Abstract
Although coronaviruses use diverse receptors, the characterization of coronaviruses with unknown receptors has been impeded by a lack of infection models1,2. Here we introduce a strategy to engineer functional customized viral receptors (CVRs). The modular design relies on building artificial receptor scaffolds comprising various modules and generating specific virus-binding domains. We identify key factors for CVRs to functionally mimic native receptors by facilitating spike proteolytic cleavage, membrane fusion, pseudovirus entry and propagation for various coronaviruses. We delineate functional SARS-CoV-2 spike receptor-binding sites for CVR design and reveal the mechanism of cell entry promoted by the N-terminal domain-targeting S2L20-CVR. We generated CVR-expressing cells for 12 representative coronaviruses from 6 subgenera, most of which lack known receptors, and show that a pan-sarbecovirus CVR supports propagation of a propagation-competent HKU3 pseudovirus and of authentic RsHuB2019A3. Using an HKU5-specific CVR, we successfully rescued wild-type and ZsGreen-HiBiT-incorporated HKU5-1 (LMH03f) and isolated a HKU5 strain from bat samples. Our study demonstrates the potential of the CVR strategy for establishing native receptor-independent infection models, providing a tool for studying viruses that lack known susceptible target cells.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 51 print issues and online access
$199.00 per year
only $3.90 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout




Similar content being viewed by others
Data availability
The cryo-EM maps and models have been deposited to the Electron Microscopy Data Bank and Protein Data Bank (PDB) with accession numbers EMD-45174 and 9C44 (global refinement of the S2L20-bound spike trimer), and EMD-45175 and 9C45 (local refinement of the NTD and S2L20 Fab variable regions). The accession numbers (NCBI GenBank or GISAID) and protein sequence information of receptor, virus, antibodies, domains and reporter genes are provided in the Methods and supplementary tables. All other data supporting the findings of this study are available with the Article and the Supplementary Information. All reagents generated in this study are available from H.Y. with a completed materials transfer agreement. Source data are provided with this paper.
References
Millet, J. K., Jaimes, J. A. & Whittaker, G. R. Molecular diversity of coronavirus host cell entry receptors. FEMS Microbiol. Rev. 45, fuaa057 (2021).
Li, F. Receptor recognition mechanisms of coronaviruses: a decade of structural studies. J. Virol. 89, 1954–1964 (2015).
Guo, H. et al. Isolation of ACE2-dependent and -independent sarbecoviruses from Chinese horseshoe bats. J. Virol. 97, e0039523 (2023).
Cui, J., Li, F. & Shi, Z.-L. Origin and evolution of pathogenic coronaviruses. Nat. Rev. Microbiol. 17, 181–192 (2019).
Walls, A. C. et al. Cryo-electron microscopy structure of a coronavirus spike glycoprotein trimer. Nature 531, 114–117 (2016).
Walls, A. C. et al. Tectonic conformational changes of a coronavirus spike glycoprotein promote membrane fusion. Proc. Natl Acad. Sci. USA 114, 11157–11162 (2017).
Walls, A. C. et al. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 181, 281–292.e6 (2020).
Peng, G. et al. Crystal structure of mouse coronavirus receptor-binding domain complexed with its murine receptor. Proc. Natl Acad. Sci. USA 108, 10696–10701 (2011).
Yan, R. et al. Structural basis for the different states of the spike protein of SARS-CoV-2 in complex with ACE2. Cell Res. 31, 717–719 (2021).
Hoffmann, M. et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181, 271–280.e8 (2020).
Saunders, N. et al. TMPRSS2 is a functional receptor for human coronavirus HKU1. Nature 624, 207–214 (2023).
McCallum, M. et al. Human coronavirus HKU1 recognition of the TMPRSS2 host receptor. Cell 187, 4231–4245.e13 (2024).
Xiong, Q. et al. Close relatives of MERS-CoV in bats use ACE2 as their functional receptors. Nature 612, 748–757 (2022).
Hofmann, H. et al. Human coronavirus NL63 employs the severe acute respiratory syndrome coronavirus receptor for cellular entry. Proc. Natl Acad. Sci. USA 102, 7988–7993 (2005).
Starr, T. N. et al. ACE2 binding is an ancestral and evolvable trait of sarbecoviruses. Nature 603, 913–918 (2022).
Lee, J. et al. Broad receptor tropism and immunogenicity of a clade 3 sarbecovirus. Cell Host Microbe 31, 1961–1973.e11 (2023).
Lim, S., Zhang, M. & Chang, T. L. ACE2-independent alternative receptors for SARS-CoV-2. Viruses 14, 2535 (2022).
Baggen, J. et al. TMEM106B is a receptor mediating ACE2-independent SARS-CoV-2 cell entry. Cell 186, 3427–3442.e22 (2023).
Verheije, M. H. et al. Redirecting coronavirus to a nonnative receptor through a virus-encoded targeting adapter. J. Virol. 80, 1250–1260 (2006).
Wan, Y. et al. Molecular mechanism for antibody-dependent enhancement of coronavirus entry. J. Virol. 94, e02015–19 (2020).
Maemura, T. et al. Antibody-dependent enhancement of SARS-CoV-2 infection is mediated by the IgG receptors FcγRIIA and FcγRIIIA but does not contribute to aberrant cytokine production by macrophages. mBio 12, e0198721 (2021).
Kibria, M. G. et al. Antibody-mediated SARS-CoV-2 entry in cultured cells. EMBO Rep. 24, e57724 (2023).
Junqueira, C. et al. FcγR-mediated SARS-CoV-2 infection of monocytes activates inflammation. Nature 606, 576–584 (2022).
Cao, L. et al. De novo design of picomolar SARS-CoV-2 miniprotein inhibitors. Science 370, 426–431 (2020).
Hoffmann, M. A. G. et al. ESCRT recruitment to SARS-CoV-2 spike induces virus-like particles that improve mRNA vaccines. Cell 186, 2380–2391.e9 (2023).
Ying, T., Feng, Y., Wang, Y., Chen, W. & Dimitrov, D. S. Monomeric IgG1 Fc molecules displaying unique Fc receptor interactions that are exploitable to treat inflammation-mediated diseases. MAbs 6, 1201–1210 (2014).
Xiang, Y. et al. Versatile and multivalent nanobodies efficiently neutralize SARS-CoV-2. Science 370, 1479–1484 (2020).
Pinto, D. et al. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature 583, 290–295 (2020).
Scheid, J. F. et al. B cell genomics behind cross-neutralization of SARS-CoV-2 variants and SARS-CoV. Cell 184, 3205–3221.e24 (2021).
Tortorici, M. A. et al. Ultrapotent human antibodies protect against SARS-CoV-2 challenge via multiple mechanisms. Science 370, 950–957 (2020).
Cui, Z. et al. Structural and functional characterizations of infectivity and immune evasion of SARS-CoV-2 Omicron. Cell 185, 860–871.e13 (2022).
McCallum, M. et al. N-terminal domain antigenic mapping reveals a site of vulnerability for SARS-CoV-2. Cell 184, 2332–2347.e16 (2021).
Sun, X. et al. Neutralization mechanism of a human antibody with pan-coronavirus reactivity including SARS-CoV-2. Nat Microbiol 7, 1063–1074 (2022).
Pinto, D. et al. Broad betacoronavirus neutralization by a stem helix-specific human antibody. Science 373, 1109–1116 (2021).
Sauer, M. M. et al. Structural basis for broad coronavirus neutralization. Nat. Struct. Mol. Biol. 28, 478–486 (2021).
Low, J. S. et al. ACE2-binding exposes the SARS-CoV-2 fusion peptide to broadly neutralizing coronavirus antibodies. Science 377, 735–742 (2022).
Piccoli, L. et al. Mapping neutralizing and immunodominant sites on the SARS-CoV-2 spike receptor-binding domain by structure-guided high-resolution serology. Cell 183, 1024–1042.e21 (2020).
Walls, A. C. et al. Unexpected receptor functional mimicry elucidates activation of coronavirus fusion. Cell 176, 1026–1039.e15 (2019).
Zhang, S. et al. Loss of spike N370 glycosylation as an important evolutionary event for the enhanced infectivity of SARS-CoV-2. Cell Res. 32, 315–318 (2022).
Ou, X. et al. Host susceptibility and structural and immunological insight of S proteins of two SARS-CoV-2 closely related bat coronaviruses. Cell Discov. 9, 78 (2023).
Chi, X. et al. Comprehensive structural analysis reveals broad-spectrum neutralizing antibodies against SARS-CoV-2 Omicron variants. Cell Discov. 9, 37 (2023).
Cao, Y. et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. Nature 608, 593–602 (2022).
Chi, X. et al. A neutralizing human antibody binds to the N-terminal domain of the Spike protein of SARS-CoV-2. Science 369, 650–655 (2020).
Li, D. et al. In vitro and in vivo functions of SARS-CoV-2 infection-enhancing and neutralizing antibodies. Cell 184, 4203–4219.e32 (2021).
Beaudoin-Bussières, G. et al. A Fc-enhanced NTD-binding non-neutralizing antibody delays virus spread and synergizes with a nAb to protect mice from lethal SARS-CoV-2 infection. Cell Rep. 38, 110368 (2022).
Barnes, C. O. et al. Structures of human antibodies bound to SARS-CoV-2 spike reveal common epitopes and recurrent features of antibodies. Cell 182, 828–842.e16 (2020).
Benton, D. J. et al. Receptor binding and priming of the spike protein of SARS-CoV-2 for membrane fusion. Nature 588, 327–330 (2020).
Starr, T. N. et al. SARS-CoV-2 RBD antibodies that maximize breadth and resistance to escape. Nature 597, 97–102 (2021).
Hunt, A. C. et al. Multivalent designed proteins neutralize SARS-CoV-2 variants of concern and confer protection against infection in mice. Sci. Transl. Med. 14, eabn1252 (2022).
Hsieh, C.-L. et al. Structure-based design of prefusion-stabilized SARS-CoV-2 spikes. Science 369, 1501–1505 (2020).
McCallum, M. et al. Molecular basis of immune evasion by the Delta and Kappa SARS-CoV-2 variants. Science 374, 1621–1626 (2021).
McCallum, M. et al. SARS-CoV-2 immune evasion by the B.1.427/B.1.429 variant of concern. Science 373, 648–654 (2021).
Shang, J. et al. Structure of mouse coronavirus spike protein complexed with receptor reveals mechanism for viral entry. PLoS Pathog. 16, e1008392 (2020).
Wang, H. et al. TMPRSS2 and glycan receptors synergistically facilitate coronavirus entry. Cell 187, 4261–4271.e17 (2024).
Pronker, M. F. et al. Sialoglycan binding triggers spike opening in a human coronavirus. Nature 624, 201–206 (2023).
Xu, J. et al. Nanobodies from camelid mice and llamas neutralize SARS-CoV-2 variants. Nature 595, 278–282 (2021).
Xia, S. et al. Inhibition of SARS-CoV-2 (previously 2019-nCoV) infection by a highly potent pan-coronavirus fusion inhibitor targeting its spike protein that harbors a high capacity to mediate membrane fusion. Cell Res. 30, 343–355 (2020).
Xia, S. et al. Structural and functional basis for pan-CoV fusion inhibitors against SARS-CoV-2 and its variants with preclinical evaluation. Signal Transduct. Target. Ther. 6, 288 (2021).
Xia, S. et al. A pan-coronavirus fusion inhibitor targeting the HR1 domain of human coronavirus spike. Sci. Adv. 5, eaav4580 (2019).
Menachery, V. D. et al. Trypsin treatment unlocks barrier for zoonotic bat coronavirus infection. J. Virol. 94, e01774–19 (2020).
Catanzaro, NJ. et al. ACE2 from Pipistrellus abramus bats is a receptor for HKU5 coronaviruses. Preprint at bioRxiv https://doi.org/10.1101/2024.03.13.584892 (2024).
Qing, E. et al. Inter-domain communication in SARS-CoV-2 spike proteins controls protease-triggered cell entry. Cell Rep. 39, 110786 (2022).
Takano, T., Kawakami, C., Yamada, S., Satoh, R. & Hohdatsu, T. Antibody-dependent enhancement occurs upon re-infection with the identical serotype virus in feline infectious peritonitis virus infection. J. Vet. Med. Sci. 70, 1315–1321 (2008).
Loo, L. et al. Fibroblast-expressed LRRC15 is a receptor for SARS-CoV-2 spike and controls antiviral and antifibrotic transcriptional programs. PLoS Biol. 21, e3001967 (2023).
Schwegmann-Weßels, C. et al. Comparison of vesicular stomatitis virus pseudotyped with the S proteins from a porcine and a human coronavirus. J. Gen. Virol. 90, 1724–1729 (2009).
Du, Y. et al. A broadly neutralizing humanized ACE2-targeting antibody against SARS-CoV-2 variants. Nat. Commun. 12, 5000 (2021).
Whitt, M. A. Generation of VSV pseudotypes using recombinant ΔG-VSV for studies on virus entry, identification of entry inhibitors, and immune responses to vaccines. J. Virol. Methods 169, 365–374 (2010).
Nie, J. et al. Quantification of SARS-CoV-2 neutralizing antibody by a pseudotyped virus-based assay. Nat. Protoc. 15, 3699–3715 (2020).
Reed, L. J. & Muench, H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 27, 493–497 (1938).
Lempp, F. A. et al. Lectins enhance SARS-CoV-2 infection and influence neutralizing antibodies. Nature 598, 342–347 (2021).
Bowen, J. E. et al. SARS-CoV-2 spike conformation determines plasma neutralizing activity elicited by a wide panel of human vaccines. Sci. Immunol. 7, eadf1421 (2022).
Russo, C. J. & Passmore, L. A. Electron microscopy: ultrastable gold substrates for electron cryomicroscopy. Science 346, 1377–1380 (2014).
Suloway, C. et al. Automated molecular microscopy: the new Leginon system. J. Struct. Biol. 151, 41–60 (2005).
Tegunov, D. & Cramer, P. Real-time cryo-electron microscopy data preprocessing with Warp. Nat. Methods 16, 1146–1152 (2019).
Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).
Punjani, A., Zhang, H. & Fleet, D. J. Non-uniform refinement: adaptive regularization improves single-particle cryo-EM reconstruction. Nat. Methods 17, 1214–1221 (2020).
Zivanov, J., Nakane, T. & Scheres, S. H. W. A Bayesian approach to beam-induced motion correction in cryo-EM single-particle analysis. IUCrJ 6, 5–17 (2019).
Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).
Zivanov, J., Nakane, T. & Scheres, S. H. W. Estimation of high-order aberrations and anisotropic magnification from cryo-EM data sets in −3.1. IUCrJ 7, 253–267 (2020).
Rosenthal, P. B. & Henderson, R. Optimal determination of particle orientation, absolute hand, and contrast loss in single-particle electron cryomicroscopy. J. Mol. Biol. 333, 721–745 (2003).
Chen, S. et al. High-resolution noise substitution to measure overfitting and validate resolution in 3D structure determination by single particle electron cryomicroscopy. Ultramicroscopy 135, 24–35 (2013).
Pettersen, E. F. et al. UCSF Chimera–a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).
Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).
Wang, R. Y.-R. et al. Automated structure refinement of macromolecular assemblies from cryo-EM maps using Rosetta. eLife 5, e17219 (2016).
Frenz, B. et al. Automatically fixing errors in glycoprotein structures with Rosetta. Structure 27, 134–139.e3 (2019).
Croll, T. I. ISOLDE: a physically realistic environment for model building into low-resolution electron-density maps. Acta Crystallogr. D 74, 519–530 (2018).
Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).
Barad, B. A. et al. EMRinger: side chain-directed model and map validation for 3D cryo-electron microscopy. Nat. Methods 12, 943–946 (2015).
Agirre, J. et al. Privateer: software for the conformational validation of carbohydrate structures. Nat. Struct. Mol. Biol. 22, 833–834 (2015).
Liebschner, D. et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr. D 75, 861–877 (2019).
Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).
Li, W. et al. Bats are natural reservoirs of SARS-like coronaviruses. Science 310, 676–679 (2005).
Hu, B. et al. Discovery of a rich gene pool of bat SARS-related coronaviruses provides new insights into the origin of SARS coronavirus. PLoS Pathog. 13, e1006698 (2017).
Ge, X.-Y. et al. Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature 503, 535–538 (2013).
Ju, X. et al. A novel cell culture system modeling the SARS-CoV-2 life cycle. PLoS Pathog. 17, e1009439 (2021).
Guo, H. et al. ACE2-independent bat sarbecovirus entry and replication in human and bat cells. mBio 13, e0256622 (2022).
Acknowledgements
The authors are grateful for the funding support from the Basic Science Center Program of the National Natural Science Foundation of China (NSFC) (32188101 to H.Y.), National Key R&D Program of China (2023YFC2605500 to Z.-L.S. and H.Y.), NSFC Excellent Young Scientists Fund (82322041 to H.Y.), other NSFC projects (32270164, 32070160 to H.Y., 323B2006 to C.-B.M. and 32300141 to H.G.), Fundamental Research Funds for the Central Universities (2042023kf0191 and 2042022kf1188 to H.Y.), and Natural Science Foundation of Hubei Province (2023AFA015 to H.Y.), and China Postdoctoral Science Foundation (2023M733708 to H.G.). This study was also supported by the National Institute of Allergy and Infectious Diseases (P01AI167966, DP1AI158186 and 75N93022C00036 to D.V.), an Investigators in the Pathogenesis of Infectious Disease Awards from the Burroughs Wellcome Fund (D.V.), the University of Washington Arnold and Mabel Beckman CryoEM center and the National Institute of Health grant S10OD032290 (to D.V.). D.V. is an Investigator of the Howard Hughes Medical Institute and the Hans Neurath Endowed Chair in Biochemistry at the University of Washington. The authors thank X.-X. Wang for providing the human sera collected post-vaccination (SARS-CoV-2 CoronaVac, Sinovac) and collected by Beijing Youan Hospital (approval number LL-2021-042-K); K. Cai for providing sera collected from Wuhan COVID-19 convalescents (identification number 2021-012-01); Z.-H. Qian for providing BANAL-20-52 related spike-expressing plasmids; L. Lu for providing EK1-related peptides; P. Zhang and B.-C. Xu for their help with the ultracentrifugation and electron microscopic analysis; Y.-J. Li for assisting in establishing the yeast cloning methods; Y. Chen for providing the MHV-A59 strain; C.-G Wu for assissting in isolating the HKU5_PaGD2014/15 stain; Q. Ding for providing the SARS-CoV-2 (ΔN-GFP) reverse genetics related plasmids; and K. Lan and the ABSL-3 facility and other core facilities of the Key Laboratory of Virology, Wuhan University.
Author information
Authors and Affiliations
Contributions
H.Y., P.L., M.-L.H., H.G. and M.M. conceived the study and designed the experiments. J.L. conducted the first experiment of this study. P.L., M.-L.H., H.G., M.M., J.-Y.S., Y.-M.C., C.-L.W., X. Yu, C.L., L.-L.S., Y.-h.S., X. Yang and J.L. established the assays and methods, designed and cloned receptor and protein constructs and conducted related experiments. P.L., M.-L.H., C.-L.W., L.-L.S., C.-B.M., Q.X., F.T. and C.L. screened the nanobodies. P.L., M.-L.H. and L.-L.S. conducted 229E and MHV authentic virus-related experiments at Wuhan University. P.L. and H.G. conducted the RsHuB2019A and HKU5 authentic virus and HKU5 negative staining for electron microscopy imaging at the Wuhan Institute of Virology. J.C. and P.L. constructed the infectious clones of HKU5 and HKU5-ZGH, respectively. M.G. conducted the SARS-CoV-2 authentic virus infection-related experiments in ABSL-III at Wuhan University. M.M. carried out cryo-EM sample preparation, data collection and processing of the S2L20-bound SARS-CoV-2 spike dataset. M.M. and D.V. built and refined the structure. M.M. and J.E.B. carried out antibody-mediated triggering assays. D.C. provided critical reagents. H.Y., Z.-L.S., D.V., P.L., M.-L.H., M.M., C.-L.W. and X. Yu analysed the data. H.Y., P.L. and M.-L.H. prepared the original draft of the manuscript. H.Y., D.V., Z.-L.S., P.L. and M.-L.H. revised the manuscript with input from all other authors. H.Y., Z.-L.S. and D.V. obtained funding to support this study. H.Y. supervised the project.
Corresponding authors
Ethics declarations
Competing interests
H.Y. has submitted a patent application to the China National Intellectual Property Administration for the utilization of artificial viral receptors and their applications.
Peer review
Peer review information
Nature thanks Shibo Jiang 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 figures and tables
Extended Data Fig. 1 Development and optimization of a modular design strategy for CVR with a type I transmembrane topology.
a, Schematic illustration of the four miniprotein-based CVRs. b, Immunofluorescence analysis of ACE2/CVRs expression and authentic SARS-CoV-2 infection in HEK293T cells stably expressing the receptors. Upper: Receptor expression examined by C-terminal fused 3×Flag tags. Lower: SARS-CoV-2 infection efficiency as indicated by intracellular N proteins at 24 hpi. Data representative of two independent authentic SARS-CoV-2 infection assays with similar results. c, Cartoon illustrating the framework of the CVRs for TM evaluation. d, Immunofluorescence analysis of the expression of the 31 CVRs in HEK293T cells by detecting the C-terminal fused 3×Flag tags. e, SARS-CoV-2 PSV entry efficiency promoted by CVRs carrying different TMs. The detailed information on the TMs is summarized in Supplementary Table 2. Data are presented as mean ± s.d. (n = 3 wells of independently infected cells), representative of two independent experiments with similar results. f, Schematic diagram showing the LCB1-based CVRs with indicated TM or TMC substitutions. g, h, SARS-CoV-2 PSV entry in HEK293T cells transiently expressing the indicated CVRs examined by RLU (g) or GFP (h). Data are presented as mean ± s.d. (n = 3 wells of independently infected cells), Statistical analysis was performed using One-way ANOVA analysis followed by Dunnett’s test. Data represented were performed in at least two independent experiments with similar results. i, Immunofluorescence analysis of the subcellular distribution of LCB1-MXRA8 TMC-based CVRs with or without EPM transiently expressed in HEK293T cells. The white dashed boxes highlight the cell surface distribution with a higher magnification. j, SARS-CoV-2 PSV entry efficiency in HEK293T cells transiently expressing CVRs with or without EPM. Data are mean ± s.d. (n = 3 wells of independently infected cells) and analyzed with unpaired two-tailed Student’s t-tests, representative of two independent experiments with similar results. Scale bars: 100 μm in b, d, h, and i. *P < 0.05, **P < 0.01, ****P < 0.0001.
Extended Data Fig. 2 Exploring factors that contribute to the receptor function of CVRs with different topologies or modules.
a, Schematic diagram showing CVRs carrying LCB1 or mNb1 displayed in either type I or type II transmembrane topology. b, Evaluation of SARS-CoV-2 or MERS-CoV PSV entry efficiency supported by the indicated CVRs with different transmembrane topologies in HEK293T cells. Data are mean ± s.d. of biological triplicates examined over three independent infection assays. Unpaired two-tailed Student’s t-tests. c, Assessment of CVR expression, SARS-CoV-2 RBD-mFc binding, and PSV entry efficiency supported by the CVRs carrying varying copies of TR23 repeats transiently expressed in HEK293T cells. Data are representative of three independent experiments. Scale bars: 100 μm. d, Schematic representation of the CVRs carrying different numbers of immunoglobulin (Ig) domains (left) or an Fc mutant with abolished dimerization ability. e, Western blot analysis of CVRs expression in HEK293T cells under either reducing or non-reducing conditions, respectively. f, Assessment of SARS-CoV-2 PSV entry efficiency in HEK293T cells transiently expressing the indicated CVRs. Data are mean ± s.d. (n = 3 wells of independently infected cells.) and analyzed by unpaired two-tailed Student’s t-tests. g, Schematic representation of the CVRs carrying different numbers of Ig-like domains (left) from mCEACAM1a. h, Western blot analysis of CVRs expression in HEK293T cells. i, SARS-CoV-2 PSV entry efficiency in HEK293T cells transiently expressing the indicated CVRs. Data are mean ± s.d. (n = 3 wells of independently infected cells). One-way ANOVA analysis followed by Dunnett’s test. j, Schematic representation of the CVRs carrying different SARS-CoV-2 RBD targeting miniproteins. k, l, Expression (k) and SARS-CoV-2 entry-supporting (l) ability of different CVRs in 293T cells. Data are mean ± s.d. (n = 3 wells of independently infected cells). One-way ANOVA analysis followed by Dunnett’s test. m, Schematic diagram showing CVRs carrying different types of VBDs, the two representative VBDs for each type are indicated. n, Immunofluorescence analyzing the expression of the indicated CVRs transiently expressed in HEK293T cells by detecting the C-terminal fused 3×Flag tags, and the SARS-CoV-2 RBD binding. Scale bars: 100 μm. o, PSV entry are supported by indicated CVRs transiently expressed in HEK293T cells. Data are mean ± s.d. (n = 3 wells of independently infected cells). Data representative of two independent transfections, expression verification, and infection assays with similar results for d-f, g-i, j-l, and m-o, respectively. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; NS, Not significant (P > 0.05).
Extended Data Fig. 3 Investigating receptor function of CVRs with VBDs connected in various ways.
a-c, Illustration (a), RBD binding efficiency (b), and PSV entry-supporting efficiency (c) of a SARS-CoV-2/MERS-CoV bi-specific CVR transiently expressed in HEK293T cells. Data are presented as mean ± s.d. (biological triplicates of infected cells), representative of three independent experiments with similar results. Unpaired two-tailed Student’s t-test. d-f, Illustration (d), expression (e), and PSV entry-supporting efficiencies (f) of CVRs carrying single or trimeric VBD. Data are mean ± s.d. (biological triplicates of infected cells), analyzed by unpaired two-tailed Student’s t-test. Representative of two independent experiments. g-i, Schematic illustration of bispecific adapter protein (g) and MERS-CoV PSV entry efficiency in BHK-21-hACE2 cells in the presence of indicated concentrations of adapter proteins (h11B11-mNb1) throughout the infection. Entry efficiency is examined by GFP intensity (h) or RLU (i). Data are mean ± s.d. (biological triplicates of infected cells), representative of three independent infection assays. j-l, Schematic illustration of FcγR (CD32a) mediated antibody-dependent coronavirus entry (j). CD32a expression, antibody (CB6) binding (k), and SARS-CoV-2 PSV entry (l) into HEK293T-CD32a cells pretreated with indicated concentration (con.) of the CB6 antibodies. Data are mean ± s.d. (n = 4 wells of biologically independent cells), examined over two independent experiments. m-o, Entry of pre-attached PSV promoted by soluble neutralizing antibody (Nb27-hFc) or bi-specific neutralizing antibody with membrane-associating ability (Nb27-hFc-h11B11). Schematic illustration (m), Nb27-hFc promoted entry (n), and Nb27-hFc-h11B11 promoted PSV entry (o) in Caco2 cells with virus pre-attachment by 1500 rpm centrifugation at 4 °C for 1 h. Data are mean ± s.d. (biological triplicates of infected cells). Data are representative of three independent infection assays. Scale bars: 100 μm for b, e, and k, and 200 μm for h. One-way ANOVA analysis followed by Dunnett’s test for i, l, n, and o. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; NS, Not significant (P > 0.05).
Extended Data Fig. 4 Comparing receptor function of CVRs with native receptors, or alternative receptors/coreceptors in different cell types.
a-c, The ability of CVRs to promote cell-cell membrane fusion, and authentic SARS-CoV-2 infection is comparable to their native receptors. Spike and receptor-mediated cell-cell fusion was demonstrated by reconstituted GFP intensity (a) and relative light unit (RLU) of Renilla luciferase activity (b). Authentic SARS-CoV-2 infection was examined by immunostaining of intracellular N proteins at 24 hpi (c). Data are mean ± s.d. (biological triplicates) for b. Data are representative of two independent fusion assays or infection assays with similar results. d, Receptor specificity of different coronavirus PSVs in HEK293T stably expressing the native receptor or the indicated CVRs. e, SARS-CoV-2 and MERS-CoV PSV entry into various cell types expressing the indicated receptors. Data representative of two independent transfection and infection assays with similar results for d and e. f, SARS-CoV-2 PSV entry efficiencies in HEK293T cells expressing different receptors or entry factors. Data are presented as mean ± s.d. (n = 3 wells of independently infected cells.), representative of two independent transfection and infection experiments. **P < 0.01, ***P < 0.001, ****P < 0.0001; NS, Not significant (P > 0.05).
Extended Data Fig. 5 Relationship between the binding affinity, neutralizing activity, and CVR entry-promoting efficiency of 25 SARS-CoV-2 RBD-targeting nanobody-fused receptors.
a, b, Assessment of the entry-promoting ability of 25 nanobody-CVRs in HEK293T cells, indicated by GFP (a) and the RLU (b), respectively. Data are presented as mean ± s.d. (n = 3 wells of independently infected cells), representative of two independent experiments. One-way ANOVA analysis followed by Dunnett’s test. Scale bars: 200 μm. c, Comparing RBD binding, neutralization, and PSV entry-promoting ability of different nanobody-fused proteins in HEK293T cells. RBD-mFc binding and PSV entry assays were conducted in HEK293T transiently expressing the 25 nanobody-CVRs. The SARS-CoV-2 PSV neutralization assay was performed in HEK293T-ACE2 in the presence of indicated nanobody-Fc recombinant proteins (10 μg ml−1). Data are representative results of two independent experiments with similar results and plotted by the mean (n = 3 wells of independently infected/bound cells). ***P < 0.001, ****P < 0.0001; NS, Not significant (P > 0.05).
Extended Data Fig. 6 Expression and antigen-binding ability of CVRs targeting distinct SARS-CoV-2 neutralizing epitopes.
a, Western blot analysis of the expression levels of indicated scFv-CVRs transiently expressed in HEK293T cells. Data are representative of two independent experiments. b, Binding of SARS-CoV-2 S-trimer to HEK293T cells expressing the indicated CVRs. Data are representative of three assays using independent preparations of proteins. c, Flow cytometry analysis of the binding efficiency of scFv-mFc with HEK293T cells transiently expressing the SARS-CoV-2 Spike proteins and ZsGreen simultaneously. The ZsGreen positive cells were gated for subsequent analysis of mFc binding efficiency. Data representative from a single experiment with mean values (n = 3 wells of biologically independent cells) indicated. d, Trypsin-mediated S2′ cleavage of SARS2-CoV-2 PSV in the presence of soluble receptors or CB6-scFv-mFc. The concentrated SARS-CoV-2 PSV particles were incubated with 100 μg ml−1 of soluble receptors or CB6-scFv-mFc for 1 h, followed by incubation with the indicated concentration of TPCK-treated trypsin for 30 min. Western blot analysis was conducted by detecting the S2P6 epitope in the S2 subunit. Data are representative of three independent assays with similar results.
Extended Data Fig. 7 Molecular basis of NTD-mediated coronavirus entry.
a-c, Package efficiency of PSVs carrying indicated sarbecoviruses spike glycoproteins (a) and the indicated mutants (b, c). Western blot was conducted by detecting the conserved S2P6 epitope. VSV-M serves as a loading control. Blots representative of two independent transfection assays for pseudovirus production. d, Structures of SARS-CoV-2 BA.4/5 spike trimer without antibody binding (left), or in complex with S2L20 (right). Dashed boxes highlighted the N370-glycan spatially proximate to the S2L20. e, Heatmap showing the inhibitory efficacy of indicated SARS-CoV-2 neutralizing antibodies against PSV entry in HEK293T-hACE2 or HEK293T-S2L20, with BSA as a control. Data are representative results of two independent neutralization assays and plotted by the mean (n = 3 wells of independently infected cells). f, Structures of SARS-CoV-2 BA.2 spike trimers with (upper) or without (lower) the binding of NTD-targeting 4A8, along with the side-view (top) and top-view (bottom) cryoEM structures of SARS-CoV-2 Wuhan-Hu-1 spike trimers with the binding of NTD-targeting CV3-13 and DH1052. Orange: NTD; Green: CTD; Red: S2L20; Magenta: 4A8; Blue: CV3-13; Pink: DH1052. g, CryoEM data processing workflow and validation of the S2L20-bound SARS-CoV-2 S CryoEM structure. h, Negative stain microscopy of prefusion SARS-CoV-2 S-glycoprotein (without stabilizing proline substitutions) incubated without antibody, or with S2H14, S2X28, or S2L20 as indicated. S2H14 is known to promote the transition to the postfusion state and was used as a control37. Scale bars: 10 nm. Data representative of images captured from two independent experiments with similar results. i, Top-view and side-view CryoEM structures depicting soluble mCEACAM1a (cyan) in complex with MHV spike trimer (gray). NTD and CTD of MHV S are indicated in orange and green, respectively.
Extended Data Fig. 8 Generation and characterization of VBDs used for CVRs customized for various coronaviruses.
a, Workflow demonstrating the customization of nanobody-based CVRs for specific coronaviruses. b, Coronavirus CTD or S1 binding in HEK293T cells transiently expressing the corresponding CVRs. Dashed lines indicate thresholds for positive ratio calculation. Data are presented as mean ± s.d. (n = 3 wells of biologically independent cells), representative of two independent experiments. c, The pan-sarbecovirus entry-promoting ability of CVR-Nb27 was evaluated by six different sarbecoviruses in 293T cells. Data are presented as mean ± s.d. (biological quadruples of infected cells), representative of two independent infection assays. Unpaired two-tailed Student’s t-tests. d, BLI analyses of binding kinetics of immobilized RBD and S1-hFc (for 229E) of the indicated coronaviruses with the indicated monomeric nanobodies. e, a summary of binding kinetics of nanobodies bound to the immobilized virus antigens. f, g, Expression (f) and entry-supporting efficiency (g) of the CVRs with or without EPM transiently expressed in the HEK293T cells. EPM: endocytosis prevention motif. Data are mean ± s.d. (n = 3 independently infected cells). Unpaired two-tailed Student’s t-tests. Experiments were performed twice with similar results, and representative data were shown. h-k, HKU1 specific 2D1-CVR exhibited a comparable receptor function to TMPRSS2. The expression (h) and HKU1-PSV entry promoting efficiency (h, i) of the two receptors were examined. The amplification of propagation-competent rVSV-HKU1-GFP in Caco2 cells expressing 2D1-CVR and TMPRSS2 was demonstrated by GFP (j) and RNA accumulation (k). Scale bars: 100 μm for f, h, and j. Data are mean ± s.d. (n = 3 biologically independent wells of cells) analyzed by unpaired two-tailed Student’s t-tests for i. Data presented are RNA copies of two independently infected cells with each point representing the mean of technical duplicates (RT-qPCR) for k. Experiments presented were independently performed twice with similar results for h-j and single time for k. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; NS, Not significant (P > 0.05).
Extended Data Fig. 9 Neutralization and inhibition assays based on CVR-expressing HEK293T cells.
a, Comparison of neutralization profiles of sera collected from COVID-19 convalescents (left) or vaccinated individuals (right) based on HEK293T cells expressing ACE2 or two different CVRs. Serum dilution: 1:200. Heatmap plotted by the mean values (biologically triplicates of infected cells), which are representative results out of two independent experiments. b, c, Neutralization assays of several broadly neutralizing antibodies against PSV entry of representative coronaviruses in HEK293T stably expressing the indicated CVRs. Neutralization curves (b) and a summary of IC50 (c) against each virus are shown. The RBD-targeting REGN 19033 (REGN) was employed as a control. /: no inhibition detected. Data are mean ± s.d. (biologically triplicates of infected cells). d, The IC50 of selected entry inhibitors against SARS-CoV-2 PSV entry was determined in both HEK293T-ACE2 or HEK293T-LCB1-CVR cells. Data are mean ± s.d. (biologically triplicates of infected cells). e, Inhibitory efficacy of inhibitors against PSV entry of SARS-CoV-2-D614G, HKU1, HKU3 and HKU5 in HEK293T cells stably expressing the indicated CVRs. Data are mean ± s.d. (biologically triplicates of infected cells), representative of two infection inhibition assays with similar results. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; NS, Not significant (P > 0.05).
Extended Data Fig. 10 Characterization of CVR-promoted amplification of replication-competent pcVSV-CoVs or authentic coronaviruses.
a, Genetic organizations and workflow for generating replicable pcVSV-HKU3 or pcVSV-HKU5. b, Successful rescue (P0) and amplification (P1) of pcVSV-HKU3 assisted by VSV-G. Representative images of an experiment that was conducted for a single time. c, d, Trypsin-enhanced cell-cell fusion (c) and VSV-G-independence (d) of pcVSV-HKU3 infection in Caco2-Nb27 (MOI: 0.001). Data are representative of four independent infection assays with similar results. e, f, Accumulation of pcVSV-HKU3 (e) or pcVSV-HKU5 (f) RNA copies in the supernatant at indicated time points. g, pcVSV-HKU5 mediated cell-cell fusion in Caco2 or Caco2-1B4 cells at MOI = 0.1. TPCK-Try + : 20 μg ml−1 TPCK-treated Trypsin in DMEM + 2% FBS. Representative of two independent experiments. h, TCID50 determination assay for RsHuB2019A in Caco2-Nb27 cells by the Red-Muench method. Caco2-Nb27 cells were inoculated with a 10-fold serial dilution of RsHuB2019A containing supernatant (Passage 6). The TCID50 was determined using immunofluorescence to detect the presence of N protein expression of the inoculated cells at 4 dpi. i, Trypsin-dependent amplification of RsHuB2019A in Huh-7 cells. The RsHuB2019A genomic RNA copies in the supernatant collected at indicated time points of infected Huh-7 cells were quantified by RT-qPCR using RdRp-specific primers. Inoculation was conducted at an MOI of 0.0001, with or without trypsin treatment. Try: 100 μg ml−1 Trypsin in DMEM. j, Genetic organizations of the HKU5 (HKU5-1 LMH03f) ΔORF5-ZsGreen-HiBiT (HKU5-ZGH). k-m, Supernatant RNA copies of HKU5-ZGH (k) in Caco2-1B4 cells, ZsGreen-HitBit signal (l), and increase in ZsGreen intensity (P0) (m). Representative of two independent infection assays are shown. Data are mean ± s.d. (n = 3 independently infected wells of cells) for l. n, Isolation of HKU5 (strain PaGD2014/15) from bat samples by Caco2-1B4 cells and its trypsin-dependent propagation. o, Vero E6 and Caco2 cells were infected with HKU5-1 (LMH03f) with or without trypsin treatment. Data are representative of three independent experiments. p, Sequencing results show L76R and K519T mutations in HKU5-1 spikes after ten passages in Caco2-1B4 cells. q, HKU5-1 after ten passages carrying mutations remains unable to infect Caco2 cells without exogenous trypsin treatment. Caco2-1B4 with CVR expression was included as a positive control. r, Efficacy of indicated antiviral reagents against HKU5-1 infection in Caco2-1B4 cells assessed by intracellular N proteins at 48 hpi. s, Efficient inhibition of HKU5 entry by EK1 and EK1C4 peptides in Caco2-1B4. t, Inhibitory effect of selected anti-viral reagents against authentic HKU5-ZGH infection in Caco2-1B4. Inhibitors were coincubated with either the cells or the viruses for 1 h and present in the culture medium during infection. The HiBiT-based luciferase activity was determined at 48 hpi to assess the inhibitory effect of selected anti-viral reagents against the infection of authentic HKU5-ZGH in Caco2-1B4. Data are representative of two independent infection assays for r-t. u, Overview of the protease cleavage sites of selected coronaviruses. The residue responsible for reduced endosomal cysteine protease activity (ECP) is marked in red, numbering based on SARS-CoV-2. The HKU5 infection efficiencies in n, o, q, r, and s were assessed using rabbit polyclonal antibodies targeting the HKU5 N protein (Cy3) at 48 hpi. Data presented are RNA copies of two independently infected cells with each point representing the mean of technical duplicates (RT-qPCR) for e, f, i, and k, representative of two independent experiments. Scale bars: 125 μm for all images.
Extended Data Fig. 11 Schematic diagram of the modular design strategy for CVRs and the potential applications of this technique.
a, Workflow outlining the process of creating artificial receptor scaffolds (ARS) and viral binding domains (VBDs) to construct functional customized viral receptors (CVRs) for establishing in vitro and in vivo infection models. b, The crucial role of CVRs in bridging infection models and virus strains, along with the potential applications of this technique in both basic research and applied science.
Supplementary information
Supplementary Figures
Supplementary Figs 1 and 2 present uncropped immunoblots from the Main and Extended Data Figs, respectively; Supplementary Fig. 3 describes gating strategies for flow cytometry analysis.
Supplementary Table 1
Virus and receptor gene information.
Supplementary Table 2
Domain sequences for CVR design.
Supplementary Table 3
Information regarding nanobodies used in this study.
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.
About this article
Cite this article
Liu, P., Huang, ML., Guo, H. et al. Design of customized coronavirus receptors. Nature 635, 978–986 (2024). https://doi.org/10.1038/s41586-024-08121-5
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41586-024-08121-5
This article is cited by
-
Designer viral receptors
Nature Reviews Microbiology (2025)
-
SARS-CoV-2 spike protein: structure, viral entry and variants
Nature Reviews Microbiology (2025)
-
Engineering virus-like particles for safe and versatile modeling of SARS-CoV-2 host interaction and immune escape
Communications Biology (2025)