1887

Abstract

Following the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in PR China in late 2019 a number of variants have emerged, with two of these – alpha and delta – subsequently growing to global prevalence. One characteristic of these variants are changes within the spike protein, in particular the receptor-binding domain (RBD). From a public health perspective, these changes have important implications for increased transmissibility and immune escape; however, their presence could also modify the intrinsic host range of the virus. Using viral pseudotyping, we examined whether the variants of concern (VOCs) alpha, beta, gamma and delta have differing host angiotensin-converting enzyme 2 (ACE2) receptor usage patterns, focusing on a range of relevant mammalian ACE2 proteins. All four VOCs were able to overcome a previous restriction for mouse ACE2, with demonstrable differences also seen for individual VOCs with rat, ferret or civet ACE2 receptors, changes that we subsequently attributed to N501Y and E484K substitutions within the spike RBD.

Funding
This study was supported by the:
  • Medical Research Council (Award MR/W005611/1)
    • Principle Award Recipient: DalanBailey
  • Medical Research Council (Award MR/V036750/1)
    • Principle Award Recipient: CatherineN Hall
  • Biotechnology and Biological Sciences Research Council (Award BB/T008784/1)
    • Principle Award Recipient: NaziaThakur
  • Biotechnology and Biological Sciences Research Council (Award BBS/E/I/00007034)
    • Principle Award Recipient: DalanBailey
  • Biotechnology and Biological Sciences Research Council (Award BBS/E/I/00007038)
    • Principle Award Recipient: DalanBailey
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001735
2022-04-04
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/jgv/103/4/jgv001735.html?itemId=/content/journal/jgv/10.1099/jgv.0.001735&mimeType=html&fmt=ahah

References

  1. Zhou P, Yang X-L, Wang X-G, Hu B, Zhang L et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020; 579:270–273 [View Article] [PubMed]
    [Google Scholar]
  2. Sarah T, Khamsing V, Eduard Baquero S, Sandie M, Max B et al. Coronaviruses with a SARS-cov-2-like receptor-binding domain allowing ACE2-mediated entry into human cells isolated from bats of indochinese peninsula. Research Square 2021
    [Google Scholar]
  3. Shi J, Wen Z, Zhong G, Yang H, Wang C et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science 2020; 368:1016–1020 [View Article] [PubMed]
    [Google Scholar]
  4. Zhang Q, Zhang H, Huang K, Yang Y, Hui X et al. SARS-ccovv-2 neutralizing serum antibodies in cats: a serological investigation. bioRxiv 20202020.04.01.021196
    [Google Scholar]
  5. Kim Y-I, Kim S-G, Kim S-M, Kim E-H, Park S-J et al. Infection and rRapid tTransmission of SARS-CoV-2 in fFerrets. Cell Host Microbe 2020; 27:704–709 [View Article] [PubMed]
    [Google Scholar]
  6. de Vries RD, Rockx B, Haagmans BL, Herfst S, Koopmans MP et al. Animal models of SARS-CoV-2 transmission. Current Opinion in Virology 2021; 50:8–16 [View Article] [PubMed]
    [Google Scholar]
  7. Conceicao C, Thakur N, Human S, Kelly JT, Logan L et al. The SARS-CoV-2 Spike protein has a broad tropism for mammalian ACE2 proteins. PLoS Biol 2020; 18:12 [View Article]
    [Google Scholar]
  8. Li Y, Wang H, Tang X, Fang S, Ma D et al. SARS-CoV-2 and three related coronaviruses utilize multiple ACE2 orthologs and are potently blocked by an improved ACE2-Ig. J Virol 2020; 94:22 [View Article] [PubMed]
    [Google Scholar]
  9. Zhao X, Chen D, Szabla R, Zheng M, Li G et al. Broad and differential animal angiotensin-converting enzyme 2 receptor usage by SARS-CoV-2. J Virol 2020; 94:18 [View Article] [PubMed]
    [Google Scholar]
  10. Zhou J, Peacock TP, Brown JC, Goldhill DH, Elrefaey AME et al. Mutations that adapt sarssars-ccovv-2 to mustelid hosts do not increase fitness in the human airway. bioRxiv 20212021.08.20.456972
    [Google Scholar]
  11. Mishra S, Mindermann S, Sharma M, Whittaker C, Mellan TA et al. Changing composition of SARS-CoV-2 lineages and rise of Delta variant in England. EClinicalMedicine 2021; 39:101064 [View Article] [PubMed]
    [Google Scholar]
  12. Lupala CS, Ye Y, Chen H, Su XD, Liu H. Mutations on RBD of SARS-CoV-2 Omicron variant result in stronger binding to human ACE2 receptor. Biochem Biophys Res Commun 2022; 590:34–41 [View Article] [PubMed]
    [Google Scholar]
  13. Newman J, Thakur N, Peacock TP, Bialy D, Elreafey AM et al. Neutralising antibody activity against SARS-CoV-2 variants, including omicron, in an elderly cohort vaccinated with BNT162B2. medRxiv 2021
    [Google Scholar]
  14. Syed AM, Ciling A, Khalid MM, Sreekumar B, Chen P-Y et al. Omicron mutations enhance infectivity and reduce antibody neutralization of SARS-CoV-2 virus-like particles. medRxiv 20222021.12.20.21268048 [View Article]
    [Google Scholar]
  15. Peacock TP, Goldhill DH, Zhou J, Baillon L, Frise R et al. The furin cleavage site in the SARS-CoV-2 spike protein is required for transmission in ferrets. Nat Microbiol 2021; 6:899–909 [View Article] [PubMed]
    [Google Scholar]
  16. Meng B, Kemp SA, Papa G, Datir R, Ferreira IATM et al. Recurrent emergence of SARS-CoV-2 spike deletion H69/V70 and its role in the Alpha variant B.1.1.7. Cell Rep 2021; 35:109292 [View Article]
    [Google Scholar]
  17. Thorne LG, Bouhaddou M, Reuschl A-K, Zuliani-Alvarez L, Polacco B et al. Evolution of enhanced innate immune evasion by the SARS-CoV-2 B.1.1.7 UK variant. bioRxiv 20212021.06.06.446826 [View Article]
    [Google Scholar]
  18. Wall EC, Wu M, Harvey R, Kelly G, Warchal S et al. AZD1222-induced neutralising antibody activity against SARS-CoV-2 Delta VOC. The Lancet 2021; 398:207–209 [View Article] [PubMed]
    [Google Scholar]
  19. Wall EC, Wu M, Harvey R, Kelly G, Warchal S et al. Neutralising antibody activity against SARS-CoV-2 VOCs B.1.617.2 and B.1.351 by BNT162b2 vaccination. The Lancet 2021; 397:2331–2333 [View Article] [PubMed]
    [Google Scholar]
  20. Thakur N, Gallo G, Elreafey AME, Bailey D. Production of Recombinant Replication-defective Lentiviruses Bearing the SARS-CoV or SARS-CoV-2 Attachment Spike Glycoprotein and Their Application in Receptor Tropism and Neutralisation Assays. Bio Protoc 2021; 11:e4249 [View Article]
    [Google Scholar]
  21. Yu J, Li Z, He X, Gebre MS, Bondzie EA et al. Deletion of the SARS-CoV-2 spike cytoplasmic tail increases infectivity in pseudovirus neutralization assays. J Virol 2021; 95:11 [View Article] [PubMed]
    [Google Scholar]
  22. Thakur N, Gallo G, Elreafey AME, Bailey D. Production of recombinant replication-defective lentiviruses bearing the SARS-CoV or SARS-CoV-2 attachment spike glycoprotein and their application in receptor tropism and neutralisation assays. Bio Protoc 2021; 11:e4249 [View Article] [PubMed]
    [Google Scholar]
  23. Dinnon KH 3rd, Leist SR, Schäfer A, Edwards CE, Martinez DR et al. A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures. Nature 2020; 586:560–566 [View Article] [PubMed]
    [Google Scholar]
  24. Gu H, Chen Q, Yang G, He L, Fan H et al. Adaptation of SARS-CoV-2 in BALB/c mice for testing vaccine efficacy. Science 2020; 369:1603–1607 [View Article] [PubMed]
    [Google Scholar]
  25. Kim Y, Gaudreault NN, Meekins DA, Perera KD, Bold D et al. Effects of spike mutations in SARS-CoV-2 variants of concern on human or animal ACE2-mediated virus entry and neutralization. bioRxiv 20212021.08.25.457627 [View Article]
    [Google Scholar]
  26. Wang R, Zhang Q, Ge J, Ren W, Zhang R et al. Analysis of SARS-CoV-2 variant mutations reveals neutralization escape mechanisms and the ability to use ACE2 receptors from additional species. Immunity 2021; 54:1611–1621 [View Article] [PubMed]
    [Google Scholar]
  27. Wei Y, Aris P, Farookhi H, Xia X. Predicting mammalian species at risk of being infected by SARS-CoV-2 from an ACE2 perspective. Sci Rep 2021; 11:1702 [View Article] [PubMed]
    [Google Scholar]
  28. Hammer AS, Quaade ML, Rasmussen TB, Fonager J, Rasmussen M et al. SARS-CoV-2 Transmission between Mink (Neovison vison) and Humans, Denmark. Emerg Infect Dis 2021; 27:547–551 [View Article] [PubMed]
    [Google Scholar]
  29. Koopmans M. SARS-CoV-2 and the human-animal interface: outbreaks on mink farms. Lancet Infect Dis 2021; 21:18–19 [View Article] [PubMed]
    [Google Scholar]
  30. Montagutelli X, Prot M, Levillayer L, Salazar EB, Jouvion G et al. The b1.351 and p.1 variants extend sars-cov-2 host range to mice. bioRxiv 20212021.03.18.436013
    [Google Scholar]
  31. Horspool AM, Ye C, Wong TY, Russ BP, Lee KS et al. SARS-CoV-2 B.1.1.7 and B.1.351 variants of concern induce lethal disease in K18-hACE2 transgenic mice despite convalescent plasma therapy. bioRxiv 20212021.05.05.442784 [View Article]
    [Google Scholar]
  32. Huiping S, Jasper C, Terrence Tsz-Tai Y, Chaemin Y, Jingchu H et al. Emerging SARS-cov-2 variants expand species tropism to rodents. Research Square 2021
    [Google Scholar]
  33. Huang H, Zhu Y, Niu Z, Zhou L, Sun Q. SARS-CoV-2 N501Y variants of concern and their potential transmission by mouse. Cell Death Differ 2021; 28:2840–2842 [View Article] [PubMed]
    [Google Scholar]
  34. Shou S, Liu M, Yang Y, Kang N, Song Y et al. Animal Models for COVID-19: Hamsters, Mouse, Ferret, Mink, Tree Shrew, and Non-human Primates. Front Microbiol 2021; 12:626553 [View Article]
    [Google Scholar]
  35. Schlottau K, Rissmann M, Graaf A, Schön J, Sehl J et al. SARS-CoV-2 in fruit bats, ferrets, pigs, and chickens: an experimental transmission study. Lancet Microbe 2020; 1:e218–e225 [View Article] [PubMed]
    [Google Scholar]
  36. Vergara-Alert J, Rodon J, Carrillo J, Te N, Izquierdo-Useros N et al. Pigs are not susceptible to SARS-CoV-2 infection but are a model for viral immunogenicity studies. Transbound Emerg Dis 2021; 68:1721–1725 [View Article] [PubMed]
    [Google Scholar]
  37. Lean FZX, Núñez A, Spiro S, Priestnall SL, Vreman S et al. Differential susceptibility of SARS-CoV-2 in animals: Evidence of ACE2 host receptor distribution in companion animals, livestock and wildlife by immunohistochemical characterisation. Transbound Emerg Dis 20211–12 [View Article] [PubMed]
    [Google Scholar]
  38. Kuchipudi SV, Surendran-Nair M, Ruden RM, Yon M, Nissly RH et al. Multiple spillovers and onward transmission of sars-cov-2 in free-living and captive white-tailed deer (odocoileus virginianus). bioRxiv 20212021.10.31.466677
    [Google Scholar]
  39. Smyth DS, Trujillo M, Gregory DA, Cheung K, Gao A et al. Tracking cryptic sars-cov-2 lineages detected in nyc wastewater. medRxiv 2021
    [Google Scholar]
  40. Patterson EI, Elia G, Grassi A, Giordano A, Desario C et al. Evidence of exposure to SARS-CoV-2 in cats and dogs from households in Italy. Nat Commun 2020; 11:6231 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001735
Loading
/content/journal/jgv/10.1099/jgv.0.001735
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error