Skip to main content

Advertisement

Log in

Structural and functional characteristics of the SARS-CoV-2 Omicron subvariant BA.2 spike protein

  • Article
  • Published:

From Nature Structural & Molecular Biology

View current issue Submit your manuscript

Abstract

The Omicron subvariant BA.2 has become the dominant circulating strain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in many countries. Here, we have characterized structural, functional and antigenic properties of the full-length BA.2 spike (S) protein and compared replication of the authentic virus in cell culture and an animal model with previously prevalent variants. BA.2 S can fuse membranes slightly more efficiently than Omicron BA.1, but still less efficiently than other previous variants. Both BA.1 and BA.2 viruses replicated substantially faster in animal lungs than the early G614 (B.1) strain in the absence of pre-existing immunity, possibly explaining the increased transmissibility despite their functionally compromised spikes. As in BA.1, mutations in the BA.2 S remodel its antigenic surfaces, leading to strong resistance to neutralizing antibodies. These results suggest that both immune evasion and replicative advantage may contribute to the heightened transmissibility of the Omicron subvariants.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Functional properties of Omicron BA.2 S.
Fig. 2: Purification of the Omicron BA.2 full-length S protein.
Fig. 3: Antigenic properties of the purified full-length BA.2 S protein.
Fig. 4: Cryo-EM structures of the full-length BA.2 S protein.
Fig. 5: Structural impact of the mutations in the BA.2 S.
Fig. 6: Antigenic surfaces of the RBD.

Similar content being viewed by others

Data availability

The atomic structure coordinates and EM maps are deposited in the Protein Data Bank (PDB) and Electron Microscopy Data Bank (EMDB) under the following accession numbers: PDB ID 8D55 and EMDB ID EMD-27205 for the RBD-down conformation, PDB ID 8D56 and EMDB ID EMD-27206 for the one-RBD-up conformation, and PDB ID 8D5A and EMDB ID EMD-27207 for the RBD-intermediate conformation. All other related data generated and/or analyzed during the current study are available from the corresponding author upon reasonable request. The initial templates for model building include PDB IDs 7KRQ and 7KRR. Source data are provided with this paper.

References

  1. Hirotsu, Y. et al. SARS-CoV-2 Omicron sublineage BA.2 replaces BA.1.1: genomic surveillance in Japan from September 2021 to March 2022. J. Infect. 85, 174–211 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Lyngse, F. P. et al. Household transmission of SARS-CoV-2 Omicron variant of concern subvariants BA.1 and BA.2 in Denmark. Nat. Commun. 13, 5760 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Mefsin, Y. M. et al. Epidemiology of infections with SARS-CoV-2 Omicron BA.2 variant, Hong Kong, January–March 2022. Emerg. Infect. Dis. 28, 1856–1858 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Smith, D. J. et al. COVID-19 mortality and vaccine coverage — Hong Kong Special Administrative Region, China, January 6, 2022–March 21, 2022. MMWR Morb. Mortal. Wkly Rep. 71, 545–548 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Yamasoba, D. et al. Virological characteristics of the SARS-CoV-2 Omicron BA.2 spike. Cell 185, 2103–2115.e19 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Wolter, N. et al. Clinical severity of omicron lineage BA.2 infection compared with BA.1 infection in South Africa. Lancet 400, 93–96 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Iketani, S. et al. Antibody evasion properties of SARS-CoV-2 Omicron sublineages. Nature 604, 553–556 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gruell, H. et al. SARS-CoV-2 Omicron sublineages exhibit distinct antibody escape patterns. Cell Host Microbe. 30, 1231–1241.e6 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Yu, J. et al. Neutralization of the SARS-CoV-2 Omicron BA.1 and BA.2 variants. N. Engl. J. Med. 386, 1579–1580 (2022).

    PubMed  Google Scholar 

  10. Mykytyn, A. Z. et al. Antigenic cartography of SARS-CoV-2 reveals that Omicron BA.1 and BA.2 are antigenically distinct. Sci Immunol. 7, eabq4450 (2022).

    CAS  PubMed  Google Scholar 

  11. Cao, Y. et al. Omicron BA.2 specifically evades broad sarbecovirus neutralizing antibodies. Preprint at bioRxiv https://doi.org/10.1101/2022.02.07.479349 (2022).

  12. Zou, J. et al. Cross-neutralization of Omicron BA.1 against BA.2 and BA.3 SARS-CoV-2. Nat. Commun. 13, 2956 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Chemaitelly, H. et al. Protection of Omicron sub-lineage infection against reinfection with another Omicron sub-lineage. Nat. Commun. 13, 4675 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Stegger, M. et al. Occurrence and significance of Omicron BA.1 infection followed by BA.2 reinfection. Preprint at medRxiv https://doi.org/10.1101/2022.02.19.22271112 (2022).

  15. Kirsebom, F. C. M. et al. COVID-19 vaccine effectiveness against the omicron (BA.2) variant in England. Lancet Infect. Dis. 22, 931–933 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Bowen, J. E. et al. Omicron spike function and neutralizing activity elicited by a comprehensive panel of vaccines. Science. 377, 890–894 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Dai, L. & Gao, G. F. Viral targets for vaccines against COVID-19. Nat. Rev. Immunol. 21, 73–82 (2021).

    CAS  PubMed  Google Scholar 

  18. Bosch, B. J., van der Zee, R., de Haan, C. A. M. & Rottier, P. J. M. The coronavirus spike protein is a class I virus fusion protein: structural and functional characterization of the fusion core complex. J. Virol. 77, 8801–8811 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Millet, J. K. & Whittaker, G. R. Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein. Proc. Natl Acad. Sci. USA 111, 15214–15219 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Tortorici, M. A. & Veesler, D. Structural insights into coronavirus entry. Adv. Virus Res 105, 93–116 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Jackson, C. B., Farzan, M., Chen, B. & Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 23, 3–20 (2022).

    CAS  PubMed  Google Scholar 

  23. Shang, J. et al. Cell entry mechanisms of SARS-CoV-2. Proc. Natl Acad. Sci. USA 117, 11727–11734 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Wrapp, D. et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 367, 1260–1263 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Harvey, W. T. et al. SARS-CoV-2 variants, spike mutations and immune escape. Nat. Rev. Microbiol. 19, 409–424 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Hong, Q. et al. Molecular basis of receptor binding and antibody neutralization of Omicron. Nature 604, 546–552 (2022).

    CAS  PubMed  Google Scholar 

  27. Stalls, V. et al. Cryo-EM structures of SARS-CoV-2 Omicron BA.2 spike. Cell Rep. 39, 111009 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, J. et al. Structural impact on SARS-CoV-2 spike protein by D614G substitution. Science 372, 525–530 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Cai, Y. et al. Structural basis for enhanced infectivity and immune evasion of SARS-CoV-2 variants. Science 373, 642–648 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang, J. et al. Membrane fusion and immune evasion by the spike protein of SARS-CoV-2 Delta variant. Science 374, 1353–1360 (2021).

    CAS  PubMed  Google Scholar 

  31. Zhang, J. et al. Structural and functional impact by SARS-CoV-2 Omicron spike mutations. Cell Rep. 39, 110729 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Cai, Y. et al. Distinct conformational states of SARS-CoV-2 spike protein. Science 369, 1586–1592 (2020).

    CAS  PubMed  Google Scholar 

  33. Ogando, N. S. et al. SARS-coronavirus-2 replication in Vero E6 cells: replication kinetics, rapid adaptation and cytopathology. J. Gen. Virol. 101, 925–940 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. McCray, P. B.Jr. et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 81, 813–821 (2007).

    CAS  PubMed  Google Scholar 

  35. Radvak, P. et al. SARS-CoV-2 B.1.1.7 (alpha) and B.1.351 (beta) variants induce pathogenic patterns in K18-hACE2 transgenic mice distinct from early strains. Nat. Commun. 12, 6559 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Tong, P. et al. Memory B cell repertoire for recognition of evolving SARS-CoV-2 spike. Cell 184, 4969–4980.e15 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Xiao, T. et al. A trimeric human angiotensin-converting enzyme 2 as an anti-SARS-CoV-2 agent. Nat. Struct. Mol. Biol. 28, 202–209 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. 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).

    CAS  PubMed  Google Scholar 

  39. Zhang, J., Xiao, T., Cai, Y. & Chen, B. Structure of SARS-CoV-2 spike protein. Curr. Opin. Virol. 50, 173–182 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Tian, F. et al. N501Y mutation of spike protein in SARS-CoV-2 strengthens its binding to receptor ACE2. eLife 10, e69091 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Mannar, D. et al. SARS-CoV-2 Omicron variant: Antibody evasion and cryo-EM structure of spike protein-ACE2 complex. Science. 375, 760–764 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Yin, W. et al. Structures of the Omicron spike trimer with ACE2 and an anti-Omicron antibody. Science. 375, 1048–1053 (2022).

  43. McCallum, M. et al. Structural basis of SARS-CoV-2 Omicron immune evasion and receptor engagement. Science 375, 864–868 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Cui, Z. et al. Structural and functional characterizations of infectivity and immune evasion of SARS-CoV-2 Omicron. Cell. 185, 860–871.e13 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Gobeil, S. M.-C. et al. Effect of natural mutations of SARS-CoV-2 on spike structure, conformation, and antigenicity. Science 373, eabi6226 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Davies, N. G. et al. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science 372, eabg3055 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hart, W. S. et al. Generation time of the alpha and delta SARS-CoV-2 variants: an epidemiological analysis. Lancet Infect. Dis. 22, 603–610 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Milne, G. et al. Does infection with or vaccination against SARS-CoV-2 lead to lasting immunity? Lancet Respir. Med 9, 1450–1466 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Levin, E. G. et al. Waning immune humoral response to BNT162b2 Covid-19 vaccine over 6 months. N. Engl. J. Med. 385, e84 (2021).

    CAS  PubMed  Google Scholar 

  51. Meng, B. et al. Altered TMPRSS2 usage by SARS-CoV-2 Omicron impacts infectivity and fusogenicity. Nature 603, 706–714 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Zeng, C. et al. Neutralization and stability of SARS-CoV-2 Omicron variant. Preprint at bioRxiv https://doi.org/10.1101/2021.12.16.472934 (2021).

  53. Halfmann, P. J. et al. SARS-CoV-2 Omicron virus causes attenuated disease in mice and hamsters. Nature 603, 687–692 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Suzuki, R. et al. Attenuated fusogenicity and pathogenicity of SARS-CoV-2 Omicron variant. Nature 603, 700–705 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Shuai, H. et al. Attenuated replication and pathogenicity of SARS-CoV-2 B.1.1.529 Omicron. Nature 603, 693–699 (2022).

    CAS  PubMed  Google Scholar 

  56. Sentis, C. et al. SARS-CoV-2 Omicron variant, lineage BA.1, is associated with lower viral load in nasopharyngeal samples compared to Delta variant. Viruses 14, 919 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Yinda, C. K. et al. K18-hACE2 mice develop respiratory disease resembling severe COVID-19. PLoS Pathog. 17, e1009195 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Romano, M., Ruggiero, A., Squeglia, F., Maga, G. & Berisio, R. A structural view of SARS-CoV-2 RNA replication machinery: RNA synthesis, proofreading and final capping. Cells 9, 1267 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Hansen, J. et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science 369, 1010–1014 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Pinto, D. et al. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature 583, 290–295 (2020).

    CAS  PubMed  Google Scholar 

  61. Tortorici, M. A. et al. Ultrapotent human antibodies protect against SARS-CoV-2 challenge via multiple mechanisms. Science 370, 950–957 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Starr, T. N. et al. SARS-CoV-2 RBD antibodies that maximize breadth and resistance to escape. Nature 597, 97–102 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Huo, J. et al. Neutralization of SARS-CoV-2 by destruction of the prefusion spike. Cell Host Microbe 28, 445–454.e6 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Chen, J. et al. Effect of the cytoplasmic domain on antigenic characteristics of HIV-1 envelope glycoprotein. Science 349, 191–195 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Chen, C. Z. et al. Identifying SARS-CoV-2 entry inhibitors through drug repurposing screens of SARS-S and MERS-S pseudotyped particles. ACS Pharmacol. Transl. Sci. 3, 1165–1175 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Millet, J. K. & Whittaker, G. R. Murine leukemia virus (MLV)-based coronavirus spike-pseudotyped particle production and infection. Bio Protoc. 6, e2035 (2016).

    PubMed  Google Scholar 

  68. Case, J. B., Bailey, A. L., Kim, A. S., Chen, R. E. & Diamond, M. S. Growth, detection, quantification, and inactivation of SARS-CoV-2. Virology 548, 39–48 (2020).

    CAS  PubMed  Google Scholar 

  69. Xie, X. et al. An infectious cDNA clone of SARS-CoV-2. Cell Host Microbe 27, 841–848.e3 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    PubMed  Google Scholar 

  71. Scheres, S. H. W. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 180, 519–530 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Croll, T. I. ISOLDE: a physically realistic environment for model building into low-resolution electron-density maps. Acta Crystallogr. D Struct. Biol. 74, 519–530 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 30, 70–82 (2021).

    CAS  PubMed  Google Scholar 

  75. Morin, A. et al. Collaboration gets the most out of software. eLife 2, e01456 (2013).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the SBGrid team for computing support, S. Harrison and J. Abraham for computing resources, K. Arnett for support and advice on the BLI experiments, and S. Harrison for critical reading of the manuscript. EM data were collected at the Harvard Cryo-EM Center for Structural Biology at Harvard Medical School. We acknowledge support for COVID-19-related structural biology research at Harvard from the Nancy Lurie Marks Family Foundation and the Massachusetts Consortium on Pathogen Readiness (MassCPR). This work was supported by Fast Grants from Emergent Ventures (to B.C. and D.R.W.), COVID-19 Awards from MassCPR (to B.C., D.R.W. and M.S.S.), and NIH grants AI147884 (to B.C.), AI141002 (to B.C.), AI127193 (to B.C. and J. Chou), AI39538 (to D.R.W.), AI170715 (to D.R.W.), AI165072 (to D.R.W.) and AI169619 (to D.R.W). This work was also supported in part by the FDA Center for Biologics Evaluation and Research (CBER) intramural SARS-CoV-2 pandemic fund (to H.X.). The clinical isolate New York-PV09158/2020 (ATCC, NR-53516) and Omicron (B.1.1.529) BA.1 were obtained through BEI Resources, National Institute of Allergy and Infectious Diseases (NIAID), NIH: SARS-Related Coronavirus 2. The Delta (B.1.617.2) and Omicron BA.2 seed viruses were kindly provided by B. Zhou and C. Davis at CDC.

Author information

Authors and Affiliations

Authors

Contributions

B.C., T.X., J.Z. and H.G. conceived the project. H.G. expressed and purified the full-length S proteins with help from W.S. and H.P. T.X. designed and performed BLI and cell–cell fusion experiments, and was assisted by H.G. J.Z. prepared cryo grids and performed EM data collection with contributions from M.L.M., processed the cryo-EM data, and built and refined the atomic models. W.T., M.K., H.J.K. and H.X. carried out the animal study and in vitro virus replication kinetics using authentic viruses. C.L.L. and M.S.S. performed the neutralization assays using the HIV-based pseudoviruses. J.L. and S.W. created the BA.2 expression construct and performed the neutralization assays using the MLV-based pseudoviruses. H.Z. and K.A. performed the flow cytometry experiments. P.T., A.G. and D.R.W. produced anti-S monoclonal antibodies. S.R.-V. contributed to cell culture and protein production. All authors analyzed the data. B.C., T.X., J.Z., H.G. and H.X. wrote the manuscript with input from all other authors.

Corresponding authors

Correspondence to Tianshu Xiao, Hang Xie or Bing Chen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Sara Osman, in collaboration with the Nature Structural & Molecular Biology team.

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 Expression and cell-cell fusion of the S protein from Omicron BA.2.

(a) Schematic representation of a full-length Omicron BA.2 spike (S) protein. The sequence is derived from an Omicron BA.2 subvariant (hCoV-19/Denmark/DCGC-327158/2022). Segments of S1 and S2 include: NTD, N-terminal domain; RBD, receptor-binding domain; CTD1, C-terminal domain 1; CTD2, C-terminal domain 2; 630 loop, residues 620-640; S1/S2, the furin cleavage site at the S1/S2 boundary; S2’, S2’ cleavage site; FP, fusion peptide; FPPR, fusion peptide proximal region; HR1, heptad repeat 1; CH, central helix region; CD, connector domain; HR2, heptad repeat 2; TM, transmembrane segment; CT, cytoplasmic tail; and tree-like symbols for glycans. Positions of all mutations (from the amino-acid sequence of Wuhan-Hu-1) are indicated and those highlighted in red rectangles are also present in at least one of the previous VOCs. (b) Expression and processing of the full-length S constructs of various variants in HEK293 cells. S protein samples prepared from HEK293 cells transiently transfected with 10 μg of the full-length S expression plasmids were detected by anti-RBD polyclonal antibodies. Bands for the uncleaved S and S1 fragment are indicated. (c) HEK293T cells transfected with the untagged, full-length S protein expression plasmids were fused with ACE2-expressing cells. Cell-cell fusion led to reconstitution of α and ω fragments of β-galactosidase to form an active enzyme, and the fusion activity was then quantified by a chemiluminescent assay. No ACE2 and no S were negative controls. Error bars were generated with measurements on three biologically independent samples. All the experiments have been repeated at least twice with similar results.

Source data

Extended Data Fig. 2 Foci images of in vitro replication kinetics of the authentic viruses.

(a) and (b) Vero E6-TMPRSS2 cells were infected with the authentic G614 (B.1), Delta (B.1617.2), Omicron-BA.1 or BA.2 viruses at MOI of 0.005. Postinfection supernatants were titrated by a focus-forming assay. Foci as a cluster of cells expressing viral antigen were imaged and counted using AID vSpot Spectrum. (a) Extended virus replication kinetics in Vero E6-TMPRSS2 cells. Focus-forming units (FFU) per ml were determined and data are expressed as mean ± sem of n = 3 independent replicates/time point/virus. (b) images of foci (dark spots) titration shown in (a) with automatic counts in the lower corner of each well. (c) Vero E6 cells cells were infected with the authentic G614 (B.1), Delta (B.1617.2), Omicron-BA.1 or BA.2 viruses at MOI of 0.005. The data are summarized in Fig. 1d. (c) Vero E6-TMPRSS2 cells were infected with the authentic G614 (B.1), Delta (B.1617.2), Omicron-BA.1 or BA.2 viruses at MOI of 0.005. The data are summarized in Fig. 1e.

Source data

Extended Data Fig. 3 Infection of HEK293-ACE2 cells by MLV-based pseudotyped viruses.

Time course for single-cycle infection of HEK293-ACE2 cells by MLV-based pseudotyped viruses with various SARS-CoV-2 variant S constructs, as indicated, all containing a CT deletion. Infection was initiated by mixing viruses and target cells, and viruses were washed out at each time point as indicated. Delta AY.4.2 is a Delta subvariant. The relative light unit (RLU) of 8-hr infection of each pseudovirus was used as 100% infectivity for normalizing data at other time points. The relative infectivity (%) equals RLU(t)/RLU(8-hr)*100. The experiments were repeated at least three times with independent samples giving similar results. Error bars are standard deviations of 4 repeats of each data point (n = 3).

Source data

Extended Data Fig. 4 Tissue-specific viral burdens and hACE2 expression in K18-hACE2 mice at 1 and 3 days post infection (dpi).

Mice were intranasally inoculated with 100 TCID50/50 μl/mouse of G614 (B.1), Delta (B.1617.2), Omicron-BA.1 or BA.2. The viral RNA copies (a) or hACE2 expression (b) in various tissue homogenates were measured by RT-qPCR. Data are expressed as geometric means (bars) with geometric standard deviation (error bars). Individual results of infected mice (n = 4 mice/time point/group) and uninfected naïve mice (n = 5) are shown.

Source data

Extended Data Fig. 5 Additional antigenic analysis of the purified full-length BA.2S protein.

(a) Antibody competition groups as described in ref. 36. Surface regions of the S trimer targeted by antibodies on S1 are highlighted by orange ellipses, including RBD-1, RBD-2, RBD-3, NTD-1 and NTD-2. The exact location of NTD-2 is uncertain and therefore marked with a dashed line. (b) Binding analysis of the prefusion S trimers from G614 and BA.2 with soluble monomeric ACE2 and selected monoclonal antibodies was performed by BLI. For ACE2 binding, purified ACE2 protein was immobilized to AR2G biosensors and dipped into the wells containing each purified S proteins at various concentrations. For antibody binding, various antibodies were immobilized to AHC biosensors and dipped into the wells containing each purified S protein at different concentrations. Binding kinetics were evaluated using a 1:1 Langmuir model except for antibody 12A2 targeting the RBD-2, which was analyzed by a bivalent binding model. The sensorgrams are in black and the fits in red. Binding constants highlighted by underlines were estimated by steady-state analysis as described in the Methods. RU, response unit. Binding constants are also summarized here and in Table 1. N.D., not determined. All experiments were repeated at least twice with essentially identical results. (c) Steady-state analysis by plotting steady-state responses against concentrations. KD values were derived from the fits. Error bars were generated by the steady-state analysis of Octet Data Analysis HT Version 12.0 (ForteBio), fitting each sensorgram to a single exponential function to extract the response at equilibrium (Req) as described in Methods.

Source data

Extended Data Fig. 6 Antigenic properties of the cell-surface BA.2S protein assessed by flow cytometry.

Antibody binding to the full-length S proteins of the G614 and Omicron variants, as well as the uncleaved wildtype spike and an S2 construct expressed on the cell surfaces analyzed by flow cytometry. BA.2_notag, the unmodified, full-length S protein from the BA.2 subvariant. BA.2_streptag, the intact BA.2S protein fused with a C-terminal twin Strep tag. The antibodies and their targets are indicated. A designed ACE2-based fusion inhibitor ACE2615-foldon-T27W was used for detecting receptor binding37. No major difference in the trimeric ACE2 binding to non-tagged BA.2 and G614 spikes was observed in this assay, probably because the BA.2 trimer is less stable than G614 in the presence of ACE2 under the extensive washing conditions for flow cytometry. MFI, mean fluorescent intensity. Error bars represent standard errors of mean from measurements using three independently transfected cell samples. The flow cytometry assays were repeated three times with essentially identical results.

Source data

Extended Data Fig. 7 Cryo-EM structures of the full-length BA.2S protein.

(a) Three structures of the BA.2S trimer, representing the closed prefusion conformation, one-RBD-intermediate conformation and one-RBD-up conformations, were modeled based on corresponding cryo-EM density maps at 2.7 Å, 3.5 Å and 2.9 Å resolution, respectively. Three protomers (a, b, c) are colored in red, blue and green, respectively. RBD locations are indicated. Particle percentage for each class in the data processing is also indicated, but it may not accurately reflect the conformation distribution of the S trimer in solution. (b) Superposition of the three conformations of the BA.2 S trimer aligned by the invariant S2 with only one protomer for each shown for clarity. Three RBDs representing the closed prefusion, one-RBD-intermediate and one-RBD-up conformations are colored in cyan, yellow and orange, respectively.

Extended Data Fig. 8 Additional ordered residues near the furin site in the BA.2 structure.

(a) Superposition of the structure of the BA.2S trimer in ribbon representation and various colors with the structures of the BA.1 S in gray and G614 S in yellow aligned by S2, showing the region near the furin cleavage site. (b) Density near the furin cleavage site in the BA.2 map. (c) Processing of the revertant BA.2 mutations near the furin cleavage site. S protein samples prepared from HEK293 cells transiently transfected with 10 μg of the full-length S expression plasmids of G614, Delta, Omicron BA.2, BA.2-K679N, BA.2-H681P and BA.2-K679N/H681P were detected by anti-RBD polyclonal antibodies. Bands for the uncleaved S and S1 fragment are indicated. The experiment was repeated three times independently with similar results.

Source data

Extended Data Fig. 9 Local regions of mutations in BA.2S.

(a) A close-up view of the unique mutations in the BA.2 RBD. Superposition of the BA.2 RBD structure in ribbon representation and cyan with the structures of the RBDs of G614 S in yellow and BA.1 in gray. The mutated residues and the N-linked glycans at Asn343 are in stick model. NAG, N-acetylglucosamine. (b) Density in the BA.2 map near the short helix formed by residues 365-371 and the model fitting. (c) Possibly ordered 70-80 loop. The N-terminal segment of the BA.2S is shortened by the three-residue deletion (L24del-P25del-P26del) and also constrained by the disulfide bond between Cys15 and Cys136. There is reasonable density in which the 70-80 loop, disordered in many previous S trimer structures, could be modeled (shown in red). Such a structured loop can create a knot in this region, however, which will need a higher resolution map to confirm.

Extended Data Table 1 Neutralization of the SARS-CoV-2 Omicron subvariants

Supplementary information

Source data

Source Data Fig. 1

Source data for plots in Fig. 1a–f.

Source Data Fig. 2

Source data for gel-filtration trace in Fig. 2b.

Source Data Fig. 2

Unprocessed gel in Fig. 2b.

Source Data Fig. 3

Source data for sensorgrams and fits in Fig. 3.

Source Data Extended Data Fig. 1

Unprocessed western blot in Extended Data Fig. 1b.

Source Data Extended Data Fig. 1

Source data for plot in Extended Data Fig. 1c.

Source Data Extended Data Fig. 2

Source data for plot in Extended Data Fig. 2a.

Source Data Extended Data Fig. 3

Source data for plot in Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Source data for plots in Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Source data for sensorgrams and fits in Extended Data Fig. 5b and plot in Extended Data Fig. 5c.

Source Data Extended Data Fig. 6

Source data for plot in Extended Data Fig. 6.

Source Data Extended Data Fig. 8

Unprocessed western blot in Extended Data Fig. 8c.

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

Zhang, J., Tang, W., Gao, H. et al. Structural and functional characteristics of the SARS-CoV-2 Omicron subvariant BA.2 spike protein. Nat Struct Mol Biol 30, 980–990 (2023). https://doi.org/10.1038/s41594-023-01023-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-023-01023-6

  • Springer Nature America, Inc.

Navigation