with the CellLight Bacman 2.0 reagents fused to GFP (green) or RFP (red) at a multiplicity of infection of 30 particles per cell, incubated at 37oC for 10–12 h, treated with 80 mM Dyn asore hydrate, 5mg/ml Chlorpromazine hydrochloride solution, 10 mM EGA, or a similar volume of DMSO vehicle as negative control, and incubated for additional 12–14 h at 37oC. Nuclei were stained with DRAQ5 (blue), cells were fixed with 4% PFA, coverslips mounted with ProLong Gold antifade reagent, and slides analyzed in a LSM 700 confocal microscope. Micrographs were taken using a 63X oil objective. Cells were infected with CellLight Bacmam 2.0 driving the expression of markers of: a, Mitochondria (leader sequence of E1 alpha pyruvate dehydrogenase fused to RFP); b, Lysosomes (Lamp1 fused to RFP); c, Golgi (human Golgi-resident enzyme N-acetylgalactosaminyltransferase 2 fused to RFP); d, Peroxisomes (peroxisomal C-terminal targeting sequence fused to GFP); e, Early endosomes (EE, Rab5a fused to GFP); or f, Late Endosomes (LE, Rab 7a fused to GFP). Scale Bars represent 25µm. Results are representative of 3 independent experiments.

Supplementary information

Source data

Source Data Fig. 1

Numerical and statistical source data.

Source Data Fig. 2

Numerical and statistical source data.

Source Data Fig. 3

Numerical and statistical source data.

Source Data Fig. 4

Numerical and statistical source data.

Source Data Extended Data Fig. 1

Numerical and statistical source data.

Source Data Extended Data Fig. 2

Numerical and statistical source data.

Source Data Extended Data Fig. 3a

Numerical and statistical source data.

Source Data Extended Data Fig. 3c

Unprocessed western blot.

Source Data Extended Data Fig. 4

Numerical and statistical source data.

Source Data Extended Data Fig. 5

Numerical and statistical source data.

Source Data Extended Data Fig. 7

Numerical and statistical source data.

Source Data Extended Data Fig. 8

Numerical and statistical source data.

Source Data Extended Data Fig. 9

Numerical and statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Costafreda, M.I., Abbasi, A., Lu, H. et al. Exosome mimicry by a HAVCR1–NPC1 pathway of endosomal fusion mediates hepatitis A virus infection. Nat Microbiol 5, 1096–1106 (2020). https://doi.org/10.1038/s41564-020-0740-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-0740-y

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