Native-like SARS-CoV-2 Spike Glycoprotein Expressed by ChAdOx1 nCoV-19/AZD1222 Vaccine
- Yasunori WatanabeYasunori WatanabeSchool of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, U.K.More by Yasunori Watanabe
- ,
- Luiza MendonçaLuiza MendonçaDivision of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, U.K.More by Luiza Mendonça
- ,
- Elizabeth R. AllenElizabeth R. AllenThe Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.More by Elizabeth R. Allen
- ,
- Andrew HoweAndrew HoweElectron Bio-imaging Centre, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, U.K.More by Andrew Howe
- ,
- Mercede LeeMercede LeeThe Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.More by Mercede Lee
- ,
- Joel D. AllenJoel D. AllenSchool of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.More by Joel D. Allen
- ,
- Himanshi ChawlaHimanshi ChawlaSchool of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.More by Himanshi Chawla
- ,
- David PulidoDavid PulidoThe Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.More by David Pulido
- ,
- Francesca DonnellanFrancesca DonnellanThe Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.More by Francesca Donnellan
- ,
- Hannah DaviesHannah DaviesThe Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.More by Hannah Davies
- ,
- Marta UlaszewskaMarta UlaszewskaThe Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.More by Marta Ulaszewska
- ,
- Sandra Belij-RammerstorferSandra Belij-RammerstorferThe Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.NIHR Oxford Biomedical Research Centre, Oxford, U.K.More by Sandra Belij-Rammerstorfer
- ,
- Susan MorrisSusan MorrisThe Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.More by Susan Morris
- ,
- Anna-Sophia KrebsAnna-Sophia KrebsDivision of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, U.K.More by Anna-Sophia Krebs
- ,
- Wanwisa DejnirattisaiWanwisa DejnirattisaiThe Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.More by Wanwisa Dejnirattisai
- ,
- Juthathip MongkolsapayaJuthathip MongkolsapayaThe Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, ThailandChinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, U.K.More by Juthathip Mongkolsapaya
- ,
- Piyada SupasaPiyada SupasaThe Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.More by Piyada Supasa
- ,
- Gavin R. ScreatonGavin R. ScreatonThe Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.Division of Medical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, U.K.More by Gavin R. Screaton
- ,
- Catherine M. GreenCatherine M. GreenThe Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K.More by Catherine M. Green
- ,
- Teresa Lambe*Teresa Lambe*Email: [email protected] (T.L.).The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.NIHR Oxford Biomedical Research Centre, Oxford, U.K.More by Teresa Lambe
- ,
- Peijun Zhang*Peijun Zhang*Email: [email protected] (P.Z).Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, U.K.Electron Bio-imaging Centre, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, U.K.More by Peijun Zhang
- ,
- Sarah C. Gilbert*Sarah C. Gilbert*Email: [email protected] (S.C.G.).The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, U.K.NIHR Oxford Biomedical Research Centre, Oxford, U.K.More by Sarah C. Gilbert
- , and
- Max Crispin*Max Crispin*Email: [email protected] (M.C.).School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, U.K.More by Max Crispin
Abstract

Vaccine development against the SARS-CoV-2 virus focuses on the principal target of the neutralizing immune response, the spike (S) glycoprotein. Adenovirus-vectored vaccines offer an effective platform for the delivery of viral antigen, but it is important for the generation of neutralizing antibodies that they produce appropriately processed and assembled viral antigen that mimics that observed on the SARS-CoV-2 virus. Here, we describe the structure, conformation, and glycosylation of the S protein derived from the adenovirus-vectored ChAdOx1 nCoV-19/AZD1222 vaccine. We demonstrate native-like post-translational processing and assembly, and reveal the expression of S proteins on the surface of cells adopting the trimeric prefusion conformation. The data presented here confirm the use of ChAdOx1 adenovirus vectors as a leading platform technology for SARS-CoV-2 vaccines.
This publication is licensed under
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Synopsis
The structure, conformation, and glycosylation of the S protein derived from the adenovirus-vectored ChAdOx1 nCoV-19/AZD1222 vaccine are described. Also, native-like post-translational processing and assembly are demonstrated.
Introduction
Results and Discussion
Expression of Prefusion Conformation SARS-CoV-2 S Glycoprotein on Cell Surfaces upon ChAdOx1 nCoV-19 Infection
Figure 1

Figure 1. ChAdOx1 nCoV-19 produces membrane associated SARS-CoV-2 S glycoprotein in native conformations able to bind its host receptor, ACE2. (A) Schematic representation of the vaccine encoded SARS-CoV-2 S protein, showing the position of N-linked glycosylation amino acid sequons (NXS/T, where X ≠ P) as branches. Protein domains are illustrated: N-terminal domain (NTD), receptor-binding domain (RBD), fusion peptide (FP), heptad repeat 1 (HR1), central helix (CH), connector domain (CD), and transmembrane domain (TM), with the additional tPA secretion signal at the N-terminus. (B) HeLa S3 cells were infected with ChAdOx1 nCoV-19 and incubated with recombinant ACE2, anti-ChAdOx1 nCoV-19 (derived from vaccinated mice), or a panel of human mAbs (Ab44, Ab45, Ab71, and Ab111, which recognize S2, RBD, trimeric S, and NTD, respectively) and compared to noninfected controls, analyzed by flow cytometry. (Left). Relative frequency of cells and AlexaFluor 488 fluorescence associated with antispike detection is plotted. Left, (blue) anti-ChAdOx1 nCoV-19; middle (red), ACE2; and right (shades of green) human mAbs. In dark gray cells infected with an irrelevant ChAdOx1 vaccine and in light gray noninfected cells are shown as a control. Experimental replicates were performed two times, and representative data are shown.
Structural Analysis of Membrane-Associated ChAdOx1 nCoV-19 Derived SARS-CoV-2 S Protein
Figure 2

Figure 2. CryoET and subtomogram average of ChAdOx1 nCoV-19 derived spike. (A) Tomographic slice of U2OS cell transduced with ChAdOx1 nCoV-19. The slice is 6.4 Å thick; PM = plasma membrane, scale bar = 100 nm. (B) Detailed view of the boxed area marked in (A). White arrowheads indicate spike proteins on the cell surface; scale bar = 50 nm. (C–E) Subtomogram average of ChAdOx1 nCoV-19 spikes at 11.6 Å resolution as indicated by Fourier-Shell correlation at 0.5 cutoff (C), shown from side view (D) and top view (E). SARS-CoV-2 atomic model (PDB 6ZB5) (29) is fitted for reference.
Site-Specific Glycan Analysis of the ChAdOx1 nCoV-19 Derived SARS-CoV-2 S Protein Reveals Native-Like Glycan Maturation
Figure 3

Figure 3. Site-specific glycan processing of SARS-CoV-2 S upon infection with ChAdOx1 nCoV-19. (A) Western blot analysis of SARS-CoV-2 spike proteins, using anti-S1 and anti-S1+S2 antibodies. Lane 1 = Protein pellet from 293F cell lysates infected with ChAdOx1 nCoV-19. Lane 2 = Reduced protein pellet from 293F infected with ChAdOx1 nCoV-19. Lane 3 = 2P-stabilized SARS-CoV-2 S protein. The white boxes correspond to gel bands that were excised for mass spectrometric analysis. (B) Site-specific N-linked glycosylation of SARS-CoV-2 S0 and S1/S2 glycoproteins. The bar graphs represent the relative quantities of digested glycopeptides possessing the identifiers of oligomannose/hybrid-type glycans (green), complex-type glycans (pink), unoccupied PNGs (gray), or not determined (N.D.) at each N-linked glycan sequon on the S protein, listed from N to C terminus. (C) Glycosylated model of the cleaved (S1/S2) SARS-CoV-2 spike. The pie charts summarize the mass spectrometric analysis of the oligomannose/hybrid (green), complex (pink), or unoccupied (gray) N-linked glycan populations. Representative glycans are modeled onto the prefusion structure of trimeric SARS-CoV-2 S glycoprotein (PDB ID: 6VSB), (3) with one RBD in the “up” conformation. The modeled glycans are colored according to oligomannose/hybrid-type glycan content with glycan sites labeled in green (80–100%), orange (30–79%), pink (0–29%), or gray (not detected).
Materials and Methods
Production of ChAdOx1 nCoV-19
Production of ChAdOx1 nCoV-19 Derived SARS-CoV-2 S
Expression and Purification of Recombinant 2P-Stabilized SARS-CoV-2 S
Western Blot Analysis
FACS Analysis for Cell Surface Spike Expression
ACE2 Expression
Isolation of Human Monoclonal Antibodies from Peripheral B Cells by Spike-Specific Single B Cells Sorting
CryoET Sample Preparation and Imaging
Glycopeptide Analysis by Liquid Chromatography–Mass Spectrometry
Glycosylated Model Construction
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscentsci.1c00080.
Supplementary Figures 1–5 and Table 1; FACS gating strategy, tomographic slices, cryo-EM images, site-specific glycosylation comparisons between recombinant proteins, and values for glycoforms observed on S proteins derived from ChAdOx1 nCoV-19 (PDF)
Movie 1: Cryo-electron tomography of U2OS cells infected with ChAdOx1 nCoV-19 (MP4)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
Early work on producing ChAdOx1 nCoV-19 was supported by Funding from the Department of Health and Social Care (DHSC) managed by the Engineering and Physical Sciences Research Council (EPSRC) for the Future Vaccine Manufacturing Research Hub Grant Reference: EP/R013756/1. This work was supported by the International AIDS Vaccine Initiative (IAVI) through Grant INV-008352/OPP1153692 and the IAVI Neutralizing Antibody Center through the Collaboration for AIDS Vaccine Discovery Grant OPP1196345/INV-008813, both funded by the Bill and Melinda Gates Foundation. This work was also supported by the National Institute for Allergy and Infectious Diseases through the Scripps Consortium for HIV Vaccine Development (CHAVD) (AI144462), the University of Southampton Coronavirus Response Fund, the Bright Future Trust, the National Institute for Allergy and Infectious Diseases Grant AI150481, and the UK Wellcome Trust Investigator Award 206422/Z/17/Z. We acknowledge Diamond for access and support of the CryoEM facilities at the UK national electron bioimaging centre (eBIC, Proposal NR18477, NR21005, and NT21004), funded by the Wellcome Trust, MRC and BBSRC. The computational aspects of this research were supported by the Wellcome Trust Core Award Grant Number 203141/Z/16/Z and the NIHR Oxford BRC. C.G. is supported by Wellcome Award 090532/Z/09/Z. National Institute for Health Research Biomedical Research Centre Funding Scheme, the Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Science (CIFMS), China (Grant Number: 2018-I2M-2-002). G.R.S. is supported as a Wellcome Trust Senior Investigator (Grant 095541/A/11/Z).
References
This article references 51 other publications.
- 1Amanat, F.; Krammer, F. SARS-CoV-2 Vaccines: Status Report. Immunity. Cell Press: April 14, 2020; pp 583– 589. DOI: 10.1016/j.immuni.2020.03.007 .
- 2Letko, M.; Marzi, A.; Munster, V. Functional Assessment of Cell Entry and Receptor Usage for SARS-CoV-2 and Other Lineage B Betacoronaviruses. Nat. Microbiol. 2020, 1– 8, DOI: 10.1038/s41564-020-0688-y
- 3Wrapp, D.; Wang, N.; Corbett, K. S.; Goldsmith, J. A.; Hsieh, C.-L.; Abiona, O.; Graham, B. S.; McLellan, J. S. Cryo-EM Structure of the 2019-NCoV Spike in the Prefusion Conformation. Science 2020, 367, 1260, DOI: 10.1126/science.abb2507[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 4Walls, A. C.; Park, Y. J.; Tortorici, M. A.; Wall, A.; McGuire, A. T.; Veesler, D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020, 181 (2), 281– 292, e6 DOI: 10.1016/j.cell.2020.02.058[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVejsLk%253D&md5=ac8a8a208d9c26f88f702fb7634ab1abStructure, Function, and Antigenicity of the SARS-CoV-2 Spike GlycoproteinWalls, Alexandra C.; Park, Young-Jun; Tortorici, M. Alejandra; Wall, Abigail; McGuire, Andrew T.; Veesler, DavidCell (Cambridge, MA, United States) (2020), 181 (2), 281-292.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emergence of SARS-CoV-2 has resulted in >90,000 infections and >3000 deaths. Coronavirus spike (S) glycoproteins promote entry into cells and are the main target of antibodies. We show that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of SARS-CoV-2 S and SARS-CoV S bind with similar affinities to human ACE2, correlating with the efficient spread of SARS-CoV-2 among humans. We found that the SARS-CoV-2 S glycoprotein harbors a furin cleavage site at the boundary between the S1/S2 subunits, which is processed during biogenesis and sets this virus apart from SARS-CoV and SARS-related CoVs. We detd. cryo-EM structures of the SARS-CoV-2 S ectodomain trimer, providing a blueprint for the design of vaccines and inhibitors of viral entry. Finally, we demonstrate that SARS-CoV S murine polyclonal antibodies potently inhibited SARS-CoV-2 S mediated entry into cells, indicating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination.
- 5Watanabe, Y.; Allen, J. D.; Wrapp, D.; McLellan, J. S.; Crispin, M. Site-Specific Glycan Analysis of the SARS-CoV-2 Spike. Science 2020, eabb9983 DOI: 10.1126/science.abb9983 .
- 6Krammer, F. SARS-CoV-2 Vaccines in Development. Nature 2020, 1– 16, DOI: 10.1038/s41586-020-2798-3
- 7Polack, F. P.; Thomas, S. J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J. L.; Pérez Marc, G.; Moreira, E. D.; Zerbini, C. Safety and Efficacy of the BNT162b2MRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383 (27), 2603– 2615, DOI: 10.1056/NEJMoa2034577[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFSjuw%253D%253D&md5=08cb7204334ed37c7949dfcdfe5af75cSafety and efficacy of the BNT162b2 mRNA Covid-19 vaccinePolack, Fernando P.; Thomas, Stephen J.; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Perez, John L.; Marc, Gonzalo Perez; Moreira, Edson D.; Zerbini, Cristiano; Bailey, Ruth; Swanson, Kena A.; Roychoudhury, Satrajit; Koury, Kenneth; Li, Ping; Kalina, Warren V.; Cooper, David; Frenck, Robert W., Jr.; Hammitt, Laura L.; Tureci, Ozlem; Nell, Haylene; Schaefer, Axel; Unal, Serhat; Tresnan, Dina B.; Mather, Susan; Dormitzer, Philip R.; Sahin, Ugur; Jansen, Kathrin U.; Gruber, William C.New England Journal of Medicine (2020), 383 (27), 2603-2615CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)A review. Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a worldwide pandemic. Safe and effective vaccines are needed urgently. methods In an ongoing multinational, placebo-controlled, observer-blinded, pivotal efficacy trial, we randomly assigned persons 16 years of age or older in a 1:1 ratio to receive two doses, 21 days apart, of either placebo or the BNT162b2 vaccine candidate (30μg per dose). BNT162b2 is a lipid nanoparticle-formulated, nucleoside-modified RNA vaccine that encodes a prefusion stabilized, membrane-anchored SARS-CoV-2 full-length spike protein. The primary end points were efficacy of the vaccine against lab.-confirmed Covid-19 and safety. results A total of 43,548 participants underwent randomization, of whom 43,448 received injections: 21,720 with BNT162b2 and 21,728 with placebo. There were 8 cases of Covid-19 with onset at least 7 days after the second dose among participants assigned to receive BNT162b2 and 162 cases among those assigned to placebo; BNT162b2 was 95% effective in preventing Covid-19 (95% credible interval, 90.3 to 97.6). Similar vaccine efficacy (generally 90 to 100%) was obsd. across subgroups defined by age, sex, race, ethnicity, baseline body-mass index, and the presence of coexisting conditions. Among 10 cases of severe Covid-19 with onset after the first dose, 9 occurred in placebo recipients and 1 in a BNT162b2 recipient. The safety profile of BNT162b2 was characterized by short-term, mild-to-moderate pain at the injection site, fatigue, and headache. The incidence of serious adverse events was low and was similar in the vaccine and placebo groups. conclusions A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older. Safety over a median of 2 mo was similar to that of other viral vaccines.
- 8Corbett, K. S.; Flynn, B.; Foulds, K. E.; Francica, J. R.; Boyoglu-Barnum, S.; Werner, A. P.; Flach, B.; O’Connell, S.; Bock, K. W.; Minai, M. Evaluation of the MRNA-1273 Vaccine against SARS-CoV-2 in Nonhuman Primates. N. Engl. J. Med. 2020, 383, 1544, DOI: 10.1056/NEJMoa2024671[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFWmsr7I&md5=8ecba6b18bc615d886ca5016b7c85496Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primatesCorbett, K. S.; Flynn, B.; Foulds, K. E.; Francica, J. R.; Boyoglu-Barnum, S.; Werner, A. P.; Flach, B.; O'Connell, S.; Bock, K. W.; Minai, M.; Nagata, B. M.; Andersen, H.; Martinez, D. R.; Noe, A. T.; Douek, N.; Donaldson, M. M.; Nji, N. N.; Alvarado, G. S.; Edwards, D. K.; Flebbe, D. R.; Lamb, E.; Doria-Rose, N. A.; Lin, B. C.; Louder, M. K.; O'Dell, S.; Schmidt, S. D.; Phung, E.; Chang, L. A.; Yap, C.; Todd, J.-P. M.; Pessaint, L.; Van Ry, A.; Browne, S.; Greenhouse, J.; Putman-Taylor, T.; Strasbaugh, A.; Campbell, T.-A.; Cook, A.; Dodson, A.; Steingrebe, K.; Shi, W.; Zhang, Y.; Abiona, O. M.; Wang, L.; Pegu, A.; Yang, E. S.; Leung, K.; Zhou, T.; Teng, I-T.; Widge, A.; Gordon, I.; Novik, L.; Gillespie, R. A.; Loomis, R. J.; Moliva, J. I.; Stewart-Jones, G.; Himansu, S.; Kong, W.-P.; Nason, M. C.; Morabito, K. M.; Ruckwardt, T. J.; Ledgerwood, J. E.; Gaudinski, M. R.; Kwong, P. D.; Mascola, J. R.; Carfi, A.; Lewis, M. G.; Baric, R. S.; McDermott, A.; Moore, I. N.; Sullivan, N. J.; Roederer, M.; Seder, R. A.; Graham, B. S.New England Journal of Medicine (2020), 383 (16), 1544-1555CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Background: Vaccines to prevent coronavirus disease 2019 (Covid-19) are urgently needed. The effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines on viral replication in both upper and lower airways is important to evaluate in nonhuman primates. Methods: Nonhuman primates received 10 or 100μg of mRNA-1273, a vaccine encoding the prefusion-stabilized spike protein of SARS-CoV-2, or no vaccine. Antibody and T-cell responses were assessed before upper- and lower-airway challenge with SARS-CoV-2. Active viral replication and viral genomes in bronchoalveolar-lavage (BAL) fluid and nasal swab specimens were assessed by polymerase chain reaction, and histopathol. anal. and viral quantification were performed on lung-tissue specimens. Results The mRNA-1273 vaccine candidate induced antibody levels exceeding those in human convalescent-phase serum, with live-virus reciprocal 50% inhibitory diln. (ID50) geometric mean titers of 501 in the 10-μg dose group and 3481 in the 100-μg dose group. Vaccination induced type 1 helper T-cell (Th1)-biased CD4 T-cell responses and low or undetectable Th2 or CD8 T-cell responses. Viral replication was not detectable in BAL fluid by day 2 after challenge in seven of eight animals in both vaccinated groups. No viral replication was detectable in the nose of any of the eight animals in the 100-μg dose group by day 2 after challenge, and limited inflammation or detectable viral genome or antigen was noted in lungs of animals in either vaccine group. conclusions Vaccination of nonhuman primates with mRNA-1273 induced robust SARS-CoV-2 neutralizing activity, rapid protection in the upper and lower airways, and no pathol. changes in the lung.
- 9Gao, Q.; Bao, L.; Mao, H.; Wang, L.; Xu, K.; Yang, M.; Li, Y.; Zhu, L.; Wang, N.; Lv, Z. Development of an Inactivated Vaccine Candidate for SARS-CoV-2. Science 2020, 369 (6499), 77– 81, DOI: 10.1126/science.abc1932[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlCmtL3P&md5=674788246758fd31fa8bb54c936be83aDevelopment of an inactivated vaccine candidate for SARS-CoV-2Gao, Qiang; Bao, Linlin; Mao, Haiyan; Wang, Lin; Xu, Kangwei; Yang, Minnan; Li, Yajing; Zhu, Ling; Wang, Nan; Lv, Zhe; Gao, Hong; Ge, Xiaoqin; Kan, Biao; Hu, Yaling; Liu, Jiangning; Cai, Fang; Jiang, Deyu; Yin, Yanhui; Qin, Chengfeng; Li, Jing; Gong, Xuejie; Lou, Xiuyu; Shi, Wen; Wu, Dongdong; Zhang, Hengming; Zhu, Lang; Deng, Wei; Li, Yurong; Lu, Jinxing; Li, Changgui; Wang, Xiangxi; Yin, Weidong; Zhang, Yanjun; Qin, ChuanScience (Washington, DC, United States) (2020), 369 (6499), 77-81CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in an unprecedented public health crisis. Because of the novelty of the virus, there are currently no SARS-CoV-2-specific treatments or vaccines available. Therefore, rapid development of effective vaccines against SARS-CoV-2 are urgently needed. Here, we developed a pilot-scale prodn. of PiCoVacc, a purified inactivated SARS-CoV-2 virus vaccine candidate, which induced SARS-CoV-2-specific neutralizing antibodies in mice, rats, and nonhuman primates. These antibodies neutralized 10 representative SARS-CoV-2 strains, suggesting a possible broader neutralizing ability against other strains. Three immunizations using two different doses, 3 or 6μg per dose, provided partial or complete protection in macaques against SARS-CoV-2 challenge, resp., without observable antibody-dependent enhancement of infection. These data support the clin. development and testing of PiCoVacc for use in humans.
- 10Liu, C.; Mendonæa, L.; Yang, Y.; Gao, Y.; Shen, C.; Liu, J.; Ni, T.; Ju, B.; Liu, C.; Tang, X. The Architecture of Inactivated SARS-CoV-2 with Postfusion Spikes Revealed by Cryo-EM and Cryo-ET. Structure 2020, 28, 1218, DOI: 10.1016/j.str.2020.10.001[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVylsb3I&md5=79c3780b2fb235c85d39a7cc855ada50The Architecture of Inactivated SARS-CoV-2 with Postfusion Spikes Revealed by Cryo-EM and Cryo-ETLiu, Chuang; Mendonca, Luiza; Yang, Yang; Gao, Yuanzhu; Shen, Chenguang; Liu, Jiwei; Ni, Tao; Ju, Bin; Liu, Congcong; Tang, Xian; Wei, Jinli; Ma, Xiaomin; Zhu, Yanan; Liu, Weilong; Xu, Shuman; Liu, Yingxia; Yuan, Jing; Wu, Jing; Liu, Zheng; Zhang, Zheng; Liu, Lei; Wang, Peiyi; Zhang, PeijunStructure (Oxford, United Kingdom) (2020), 28 (11), 1218-1224.e4CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)The ongoing global pandemic of coronavirus disease 2019 (COVID-19) resulted from the outbreak of SARS-CoV-2 in Dec. 2019. Currently, multiple efforts are being made to rapidly develop vaccines and treatments to fight COVID-19. Current vaccine candidates use inactivated SARS-CoV-2 viruses; therefore, it is important to understand the architecture of inactivated SARS-CoV-2. We have genetically and structurally characterized β-propiolactone-inactivated viruses from a propagated and purified clin. strain of SARS-CoV-2. We obsd. that the virus particles are roughly spherical or moderately pleiomorphic. Although a small fraction of prefusion spikes are found, most spikes appear nail shaped, thus resembling a postfusion state, where the S1 protein of the spike has disassocd. from S2. Cryoelectron tomog. and subtomogram averaging of these spikes yielded a d. map that closely matches the overall structure of the SARS-CoV postfusion spike and its corresponding glycosylation site. Our findings have major implications for SARS-CoV-2 vaccine design, esp. those using inactivated viruses.
- 11Zost, S. J.; Gilchuk, P.; Case, J. B.; Binshtein, E.; Chen, R. E.; Nkolola, J. P.; Schäfer, A.; Reidy, J. X.; Trivette, A.; Nargi, R. S. Potently Neutralizing and Protective Human Antibodies against SARS-CoV-2. Nature 2020, 584 (7821), 443– 449, DOI: 10.1038/s41586-020-2548-6[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsF2ntLrM&md5=d39be143814010bd4eacc7b69f2e85cbPotently neutralizing and protective human antibodies against SARS-CoV-2Zost, Seth J.; Gilchuk, Pavlo; Case, James Brett; Binshtein, Elad; Chen, Rita E.; Nkolola, Joseph P.; Schafer, Alexandra; Reidy, Joseph X.; Trivette, Andrew; Nargi, Rachel S.; Sutton, Rachel E.; Suryadevara, Naveenchandra; Martinez, David R.; Williamson, Lauren E.; Chen, Elaine C.; Jones, Taylor; Day, Samuel; Myers, Luke; Hassan, Ahmed O.; Kafai, Natasha M.; Winkler, Emma S.; Fox, Julie M.; Shrihari, Swathi; Mueller, Benjamin K.; Meiler, Jens; Chandrashekar, Abishek; Mercado, Noe B.; Steinhardt, James J.; Ren, Kuishu; Loo, Yueh-Ming; Kallewaard, Nicole L.; McCune, Broc T.; Keeler, Shamus P.; Holtzman, Michael J.; Barouch, Dan H.; Gralinski, Lisa E.; Baric, Ralph S.; Thackray, Larissa B.; Diamond, Michael S.; Carnahan, Robert H.; Crowe Jr, James E.Nature (London, United Kingdom) (2020), 584 (7821), 443-449CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The ongoing pandemic of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major threat to global health and the medical countermeasures available so far are limited. Moreover, we currently lack a thorough understanding of the mechanisms of humoral immunity to SARS-CoV-2. We analyze a large panel of human monoclonal antibodies that target the spike (S) glycoprotein, and identify several that exhibit potent neutralizing activity and fully block the receptor-binding domain of the S protein (SRBD) from interacting with human angiotensin-converting enzyme 2 (ACE2). Using competition-binding, structural, and functional studies, we show that the monoclonal antibodies can be clustered into classes that recognize distinct epitopes on the SRBD, as well as distinct conformational states of the S trimer. Two potently neutralizing monoclonal antibodies, COV2-2196 and COV2-2130, which recognize non-overlapping sites, bound simultaneously to the S protein and neutralized wild-type SARS-CoV-2 virus in a synergistic manner. In 2 mouse models of SARS-CoV-2 infection, passive transfer of COV2-2196, COV2-2130, or a combination of both of these antibodies protected mice from wt. loss and reduced the viral burden and levels of inflammation in the lungs. In addn., passive transfer of either of 2 of the most potent ACE2-blocking monoclonal antibodies (COV2-2196 or COV2-2381) as monotherapy protected rhesus macaques from SARS-CoV-2 infection. These results identify protective epitopes on the SRBD and provide a structure-based framework for rational vaccine design and the selection of robust immunotherapeutic agents.
- 12Robbiani, D. F.; Gaebler, C.; Muecksch, F.; Lorenzi, J. C. C.; Wang, Z.; Cho, A.; Agudelo, M.; Barnes, C. O.; Gazumyan, A.; Finkin, S. Convergent Antibody Responses to SARS-CoV-2 in Convalescent Individuals. Nature 2020, 584 (7821), 437– 442, DOI: 10.1038/s41586-020-2456-9[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFegtb3P&md5=952577f69ffe8282d0b2451b17a5ecadConvergent antibody responses to SARS-CoV-2 in convalescent individualsRobbiani, Davide F.; Gaebler, Christian; Muecksch, Frauke; Lorenzi, Julio C. C.; Wang, Zijun; Cho, Alice; Agudelo, Marianna; Barnes, Christopher O.; Gazumyan, Anna; Finkin, Shlomo; Hagglof, Thomas; Oliveira, Thiago Y.; Viant, Charlotte; Hurley, Arlene; Hoffmann, Hans-Heinrich; Millard, Katrina G.; Kost, Rhonda G.; Cipolla, Melissa; Gordon, Kristie; Bianchini, Filippo; Chen, Spencer T.; Ramos, Victor; Patel, Roshni; Dizon, Juan; Shimeliovich, Irina; Mendoza, Pilar; Hartweger, Harald; Nogueira, Lilian; Pack, Maggi; Horowitz, Jill; Schmidt, Fabian; Weisblum, Yiska; Michailidis, Eleftherios; Ashbrook, Alison W.; Waltari, Eric; Pak, John E.; Huey-Tubman, Kathryn E.; Koranda, Nicholas; Hoffman, Pauline R.; West Jr, Anthony P.; Rice, Charles M.; Hatziioannou, Theodora; Bjorkman, Pamela J.; Bieniasz, Paul D.; Caskey, Marina; Nussenzweig, Michel C.Nature (London, United Kingdom) (2020), 584 (7821), 437-442CODEN: NATUAS; ISSN:0028-0836. (Nature Research)During the coronavirus disease-2019 (COVID-19) pandemic, severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has led to the infection of millions of people and has claimed hundreds of thousands of lives. The entry of the virus into cells depends on the receptor-binding domain (RBD) of the spike (S) protein of SARS-CoV-2. Although there is currently no vaccine, it is likely that antibodies will be essential for protection. However, little is known about the human antibody response to SARS-CoV-2. We report on 149 COVID-19-convalescent individuals. Plasma samples collected an av. of 39 days after the onset of symptoms had variable half-maximal pseudovirus neutralizing titers; titers were <50 in 33% of samples, <1000 in 79% of samples, and only 1% of samples had titers >5000. Antibody sequencing revealed the expansion of clones of RBD-specific memory B cells that expressed closely related antibodies in different individuals. Despite low plasma titers, antibodies to 3 distinct epitopes on the RBD neutralized the virus with half-maximal inhibitory concns. (IC50 values) as low as 2 ng/mL. In conclusion, most convalescent plasma samples obtained from individuals who recover from COVID-19 do not contain high levels of neutralizing activity. Nevertheless, rare but recurring RBD-specific antibodies with potent antiviral activity were found in all individuals tested, suggesting that a vaccine designed to elicit such antibodies could be broadly effective.
- 13Du, S.; Cao, Y.; Zhu, Q.; Yu, P.; Qi, F.; Wang, G.; Du, X.; Bao, L.; Deng, W.; Zhu, H. Structurally Resolved SARS-CoV-2 Antibody Shows High Efficacy in Severely Infected Hamsters and Provides a Potent Cocktail Pairing Strategy. Cell 2020, 183, 1013, DOI: 10.1016/j.cell.2020.09.035[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFGqsLvJ&md5=230461817cefd12ef9eccbe6d6aa6a73Structurally resolved SARS-CoV-2 antibody shows high efficacy in severely infected hamsters and provides a potent cocktail pairing strategyDu, Shuo; Cao, Yunlong; Zhu, Qinyu; Yu, Pin; Qi, Feifei; Wang, Guopeng; Du, Xiaoxia; Bao, Linlin; Deng, Wei; Zhu, Hua; Liu, Jiangning; Nie, Jianhui; Zheng, Yinghui; Liang, Haoyu; Liu, Ruixue; Gong, Shuran; Xu, Hua; Yisimayi, Ayijiang; Lv, Qi; Wang, Bo; He, Runsheng; Han, Yunlin; Zhao, Wenjie; Bai, Yali; Qu, Yajin; Gao, Xiang; Ji, Chenggong; Wang, Qisheng; Gao, Ning; Huang, Weijin; Wang, Youchun; Xie, X. Sunney; Su, Xiao-dong; Xiao, Junyu; Qin, ChuanCell (Cambridge, MA, United States) (2020), 183 (4), 1013-1023.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Understanding how potent neutralizing antibodies (NAbs) inhibit SARS-CoV-2 is crit. for effective therapeutic development. We previously described BD-368-2, a SARS-CoV-2 NAb with high potency; however, its neutralization mechanism is largely unknown. Here, we report the 3.5-Å cryo-EM structure of BD-368-2/trimeric-spike complex, revealing that BD-368-2 fully blocks ACE2 recognition by occupying all three receptor-binding domains (RBDs) simultaneously, regardless of their "up" or "down" conformations. Also, BD-368-2 treats infected adult hamsters at low dosages and at various administering windows, in contrast to placebo hamsters that manifested severe interstitial pneumonia. Moreover, BD-368-2's epitope completely avoids the common binding site of VH3-53/VH3-66 recurrent NAbs, evidenced by tripartite co-crystal structures with RBDs. Pairing BD-368-2 with a potent recurrent NAb neutralizes SARS-CoV-2 pseudovirus at pM level and rescues mutation-induced neutralization escapes. Together, our results rationalized a new RBD epitope that leads to high neutralization potency and demonstrated BD-368-2's therapeutic potential in treating COVID-19.
- 14Barnes, C. O.; Jette, C. A.; Abernathy, M. E.; Dam, K.-M. A.; Esswein, S. R.; Gristick, H. B.; Malyutin, A. G.; Sharaf, N. G.; Huey-Tubman, K. E.; Lee, Y. E. SARS-CoV-2 Neutralizing Antibody Structures Inform Therapeutic Strategies. Nature 2020, 1– 9, DOI: 10.1038/s41586-020-2852-1
- 15Brouwer, P. J. M.; Caniels, T. G.; van der Straten, K.; Snitselaar, J. L.; Aldon, Y.; Bangaru, S.; Torres, J. L.; Okba, N. M. A.; Claireaux, M.; Kerster, G. Potent Neutralizing Antibodies from COVID-19 Patients Define Multiple Targets of Vulnerability. Science 2020, eabc5902 DOI: 10.1126/science.abc5902
- 16Folegatti, P. M.; Ewer, K. J.; Aley, P. K.; Angus, B.; Becker, S.; Belij-Rammerstorfer, S.; Bellamy, D.; Bibi, S.; Bittaye, M.; Clutterbuck, E. A.; Safety and Immunogenicity of the ChAdOx1 NCoV-19 Vaccine against SARS-CoV-2: A Preliminary Report of a Phase 1/2, Single-Blind, Randomised Controlled Trial. Lancet 2020, 0 (0). DOI: 10.1016/S0140-6736(20)31604-4 .
- 17van Doremalen, N.; Lambe, T.; Spencer, A.; Belij-Rammerstorfer, S.; Purushotham, J. N.; Port, J. R.; Avanzato, V. A.; Bushmaker, T.; Flaxman, A.; Ulaszewska, M. ChAdOx1 NCoV-19 Vaccine Prevents SARS-CoV-2 Pneumonia in Rhesus Macaques. Nature 2020, 1– 5, DOI: 10.1038/s41586-020-2608-y
- 18Graham, S. P.; McLean, R. K.; Spencer, A. J.; Belij-Rammerstorfer, S.; Wright, D.; Ulaszewska, M.; Edwards, J. C.; Hayes, J. W. P.; Martini, V.; Thakur, N. Evaluation of the Immunogenicity of Prime-Boost Vaccination with the Replication-Deficient Viral Vectored COVID-19 Vaccine Candidate ChAdOx1 NCoV-19. npj Vaccines 2020, 5 (1), 1– 6, DOI: 10.1038/s41541-020-00221-3
- 19Ramasamy, M. N.; Minassian, A. M.; Ewer, K. J.; Flaxman, A. L.; Folegatti, P. M.; Owens, D. R.; Voysey, M.; Aley, P. K.; Angus, B.; Babbage, G.; Safety and Immunogenicity of ChAdOx1 NCoV-19 Vaccine Administered in a Prime-Boost Regimen in Young and Old Adults (COV002): A Single-Blind, Randomised, Controlled, Phase 2/3 Trial. Lancet 2020, 0 (0). DOI: 10.1016/s0140-6736(20)32466-1 .
- 20Sebastian, S.; Flaxman, A.; Cha, K. M.; Ulaszewska, M.; Gilbride, C.; Sharpe, H.; Wright, E.; Spencer, A. J.; Dowall, S.; Hewson, R.; A Multi-Filovirus Vaccine Candidate: Co-Expression of Ebola, Sudan, and Marburg Antigens in a Single Vector. Vaccines 2020, 8 (2). DOI: 10.3390/vaccines8020241 .
- 21Zhang, L.; Jackson, C. B.; Mou, H.; Ojha, A.; Peng, H.; Quinlan, B. D.; Rangarajan, E. S.; Pan, A.; Vanderheiden, A.; Suthar, M. S. SARS-CoV-2 Spike-Protein D614G Mutation Increases Virion Spike Density and Infectivity. Nat. Commun. 2020, 11 (1), 1– 9, DOI: 10.1038/s41467-020-19808-4
- 22Pallesen, J.; Wang, N.; Corbett, K. S.; Wrapp, D.; Kirchdoerfer, R. N.; Turner, H. L.; Cottrell, C. A.; Becker, M. M.; Wang, L.; Shi, W. Immunogenicity and Structures of a Rationally Designed Prefusion MERS-CoV Spike Antigen. Proc. Natl. Acad. Sci. U. S. A. 2017, 114 (35), E7348– E7357, DOI: 10.1073/pnas.1707304114[Crossref], [PubMed], [CAS], Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlWmsrfI&md5=3054f8054d852ed4192fd8f1fb6c870dImmunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigenPallesen, Jesper; Wang, Nianshuang; Corbett, Kizzmekia S.; Wrapp, Daniel; Kirchdoerfer, Robert N.; Turner, Hannah L.; Cottrell, Christopher A.; Becker, Michelle M.; Wang, Lingshu; Shi, Wei; Kong, Wing-Pui; Andres, Erica L.; Kettenbach, Arminja N.; Denison, Mark R.; Chappell, James D.; Graham, Barney S.; Ward, Andrew B.; McLellan, Jason S.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (35), E7348-E7357CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) is a lineage C betacoronavirus that since its emergence in 2012 has caused outbreaks in human populations with case-fatality rates of ∼36%. As in other coronaviruses, the spike (S) glycoprotein of MERS-CoV mediates receptor recognition and membrane fusion and is the primary target of the humoral immune response during infection. Here we use structure-based design to develop a generalizable strategy for retaining coronavirus S proteins in the antigenically optimal prefusion conformation and demonstrate that our engineered immunogen is able to elicit high neutralizing antibody titers against MERS-CoV. We also detd. high-resoln. structures of the trimeric MERS-CoV S ectodomain in complex with G4, a stem-directed neutralizing antibody. The structures reveal that G4 recognizes a glycosylated loop that is variable among coronaviruses and they define four conformational states of the trimer wherein each receptor-binding domain is either tightly packed at the membrane-distal apex or rotated into a receptor-accessible conformation. Our studies suggest a potential mechanism for fusion initiation through sequential receptor-binding events and provide a foundation for the structure-based design of coronavirus vaccines.
- 23Walls, A. C.; Tortorici, M. A.; Snijder, J.; Xiong, X.; Bosch, B. J.; Rey, F. A.; Veesler, D. Tectonic Conformational Changes of a Coronavirus Spike Glycoprotein Promote Membrane Fusion. Proc. Natl. Acad. Sci. U. S. A. 2017, 114 (42), 11157– 11162, DOI: 10.1073/pnas.1708727114[Crossref], [PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1SksLjI&md5=a5b64abf0a761b46c0098e63f57369c4Tectonic conformational changes of a coronavirus spike glycoprotein promote membrane fusionWalls, Alexandra C.; Tortorici, M. Alejandra; Snijder, Joost; Xiong, Xiaoli; Bosch, Berend-Jan; Rey, Felix A.; Veesler, DavidProceedings of the National Academy of Sciences of the United States of America (2017), 114 (42), 11157-11162CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The tremendous pandemic potential of coronaviruses was demonstrated twice in the past few decades by 2 global outbreaks of deadly pneumonia. The coronavirus spike (S) glycoprotein initiates infection by promoting fusion of the viral and cellular membranes through conformational changes that remain largely uncharacterized. Here, we report the cryo-electron microscopy (cryo-EM) structure of a coronavirus S glycoprotein in the post-fusion state, showing large-scale secondary, tertiary, and quaternary rearrangements compared with the pre-fusion trimer and rationalizing the free-energy landscape of this conformational machine. We also biochem. characterized the mol. events assocd. with refolding of the metastable pre-fusion S glycoprotein to the post-fusion conformation using limited proteolysis, mass spectrometry, and single-particle EM. The obsd. similarity between post-fusion coronavirus S and paramyxovirus F structures demonstrated that a conserved refolding trajectory mediates entry of these viruses and supports the evolutionary relatedness of their fusion subunits. Finally, our data provide a structural framework for understanding the mode of neutralization of antibodies targeting the fusion machinery and for engineering next-generation subunit vaccines or inhibitors against this medically important virus family.
- 24Berger, I.; Schaffitzel, C. The SARS-CoV-2 Spike Protein: Balancing Stability and Infectivity. Cell Research; Springer Nature: November 2, 2020; pp 1059– 1060. DOI: 10.1038/s41422-020-00430-4 .
- 25Himes, B. A.; Zhang, P. EmClarity: Software for High-Resolution Cryo-Electron Tomography and Subtomogram Averaging. Nat. Methods 2018, 15 (11), 955– 961, DOI: 10.1038/s41592-018-0167-z[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvF2lsbrE&md5=1358ebf4bdbb9e14df75ffafec9495ebemClarity: software for high-resolution cryo-electron tomography and subtomogram averagingHimes, Benjamin A.; Zhang, PeijunNature Methods (2018), 15 (11), 955-961CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)Macromol. complexes are intrinsically flexible and often challenging to purify for structure detn. by single-particle cryo-electron microscopy (cryo-EM). Such complexes can be studied by cryo-electron tomog. (cryo-ET) combined with subtomogram alignment and classification, which in exceptional cases achieves subnanometer resoln., yielding insight into structure-function relationships. However, it remains challenging to apply this approach to specimens that exhibit conformational or compositional heterogeneity or are present in low abundance. To address this, we developed emClarity (https://github.com/bHimes/emClarity/wiki), a GPU-accelerated image-processing package featuring an iterative tomog. tilt-series refinement algorithm that uses subtomograms as fiducial markers and a 3D-sampling-function-compensated, multi-scale principal component anal. classification method. We demonstrate that our approach offers substantial improvement in the resoln. of maps and in the sepn. of different functional states of macromol. complexes compared with current state-of-the-art software.
- 26Turoňová, B.; Sikora, M.; Schürmann, C.; Hagen, W.; Welsch, S.; Blanc, F.; von Bülow, S.; Gecht, M.; Bagola, K.; Hörner, C.; In Situ Structural Analysis of SARS-CoV-2 Spike Reveals Flexibility Mediated by Three Hinges. bioRxiv 2020, DOI: 10.1101/2020.06.26.173476 .
- 27Yao, H.; Song, Y.; Chen, Y.; Wu, N.; Xu, J.; Sun, C.; Zhang, J.; Weng, T.; Zhang, Z.; Wu, Z. Molecular Architecture of the SARS-CoV-2 Virus. Cell 2020, 183, 730, DOI: 10.1016/j.cell.2020.09.018[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFKjs7nM&md5=7bd110334cb42931034a4eb5f9d2ff00Molecular Architecture of the SARS-CoV-2 VirusYao, Hangping; Song, Yutong; Chen, Yong; Wu, Nanping; Xu, Jialu; Sun, Chujie; Zhang, Jiaxing; Weng, Tianhao; Zhang, Zheyuan; Wu, Zhigang; Cheng, Linfang; Shi, Danrong; Lu, Xiangyun; Lei, Jianlin; Crispin, Max; Shi, Yigong; Li, Lanjuan; Li, SaiCell (Cambridge, MA, United States) (2020), 183 (3), 730-738.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)SARS-CoV-2 is an enveloped virus responsible for the COVID-19 pandemic. Despite recent advances in the structural elucidation of SARS-CoV-2 proteins, the detailed architecture of the intact virus remains to be unveiled. Here we report the mol. assembly of the authentic SARS-CoV-2 virus using cryoelectron tomog. (cryo-ET) and subtomogram averaging (STA). Native structures of the S proteins in pre- and postfusion conformations were detd. to av. resolns. of 8.7-11 Å. Compns. of the N-linked glycans from the native spikes were analyzed by mass spectrometry, which revealed overall processing states of the native glycans highly similar to that of the recombinant glycoprotein glycans. The native conformation of the ribonucleoproteins (RNPs) and their higher-order assemblies were revealed. Overall, these characterizations revealed the architecture of the SARS-CoV-2 virus in exceptional detail and shed light on how the virus packs its ∼30-kb-long single-segmented RNA in the ∼80-nm-diam. lumen.
- 28Ke, Z.; Oton, J.; Qu, K.; Cortese, M.; Zila, V.; McKeane, L.; Nakane, T.; Zivanov, J.; Neufeldt, C. J.; Cerikan, B. Structures and Distributions of SARS-CoV-2 Spike Proteins on Intact Virions. Nature 2020, 1– 5, DOI: 10.1038/s41586-020-2665-2
- 29Toelzer, C.; Gupta, K.; Yadav, S. K. N.; Borucu, U.; Davidson, A. D.; Kavanagh Williamson, M.; Shoemark, D. K.; Garzoni, F.; Staufer, O.; Milligan, R. Free Fatty Acid Binding Pocket in the Locked Structure of SARS-CoV-2 Spike Protein. Science 2020, 370 (6517), 725– 730, DOI: 10.1126/science.abd3255[Crossref], [PubMed], [CAS], Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1Kqsr7I&md5=ab0610e4c61dd1cbf7500004cee9bd30Free fatty acid binding pocket in the locked structure of SARS-CoV-2 spike proteinToelzer, Christine; Gupta, Kapil; Yadav, Sathish K. N.; Borucu, Ufuk; Davidson, Andrew D.; Kavanagh Williamson, Maia; Shoemark, Deborah K.; Garzoni, Frederic; Staufer, Oskar; Milligan, Rachel; Capin, Julien; Mulholland, Adrian J.; Spatz, Joachim; Fitzgerald, Daniel; Berger, Imre; Schaffitzel, ChristianeScience (Washington, DC, United States) (2020), 370 (6517), 725-730CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents a global crisis. Key to SARS-CoV-2 therapeutic development is unraveling the mechanisms that drive high infectivity, broad tissue tropism, and severe pathol. Our 2.85-angstrom cryo-electron microscopy structure of SARS-CoV-2 spike (S) glycoprotein reveals that the receptor-binding domains tightly bind the essential free fatty acid linoleic acid (LA) in 3 composite binding pockets. A similar pocket also appears to be present in the highly pathogenic severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). LA binding stabilizes a locked S conformation, resulting in reduced angiotensin-converting enzyme 2 (ACE2) interaction in vitro. In human cells, LA supplementation synergizes with the COVID-19 drug remdesivir, suppressing SARS-CoV-2 replication. Our structure directly links LA and S, setting the stage for intervention strategies that target LA binding by SARS-CoV-2.
- 30Watanabe, Y.; Bowden, T. A.; Wilson, I. A.; Crispin, M. Exploitation of Glycosylation in Enveloped Virus Pathobiology. Biochim. Biophys. Acta, Gen. Subj. 2019, 1863 (10), 1480– 1497, DOI: 10.1016/j.bbagen.2019.05.012[Crossref], [PubMed], [CAS], Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtVynsbvE&md5=118babe3b318937a35757e0cd5b57521Exploitation of glycosylation in enveloped virus pathobiologyWatanabe, Yasunori; Bowden, Thomas A.; Wilson, Ian A.; Crispin, MaxBiochimica et Biophysica Acta, General Subjects (2019), 1863 (10), 1480-1497CODEN: BBGSB3; ISSN:0304-4165. (Elsevier B.V.)A review. Glycosylation is a ubiquitous post-translational modification responsible for a multitude of crucial biol. roles. As obligate parasites, viruses exploit host-cell machinery to glycosylate their own proteins during replication. Viral envelope proteins from a variety of human pathogens including HIV-1, influenza virus, Lassa virus, SARS, Zika virus, dengue virus, and Ebola virus have evolved to be extensively glycosylated. These host-cell derived glycans facilitate diverse structural and functional roles during the viral life-cycle, ranging from immune evasion by glycan shielding to enhancement of immune cell infection. In this review, we highlight the imperative and auxiliary roles glycans play, and how specific oligosaccharide structures facilitate these functions during viral pathogenesis. We discuss the growing efforts to exploit viral glycobiol. in the development of anti-viral vaccines and therapies.
- 31Cao, L.; Pauthner, M.; Andrabi, R.; Rantalainen, K.; Berndsen, Z.; Diedrich, J. K.; Menis, S.; Sok, D.; Bastidas, R.; Park, S.-K. R. Differential Processing of HIV Envelope Glycans on the Virus and Soluble Recombinant Trimer. Nat. Commun. 2018, 9 (1), 3693, DOI: 10.1038/s41467-018-06121-4[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c3ot1aksA%253D%253D&md5=d4daee53f36dffe65dff73789b3bf39eDifferential processing of HIV envelope glycans on the virus and soluble recombinant trimerCao Liwei; Diedrich Jolene K; Park Sung-Kyu Robin; Delahunty Claire M; He Lin; Yates John R 3rd; Paulson James C; Cao Liwei; Pauthner Matthias; Andrabi Raiees; Menis Sergey; Sok Devin; Bastidas Raiza; Guenaga Javier; Wyatt Richard T; Schief William R; Burton Dennis R; Paulson James C; Cao Liwei; Pauthner Matthias; Andrabi Raiees; Rantalainen Kimmo; Berndsen Zachary; Menis Sergey; Sok Devin; Bastidas Raiza; Guenaga Javier; Wyatt Richard T; Schief William R; Ward Andrew B; Burton Dennis R; Paulson James C; Rantalainen Kimmo; Berndsen Zachary; Ward Andrew B; Burton Dennis RNature communications (2018), 9 (1), 3693 ISSN:.As the sole target of broadly neutralizing antibodies (bnAbs) to HIV, the envelope glycoprotein (Env) trimer is the focus of vaccination strategies designed to elicit protective bnAbs in humans. Because HIV Env is densely glycosylated with 75-90 N-glycans per trimer, most bnAbs use or accommodate them in their binding epitope, making the glycosylation of recombinant Env a key aspect of HIV vaccine design. Upon analysis of three HIV strains, we here find that site-specific glycosylation of Env from infectious virus closely matches Envs from corresponding recombinant membrane-bound trimers. However, viral Envs differ significantly from recombinant soluble, cleaved (SOSIP) Env trimers, strongly impacting antigenicity. These results provide a benchmark for virus Env glycosylation needed for the design of soluble Env trimers as part of an overall HIV vaccine strategy.
- 32Cao, L.; Diedrich, J. K.; Kulp, D. W.; Pauthner, M.; He, L.; Park, S.-K. R.; Sok, D.; Su, C. Y.; Delahunty, C. M.; Menis, S. Global Site-Specific N-Glycosylation Analysis of HIV Envelope Glycoprotein. Nat. Commun. 2017, 8, 14954, DOI: 10.1038/ncomms14954[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlt12nu7w%253D&md5=eb9e80327cbc6e95a3c8ee602e7c14cdGlobal site-specific N-glycosylation analysis of HIV envelope glycoproteinCao, Liwei; Diedrich, Jolene K.; Kulp, Daniel W.; Pauthner, Matthias; He, Lin; Park, Sung-Kyu Robin; Sok, Devin; Su, Ching Yao; Delahunty, Claire M.; Menis, Sergey; Andrabi, Raiees; Guenaga, Javier; Georgeson, Erik; Kubitz, Michael; Adachi, Yumiko; Burton, Dennis R.; Schief, William R.; Yates, John R., III; Paulson, James C.Nature Communications (2017), 8 (), 14954CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)HIV-1 envelope glycoprotein (Env) is the sole target for broadly neutralizing antibodies (bnAbs) and the focus for design of an antibody-based HIV vaccine. The Env trimer is covered by ∼90N-linked glycans, which shield the underlying protein from immune surveillance. BNAbs to HIV develop during infection, with many showing dependence on glycans for binding to Env. The ability to routinely assess the glycan type at each glycosylation site may facilitate design of improved vaccine candidates. Here we present a general mass spectrometry-based proteomics strategy that uses specific endoglycosidases to introduce mass signatures that distinguish peptide glycosites that are unoccupied or occupied by high-mannose/hybrid or complex-type glycans. The method yields >95% sequence coverage for Env, provides semi-quant. anal. of the glycosylation status at each glycosite. We find that most glycosites in recombinant Env trimers are fully occupied by glycans, varying in the proportion of high-mannose/hybrid and complex-type glycans.
- 33Struwe, W. B.; Chertova, E.; Allen, J. D.; Seabright, G. E.; Watanabe, Y.; Harvey, D. J.; Medina-Ramirez, M.; Roser, J. D.; Smith, R.; Westcott, D. Site-Specific Glycosylation of Virion-Derived HIV-1 Env Is Mimicked by a Soluble Trimeric Immunogen. Cell Rep. 2018, 24 (8), 1958– 1966, e5 DOI: 10.1016/j.celrep.2018.07.080[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFGqtLzL&md5=2113b0b4d0702bc1cb54ade8dfc1b0bbSite-Specific Glycosylation of Virion-Derived HIV-1 Env Is Mimicked by a Soluble Trimeric ImmunogenStruwe, Weston B.; Chertova, Elena; Allen, Joel D.; Seabright, Gemma E.; Watanabe, Yasunori; Harvey, David J.; Medina-Ramirez, Max; Roser, James D.; Smith, Rodman; Westcott, David; Keele, Brandon F.; Bess, Julian W. Jr.; Sanders, Rogier W.; Lifson, Jeffrey D.; Moore, John P.; Crispin, MaxCell Reports (2018), 24 (8), 1958-1966.e5CODEN: CREED8; ISSN:2211-1247. (Cell Press)Many broadly neutralizing antibodies (bnAbs) against HIV-1 recognize and/or penetrate the glycan shield on native, virion-assocd. envelope glycoprotein (Env) spikes. The same bnAbs also bind to recombinant, sol. trimeric immunogens based on the SOSIP design. While SOSIP trimers are close structural and antigenic mimics of virion Env, the extent to which their glycan structures resemble ones on infectious viruses is undefined. Here, we compare the overall glycosylation of gp120 and gp41 subunits from BG505 (clade A) virions produced in a lymphoid cell line with those from recombinant BG505 SOSIP trimers, including CHO-derived clin. grade material. We also performed detailed site-specific analyses of gp120. Glycans relevant to key bnAb epitopes are generally similar on the recombinant SOSIP and virion-derived Env proteins, although the latter do contain hotspots of elevated glycan processing. Knowledge of native vs. recombinant Env glycosylation will guide vaccine design and manufg. programs.
- 34Watanabe, Y.; Berndsen, Z. T.; Raghwani, J.; Seabright, G. E.; Allen, J. D.; Pybus, O. G.; McLellan, J. S.; Wilson, I. A.; Bowden, T. A.; Ward, A. B. Vulnerabilities in Coronavirus Glycan Shields despite Extensive Glycosylation. Nat. Commun. 2020, 11 (1), 2688, DOI: 10.1038/s41467-020-16567-0[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVCmu77L&md5=2c0ce9b10f2a8d10d6889e975e9e09b3Vulnerabilities in coronavirus glycan shields despite extensive glycosylationWatanabe, Yasunori; Berndsen, Zachary T.; Raghwani, Jayna; Seabright, Gemma E.; Allen, Joel D.; Pybus, Oliver G.; McLellan, Jason S.; Wilson, Ian A.; Bowden, Thomas A.; Ward, Andrew B.; Crispin, MaxNature Communications (2020), 11 (1), 2688CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: Severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) coronaviruses (CoVs) are zoonotic pathogens with high fatality rates and pandemic potential. Vaccine development focuses on the principal target of the neutralizing humoral immune response, the spike (S) glycoprotein. Coronavirus S proteins are extensively glycosylated, encoding around 66-87 N-linked glycosylation sites per trimeric spike. Here, we reveal a specific area of high glycan d. on MERS S that results in the formation of oligomannose-type glycan clusters, which were absent on SARS and HKU1 CoVs. We provide a comparison of the global glycan d. of coronavirus spikes with other viral proteins including HIV-1 envelope, Lassa virus glycoprotein complex, and influenza hemagglutinin, where glycosylation plays a known role in shielding immunogenic epitopes. Overall, our data reveal how organization of glycosylation across class I viral fusion proteins influence not only individual glycan compns. but also the immunol. pressure across the protein surface.
- 35Zhao, P.; Praissman, J. L.; Grant, O. C.; Chen, B.; Brief, I.; Cai, Y.; Xiao, T.; Rosenbalm, K. E.; Aoki, K.; Kellman, B. P. Virus-Receptor Interactions of Glycosylated SARS-CoV-2 Spike and Human ACE2 Receptor. Cell Host Microbe 2020, 28, 1– 16, DOI: 10.1016/j.chom.2020.08.004
- 36Brun, J.; Vasiljevic, S.; Gangadharan, B.; Hensen, M.; Chandran, A. V.; Hill, M. L.; Kiappes, J. L.; Dwek, R. A.; Alonzi, D. S.; Struwe, W. B.; Analysis of SARS-CoV-2 Spike Glycosylation Reveals Shedding of a Vaccine Candidate. bioRxiv . bioRxiv November 16, 2020; DOI: 10.1101/2020.11.16.384594 .
- 37Morris, S. J.; Sebastian, S.; Spencer, A. J.; Gilbert, S. C. Simian Adenoviruses as Vaccine Vectors. Future Virology; Future Medicine Ltd: September 1, 2016; pp 649– 659. DOI: 10.2217/fvl-2016-0070 .
- 38Dicks, M. D. J.; Spencer, A. J.; Coughlan, L.; Bauza, K.; Gilbert, S. C.; Hill, A. V. S.; Cottingham, M. G. Differential Immunogenicity between HAdV-5 and Chimpanzee Adenovirus Vector ChAdOx1 Is Independent of Fiber and Penton RGD Loop Sequences in Mice. Sci. Rep. 2015, 5 (1), 1– 15, DOI: 10.1038/srep16756
- 39Essalmani, R.; Jain, J.; Susan-Resiga, D.; Andréo, U.; Evagelidis, A.; Derbali, R. M.; Huynh, D. N.; Dallaire, F.; Laporte, M.; Delpal, A.; Furin Cleaves SARS-CoV-2 Spike-Glycoprotein at S1/S2 and S2’ for Viral Fusion/Entry: Indirect Role of TMPRSS2. bioRxiv 2020, DOI: 10.1101/2020.12.18.423106 .
- 40Uhlen, M.; Fagerberg, L.; Hallstrom, B. M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; Asplund, A. Tissue-Based Map of the Human Proteome. Science 2015, 347 (6220), 1260419– 1260419, DOI: 10.1126/science.1260419[Crossref], [PubMed], [CAS], Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MvptFakug%253D%253D&md5=95510888806e5feb6b8024ee2a417d1cProteomics. Tissue-based map of the human proteomeUhlen Mathias; Fagerberg Linn; Oksvold Per; Sivertsson ¡ÑÜAsa; Lundberg Emma; Odeberg Jacob; Alm Tove; Nilsson Peter; Schwenk Jochen M; von Feilitzen Kalle; Forsberg Mattias; Persson Lukas; Johansson Fredric; Zwahlen Martin; Hallstrom Bjorn M; Lindskog Cecilia; Kampf Caroline; Asplund Anna; Olsson IngMarie; Djureinovic Dijana; Edqvist Per-Henrik; Ponten Fredrik; Mardinoglu Adil; Sjostedt Evelina; Edlund Karolina; Navani Sanjay; Szigyarto Cristina Al-Khalili; Takanen Jenny Ottosson; Hober Sophia; Berling Holger; Tegel Hanna; Rockberg Johan; Hamsten Marica; Mulder Jan; von Heijne Gunnar; Nielsen JensScience (New York, N.Y.) (2015), 347 (6220), 1260419 ISSN:.Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body.
- 41Hamming, I.; Timens, W.; Bulthuis, M. L. C.; Lely, A. T.; Navis, G. J.; van Goor, H. Tissue Distribution of ACE2 Protein, the Functional Receptor for SARS Coronavirus. A First Step in Understanding SARS Pathogenesis. J. Pathol. 2004, 203 (2), 631– 637, DOI: 10.1002/path.1570[Crossref], [PubMed], [CAS], Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlsVWktbY%253D&md5=0e7e65a40dd1d6cc50c0d1d28acf41cfTissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesisHamming, I.; Timens, W.; Bulthuis, M. L. C.; Lely, A. T.; Navis, G. J.; van Goor, H.Journal of Pathology (2004), 203 (2), 631-637CODEN: JPTLAS; ISSN:0022-3417. (John Wiley & Sons Ltd.)Severe acute respiratory syndrome (SARS) is an acute infectious disease that spreads mainly via the respiratory route. A distinct coronavirus (SARS-CoV) has been identified as the etiol. agent of SARS. Recently, a metallopeptidase named angiotensin-converting enzyme 2 (ACE2) has been identified as the functional receptor for SARS-CoV. Although ACE2 mRNA is known to be present in virtually all organs, its protein expression is largely unknown. Since identifying the possible route of infection has major implications for understanding the pathogenesis and future treatment strategies for SARS, the present study investigated the localization of ACE2 protein in various human organs (oral and nasal mucosa, nasopharynx, lung, stomach, small intestine, colon, skin, lymph nodes, thymus, bone marrow, spleen, liver, kidney, and brain). The most remarkable finding was the surface expression of ACE2 protein on lung alveolar epithelial cells and enterocytes of the small intestine. Furthermore, ACE2 was present in arterial and venous endothelial cells and arterial smooth muscle cells in all organs studied. In conclusion, ACE2 is abundantly present in humans in the epithelia of the lung and small intestine, which might provide possible routes of entry for the SARS-CoV. This epithelial expression, together with the presence of ACE2 in vascular endothelium, also provides a first step in understanding the pathogenesis of the main SARS disease manifestations.
- 42Sungnak, W.; Huang, N.; Bécavin, C.; Berg, M.; Queen, R.; Litvinukova, M.; Talavera-López, C.; Maatz, H.; Reichart, D.; Sampaziotis, F. SARS-CoV-2 Entry Factors Are Highly Expressed in Nasal Epithelial Cells Together with Innate Immune Genes. Nat. Med. 2020, 26 (5), 681– 687, DOI: 10.1038/s41591-020-0868-6[Crossref], [PubMed], [CAS], Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXotVCjurY%253D&md5=cc313df9c190333b98af43951a2146faSARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genesSungnak, Waradon; Huang, Ni; Becavin, Christophe; Berg, Marijn; Queen, Rachel; Litvinukova, Monika; Talavera-Lopez, Carlos; Maatz, Henrike; Reichart, Daniel; Sampaziotis, Fotios; Worlock, Kaylee B.; Yoshida, Masahiro; Barnes, Josephine L.; HCA Lung Biological NetworkNature Medicine (New York, NY, United States) (2020), 26 (5), 681-687CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)We investigated SARS-CoV-2 potential tropism by surveying expression of viral entry-assocd. genes in single-cell RNA-sequencing data from multiple tissues from healthy human donors. We co-detected these transcripts in specific respiratory, corneal and intestinal epithelial cells, potentially explaining the high efficiency of SARS-CoV-2 transmission. These genes are co-expressed in nasal epithelial cells with genes involved in innate immunity, highlighting the cells' potential role in initial viral infection, spread and clearance. The study offers a useful resource for further lines of inquiry with valuable clin. samples from COVID-19 patients and we provide our data in a comprehensive, open and user-friendly fashion at www.covid19cellatlas.org.
- 43Kornfeld, R.; Kornfeld, S. Assembly of Asparagine-Linked Oligosaccharides. Annu. Rev. Biochem. 1985, 54 (1), 631– 664, DOI: 10.1146/annurev.bi.54.070185.003215[Crossref], [PubMed], [CAS], Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL2M3nvVSjsA%253D%253D&md5=e4e0de7e37f61bbe1c7bb100927460ceAssembly of asparagine-linked oligosaccharidesKornfeld R; Kornfeld SAnnual review of biochemistry (1985), 54 (), 631-64 ISSN:0066-4154.There is no expanded citation for this reference.
- 44Gagneux, P.; Aebi, M.; Varki, A. Evolution of Glycan Diversity; Cold Spring Harbor Laboratory Press: 2015. DOI: 10.1101/GLYCOBIOLOGY.3E.020 .
- 45Mastronarde, D. N. Automated Electron Microscope Tomography Using Robust Prediction of Specimen Movements. J. Struct. Biol. 2005, 152 (1), 36– 51, DOI: 10.1016/j.jsb.2005.07.007[Crossref], [PubMed], [CAS], Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2MrjtVSrtw%253D%253D&md5=16b6502753c0f1f63ff559a784bce8f0Automated electron microscope tomography using robust prediction of specimen movementsMastronarde David NJournal of structural biology (2005), 152 (1), 36-51 ISSN:1047-8477.A new method was developed to acquire images automatically at a series of specimen tilts, as required for tomographic reconstruction. The method uses changes in specimen position at previous tilt angles to predict the position at the current tilt angle. Actual measurement of the position or focus is skipped if the statistical error of the prediction is low enough. This method allows a tilt series to be acquired rapidly when conditions are good but falls back toward the traditional approach of taking focusing and tracking images when necessary. The method has been implemented in a program, SerialEM, that provides an efficient environment for data acquisition. This program includes control of an energy filter as well as a low-dose imaging mode, in which tracking and focusing occur away from the area of interest. The program can automatically acquire a montage of overlapping frames, allowing tomography of areas larger than the field of the CCD camera. It also includes tools for navigating between specimen positions and finding regions of interest.
- 46Hagen, W. J. H.; Wan, W.; Briggs, J. A. G. Implementation of a Cryo-Electron Tomography Tilt-Scheme Optimized for High Resolution Subtomogram Averaging. J. Struct. Biol. 2017, 197 (2), 191– 198, DOI: 10.1016/j.jsb.2016.06.007[Crossref], [PubMed], [CAS], Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2s7gvFGksw%253D%253D&md5=e729954381b30719b7f07eaff9eaa496Implementation of a cryo-electron tomography tilt-scheme optimized for high resolution subtomogram averagingHagen Wim J H; Wan William; Briggs John A GJournal of structural biology (2017), 197 (2), 191-198 ISSN:.Cryo-electron tomography (cryoET) allows 3D structural information to be obtained from cells and other biological samples in their close-to-native state. In combination with subtomogram averaging, detailed structures of repeating features can be resolved. CryoET data is collected as a series of images of the sample from different tilt angles; this is performed by physically rotating the sample in the microscope between each image. The angles at which the images are collected, and the order in which they are collected, together are called the tilt-scheme. Here we describe a "dose-symmetric tilt-scheme" that begins at low tilt and then alternates between increasingly positive and negative tilts. This tilt-scheme maximizes the amount of high-resolution information maintained in the tomogram for subsequent subtomogram averaging, and may also be advantageous for other applications. We describe implementation of the tilt-scheme in combination with further data-collection refinements including setting thresholds on acceptable drift and improving focus accuracy. Requirements for microscope set-up are introduced, and a macro is provided which automates the application of the tilt-scheme within SerialEM.
- 47Zheng, S. Q.; Palovcak, E.; Armache, J. P.; Verba, K. A.; Cheng, Y.; Agard, D. A. MotionCor2: Anisotropic Correction of Beam-Induced Motion for Improved Cryo-Electron Microscopy. Nat. Methods 2017, 14 (4), 331– 332, DOI: 10.1038/nmeth.4193[Crossref], [PubMed], [CAS], Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjt1ags7g%253D&md5=5f4e225ef8123dacd8475d526175e1d2MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopyZheng, Shawn Q.; Palovcak, Eugene; Armache, Jean-Paul; Verba, Kliment A.; Cheng, Yifan; Agard, David A.Nature Methods (2017), 14 (4), 331-332CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)A review on anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Here we describe MotionCor2, a software tool for anisotropic correction of beam-induced motion. Overall, MotionCor2 is extremely robust and sufficiently accurate at correcting local motions so that the very time-consuming and computationally intensive particle polishing in RELION can be skipped, importantly, it also works on a wide range of data sets, including cryo tomog. tilt series.
- 48Kremer, J. R.; Mastronarde, D. N.; McIntosh, J. R. Computer Visualization of Three-Dimensional Image Data Using IMOD. J. Struct. Biol. 1996, 116 (1), 71– 76, DOI: 10.1006/jsbi.1996.0013[Crossref], [PubMed], [CAS], Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK28zmtFyqtg%253D%253D&md5=b6ab779bdec60f8644e254124c4dbbe7Computer visualization of three-dimensional image data using IMODKremer J R; Mastronarde D N; McIntosh J RJournal of structural biology (1996), 116 (1), 71-6 ISSN:1047-8477.We have developed a computer software package, IMOD, as a tool for analyzing and viewing three-dimensional biological image data. IMOD is useful for studying and modeling data from tomographic, serial section, and optical section reconstructions. The software allows image data to be visualized by several different methods. Models of the image data can be visualized by volume or contour surface rendering and can yield quantitative information.
- 49Pettersen, E. F.; Goddard, T. D.; Huang, C. C.; Couch, G. S.; Greenblatt, D. M.; Meng, E. C.; Ferrin, T. E. UCSF Chimera - A Visualization System for Exploratory Research and Analysis. J. Comput. Chem. 2004, 25 (13), 1605– 1612, DOI: 10.1002/jcc.20084[Crossref], [PubMed], [CAS], Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmvVOhsbs%253D&md5=944b175f440c1ff323705987cf937ee7UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 50Shevchenko, A.; Tomas, H.; Havli, J.; Olsen, J. V.; Mann, M. In-Gel Digestion for Mass Spectrometric Characterization of Proteins and Proteomes. Nat. Protoc. 2006, 1 (6), 2856– 2860, DOI: 10.1038/nprot.2006.468[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtFGjt7nM&md5=7c82516bf8197dff95fe73a371af2f0cIn-gel digestion for mass spectrometric characterization of proteins and proteomesShevchenko, Andrej; Tomas, Henrik; Havlis, Jan; Olsen, Jesper V.; Mann, MatthiasNature Protocols (2006), 1 (6), 2856-2860CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)In-gel digestion of proteins isolated by gel electrophoresis is a cornerstone of mass spectrometry (MS)-driven proteomics. The 10-yr-old recipe by Shevchenko et al. has been optimized to increase the speed and sensitivity of anal. The protocol is for the in-gel digestion of both silver and Coomassie-stained protein spots or bands and can be followed by MALDI-MS or LC-MS/MS anal. to identify proteins at sensitivities better than a few femtomoles of protein starting material.
- 51Emsley, P.; Crispin, M. Structural Analysis of Glycoproteins: Building N-Linked Glycans with Coot. Acta Crystallogr. Sect. D Struct. Biol. 2018, 74 (4), 256– 263, DOI: 10.1107/S2059798318005119[Crossref], [PubMed], [CAS], Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1KhsL8%253D&md5=b6324f66f05fbd26eed4d6b1cc6eb89eStructural analysis of glycoproteins: building N-linked glycans with CootEmsley, Paul; Crispin, MaxActa Crystallographica, Section D: Structural Biology (2018), 74 (4), 256-263CODEN: ACSDAD; ISSN:2059-7983. (International Union of Crystallography)Coot is a graphics application that is used to build or manipulate macromol. models; its particular forte is manipulation of the model at the residue level. The model-building tools of Coot have been combined and extended to assist or automate the building of N-linked glycans. The model is built by the addn. of monosaccharides, placed by variation of internal coordinates. The subsequent model is refined by real-space refinement, which is stabilized with modified and addnl. restraints. It is hoped that these enhanced building tools will help to reduce building errors of N-linked glycans and improve our knowledge of the structures of glycoproteins.
Cited By
This article is cited by 4 publications.
- Suman Maity, Atanu Acharya. Many Roles of Carbohydrates: A Computational Spotlight on the Coronavirus S Protein Binding. ACS Applied Bio Materials 2023, Article ASAP.
- Serge Perez, Olga Makshakova. Multifaceted Computational Modeling in Glycoscience. Chemical Reviews 2022, 122 (20) , 15914-15970. https://doi.org/10.1021/acs.chemrev.2c00060
- Victor Yin, Szu-Hsueh Lai, Tom G. Caniels, Philip J. M. Brouwer, Mitch Brinkkemper, Yoann Aldon, Hejun Liu, Meng Yuan, Ian A. Wilson, Rogier W. Sanders, Marit J. van Gils, Albert J. R. Heck. Probing Affinity, Avidity, Anticooperativity, and Competition in Antibody and Receptor Binding to the SARS-CoV-2 Spike by Single Particle Mass Analyses. ACS Central Science 2021, 7 (11) , 1863-1873. https://doi.org/10.1021/acscentsci.1c00804
- Joel D. Allen, Himanshi Chawla, Firdaus Samsudin, Lorena Zuzic, Aishwary Tukaram Shivgan, Yasunori Watanabe, Wan-ting He, Sean Callaghan, Ge Song, Peter Yong, Philip J. M. Brouwer, Yutong Song, Yongfei Cai, Helen M. E. Duyvesteyn, Tomas Malinauskas, Joeri Kint, Paco Pino, Maria J. Wurm, Martin Frank, Bing Chen, David I. Stuart, Rogier W. Sanders, Raiees Andrabi, Dennis R. Burton, Sai Li, Peter J. Bond, Max Crispin. Site-Specific Steric Control of SARS-CoV-2 Spike Glycosylation. Biochemistry 2021, 60 (27) , 2153-2169. https://doi.org/10.1021/acs.biochem.1c00279
Abstract
Figure 1
Figure 1. ChAdOx1 nCoV-19 produces membrane associated SARS-CoV-2 S glycoprotein in native conformations able to bind its host receptor, ACE2. (A) Schematic representation of the vaccine encoded SARS-CoV-2 S protein, showing the position of N-linked glycosylation amino acid sequons (NXS/T, where X ≠ P) as branches. Protein domains are illustrated: N-terminal domain (NTD), receptor-binding domain (RBD), fusion peptide (FP), heptad repeat 1 (HR1), central helix (CH), connector domain (CD), and transmembrane domain (TM), with the additional tPA secretion signal at the N-terminus. (B) HeLa S3 cells were infected with ChAdOx1 nCoV-19 and incubated with recombinant ACE2, anti-ChAdOx1 nCoV-19 (derived from vaccinated mice), or a panel of human mAbs (Ab44, Ab45, Ab71, and Ab111, which recognize S2, RBD, trimeric S, and NTD, respectively) and compared to noninfected controls, analyzed by flow cytometry. (Left). Relative frequency of cells and AlexaFluor 488 fluorescence associated with antispike detection is plotted. Left, (blue) anti-ChAdOx1 nCoV-19; middle (red), ACE2; and right (shades of green) human mAbs. In dark gray cells infected with an irrelevant ChAdOx1 vaccine and in light gray noninfected cells are shown as a control. Experimental replicates were performed two times, and representative data are shown.
Figure 2
Figure 2. CryoET and subtomogram average of ChAdOx1 nCoV-19 derived spike. (A) Tomographic slice of U2OS cell transduced with ChAdOx1 nCoV-19. The slice is 6.4 Å thick; PM = plasma membrane, scale bar = 100 nm. (B) Detailed view of the boxed area marked in (A). White arrowheads indicate spike proteins on the cell surface; scale bar = 50 nm. (C–E) Subtomogram average of ChAdOx1 nCoV-19 spikes at 11.6 Å resolution as indicated by Fourier-Shell correlation at 0.5 cutoff (C), shown from side view (D) and top view (E). SARS-CoV-2 atomic model (PDB 6ZB5) (29) is fitted for reference.
Figure 3
Figure 3. Site-specific glycan processing of SARS-CoV-2 S upon infection with ChAdOx1 nCoV-19. (A) Western blot analysis of SARS-CoV-2 spike proteins, using anti-S1 and anti-S1+S2 antibodies. Lane 1 = Protein pellet from 293F cell lysates infected with ChAdOx1 nCoV-19. Lane 2 = Reduced protein pellet from 293F infected with ChAdOx1 nCoV-19. Lane 3 = 2P-stabilized SARS-CoV-2 S protein. The white boxes correspond to gel bands that were excised for mass spectrometric analysis. (B) Site-specific N-linked glycosylation of SARS-CoV-2 S0 and S1/S2 glycoproteins. The bar graphs represent the relative quantities of digested glycopeptides possessing the identifiers of oligomannose/hybrid-type glycans (green), complex-type glycans (pink), unoccupied PNGs (gray), or not determined (N.D.) at each N-linked glycan sequon on the S protein, listed from N to C terminus. (C) Glycosylated model of the cleaved (S1/S2) SARS-CoV-2 spike. The pie charts summarize the mass spectrometric analysis of the oligomannose/hybrid (green), complex (pink), or unoccupied (gray) N-linked glycan populations. Representative glycans are modeled onto the prefusion structure of trimeric SARS-CoV-2 S glycoprotein (PDB ID: 6VSB), (3) with one RBD in the “up” conformation. The modeled glycans are colored according to oligomannose/hybrid-type glycan content with glycan sites labeled in green (80–100%), orange (30–79%), pink (0–29%), or gray (not detected).
References
ARTICLE SECTIONSThis article references 51 other publications.
- 1Amanat, F.; Krammer, F. SARS-CoV-2 Vaccines: Status Report. Immunity. Cell Press: April 14, 2020; pp 583– 589. DOI: 10.1016/j.immuni.2020.03.007 .
- 2Letko, M.; Marzi, A.; Munster, V. Functional Assessment of Cell Entry and Receptor Usage for SARS-CoV-2 and Other Lineage B Betacoronaviruses. Nat. Microbiol. 2020, 1– 8, DOI: 10.1038/s41564-020-0688-y
- 3Wrapp, D.; Wang, N.; Corbett, K. S.; Goldsmith, J. A.; Hsieh, C.-L.; Abiona, O.; Graham, B. S.; McLellan, J. S. Cryo-EM Structure of the 2019-NCoV Spike in the Prefusion Conformation. Science 2020, 367, 1260, DOI: 10.1126/science.abb2507[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFemt70%253D&md5=27d08cbb9a43d1da051a8a92a9f68aa5Cryo-EM structure of the 2019-nCoV spike in the prefusion conformationWrapp, Daniel; Wang, Nianshuang; Corbett, Kizzmekia S.; Goldsmith, Jory A.; Hsieh, Ching-Lin; Abiona, Olubukola; Graham, Barney S.; McLellan, Jason S.Science (Washington, DC, United States) (2020), 367 (6483), 1260-1263CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The outbreak of a novel coronavirus (2019-nCoV) represents a pandemic threat that has been declared a public health emergency of international concern. The CoV spike (S) glycoprotein is a key target for vaccines, therapeutic antibodies, and diagnostics. To facilitate medical countermeasure development, we detd. a 3.5-angstrom-resoln. cryo-electron microscopy structure of the 2019-nCoV S trimer in the prefusion conformation. The predominant state of the trimer has one of the three receptor-binding domains (RBDs) rotated up in a receptor-accessible conformation. We also provide biophys. and structural evidence that the 2019-nCoV S protein binds angiotensin-converting enzyme 2 (ACE2) with higher affinity than does severe acute respiratory syndrome (SARS)-CoV S. Addnl., we tested several published SARS-CoV RBD-specific monoclonal antibodies and found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs. The structure of 2019-nCoV S should enable the rapid development and evaluation of medical countermeasures to address the ongoing public health crisis.
- 4Walls, A. C.; Park, Y. J.; Tortorici, M. A.; Wall, A.; McGuire, A. T.; Veesler, D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 2020, 181 (2), 281– 292, e6 DOI: 10.1016/j.cell.2020.02.058[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvVejsLk%253D&md5=ac8a8a208d9c26f88f702fb7634ab1abStructure, Function, and Antigenicity of the SARS-CoV-2 Spike GlycoproteinWalls, Alexandra C.; Park, Young-Jun; Tortorici, M. Alejandra; Wall, Abigail; McGuire, Andrew T.; Veesler, DavidCell (Cambridge, MA, United States) (2020), 181 (2), 281-292.e6CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The emergence of SARS-CoV-2 has resulted in >90,000 infections and >3000 deaths. Coronavirus spike (S) glycoproteins promote entry into cells and are the main target of antibodies. We show that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of SARS-CoV-2 S and SARS-CoV S bind with similar affinities to human ACE2, correlating with the efficient spread of SARS-CoV-2 among humans. We found that the SARS-CoV-2 S glycoprotein harbors a furin cleavage site at the boundary between the S1/S2 subunits, which is processed during biogenesis and sets this virus apart from SARS-CoV and SARS-related CoVs. We detd. cryo-EM structures of the SARS-CoV-2 S ectodomain trimer, providing a blueprint for the design of vaccines and inhibitors of viral entry. Finally, we demonstrate that SARS-CoV S murine polyclonal antibodies potently inhibited SARS-CoV-2 S mediated entry into cells, indicating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination.
- 5Watanabe, Y.; Allen, J. D.; Wrapp, D.; McLellan, J. S.; Crispin, M. Site-Specific Glycan Analysis of the SARS-CoV-2 Spike. Science 2020, eabb9983 DOI: 10.1126/science.abb9983 .
- 6Krammer, F. SARS-CoV-2 Vaccines in Development. Nature 2020, 1– 16, DOI: 10.1038/s41586-020-2798-3
- 7Polack, F. P.; Thomas, S. J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J. L.; Pérez Marc, G.; Moreira, E. D.; Zerbini, C. Safety and Efficacy of the BNT162b2MRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383 (27), 2603– 2615, DOI: 10.1056/NEJMoa2034577[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFSjuw%253D%253D&md5=08cb7204334ed37c7949dfcdfe5af75cSafety and efficacy of the BNT162b2 mRNA Covid-19 vaccinePolack, Fernando P.; Thomas, Stephen J.; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Perez, John L.; Marc, Gonzalo Perez; Moreira, Edson D.; Zerbini, Cristiano; Bailey, Ruth; Swanson, Kena A.; Roychoudhury, Satrajit; Koury, Kenneth; Li, Ping; Kalina, Warren V.; Cooper, David; Frenck, Robert W., Jr.; Hammitt, Laura L.; Tureci, Ozlem; Nell, Haylene; Schaefer, Axel; Unal, Serhat; Tresnan, Dina B.; Mather, Susan; Dormitzer, Philip R.; Sahin, Ugur; Jansen, Kathrin U.; Gruber, William C.New England Journal of Medicine (2020), 383 (27), 2603-2615CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)A review. Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a worldwide pandemic. Safe and effective vaccines are needed urgently. methods In an ongoing multinational, placebo-controlled, observer-blinded, pivotal efficacy trial, we randomly assigned persons 16 years of age or older in a 1:1 ratio to receive two doses, 21 days apart, of either placebo or the BNT162b2 vaccine candidate (30μg per dose). BNT162b2 is a lipid nanoparticle-formulated, nucleoside-modified RNA vaccine that encodes a prefusion stabilized, membrane-anchored SARS-CoV-2 full-length spike protein. The primary end points were efficacy of the vaccine against lab.-confirmed Covid-19 and safety. results A total of 43,548 participants underwent randomization, of whom 43,448 received injections: 21,720 with BNT162b2 and 21,728 with placebo. There were 8 cases of Covid-19 with onset at least 7 days after the second dose among participants assigned to receive BNT162b2 and 162 cases among those assigned to placebo; BNT162b2 was 95% effective in preventing Covid-19 (95% credible interval, 90.3 to 97.6). Similar vaccine efficacy (generally 90 to 100%) was obsd. across subgroups defined by age, sex, race, ethnicity, baseline body-mass index, and the presence of coexisting conditions. Among 10 cases of severe Covid-19 with onset after the first dose, 9 occurred in placebo recipients and 1 in a BNT162b2 recipient. The safety profile of BNT162b2 was characterized by short-term, mild-to-moderate pain at the injection site, fatigue, and headache. The incidence of serious adverse events was low and was similar in the vaccine and placebo groups. conclusions A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older. Safety over a median of 2 mo was similar to that of other viral vaccines.
- 8Corbett, K. S.; Flynn, B.; Foulds, K. E.; Francica, J. R.; Boyoglu-Barnum, S.; Werner, A. P.; Flach, B.; O’Connell, S.; Bock, K. W.; Minai, M. Evaluation of the MRNA-1273 Vaccine against SARS-CoV-2 in Nonhuman Primates. N. Engl. J. Med. 2020, 383, 1544, DOI: 10.1056/NEJMoa2024671[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFWmsr7I&md5=8ecba6b18bc615d886ca5016b7c85496Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primatesCorbett, K. S.; Flynn, B.; Foulds, K. E.; Francica, J. R.; Boyoglu-Barnum, S.; Werner, A. P.; Flach, B.; O'Connell, S.; Bock, K. W.; Minai, M.; Nagata, B. M.; Andersen, H.; Martinez, D. R.; Noe, A. T.; Douek, N.; Donaldson, M. M.; Nji, N. N.; Alvarado, G. S.; Edwards, D. K.; Flebbe, D. R.; Lamb, E.; Doria-Rose, N. A.; Lin, B. C.; Louder, M. K.; O'Dell, S.; Schmidt, S. D.; Phung, E.; Chang, L. A.; Yap, C.; Todd, J.-P. M.; Pessaint, L.; Van Ry, A.; Browne, S.; Greenhouse, J.; Putman-Taylor, T.; Strasbaugh, A.; Campbell, T.-A.; Cook, A.; Dodson, A.; Steingrebe, K.; Shi, W.; Zhang, Y.; Abiona, O. M.; Wang, L.; Pegu, A.; Yang, E. S.; Leung, K.; Zhou, T.; Teng, I-T.; Widge, A.; Gordon, I.; Novik, L.; Gillespie, R. A.; Loomis, R. J.; Moliva, J. I.; Stewart-Jones, G.; Himansu, S.; Kong, W.-P.; Nason, M. C.; Morabito, K. M.; Ruckwardt, T. J.; Ledgerwood, J. E.; Gaudinski, M. R.; Kwong, P. D.; Mascola, J. R.; Carfi, A.; Lewis, M. G.; Baric, R. S.; McDermott, A.; Moore, I. N.; Sullivan, N. J.; Roederer, M.; Seder, R. A.; Graham, B. S.New England Journal of Medicine (2020), 383 (16), 1544-1555CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Background: Vaccines to prevent coronavirus disease 2019 (Covid-19) are urgently needed. The effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines on viral replication in both upper and lower airways is important to evaluate in nonhuman primates. Methods: Nonhuman primates received 10 or 100μg of mRNA-1273, a vaccine encoding the prefusion-stabilized spike protein of SARS-CoV-2, or no vaccine. Antibody and T-cell responses were assessed before upper- and lower-airway challenge with SARS-CoV-2. Active viral replication and viral genomes in bronchoalveolar-lavage (BAL) fluid and nasal swab specimens were assessed by polymerase chain reaction, and histopathol. anal. and viral quantification were performed on lung-tissue specimens. Results The mRNA-1273 vaccine candidate induced antibody levels exceeding those in human convalescent-phase serum, with live-virus reciprocal 50% inhibitory diln. (ID50) geometric mean titers of 501 in the 10-μg dose group and 3481 in the 100-μg dose group. Vaccination induced type 1 helper T-cell (Th1)-biased CD4 T-cell responses and low or undetectable Th2 or CD8 T-cell responses. Viral replication was not detectable in BAL fluid by day 2 after challenge in seven of eight animals in both vaccinated groups. No viral replication was detectable in the nose of any of the eight animals in the 100-μg dose group by day 2 after challenge, and limited inflammation or detectable viral genome or antigen was noted in lungs of animals in either vaccine group. conclusions Vaccination of nonhuman primates with mRNA-1273 induced robust SARS-CoV-2 neutralizing activity, rapid protection in the upper and lower airways, and no pathol. changes in the lung.
- 9Gao, Q.; Bao, L.; Mao, H.; Wang, L.; Xu, K.; Yang, M.; Li, Y.; Zhu, L.; Wang, N.; Lv, Z. Development of an Inactivated Vaccine Candidate for SARS-CoV-2. Science 2020, 369 (6499), 77– 81, DOI: 10.1126/science.abc1932[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlCmtL3P&md5=674788246758fd31fa8bb54c936be83aDevelopment of an inactivated vaccine candidate for SARS-CoV-2Gao, Qiang; Bao, Linlin; Mao, Haiyan; Wang, Lin; Xu, Kangwei; Yang, Minnan; Li, Yajing; Zhu, Ling; Wang, Nan; Lv, Zhe; Gao, Hong; Ge, Xiaoqin; Kan, Biao; Hu, Yaling; Liu, Jiangning; Cai, Fang; Jiang, Deyu; Yin, Yanhui; Qin, Chengfeng; Li, Jing; Gong, Xuejie; Lou, Xiuyu; Shi, Wen; Wu, Dongdong; Zhang, Hengming; Zhu, Lang; Deng, Wei; Li, Yurong; Lu, Jinxing; Li, Changgui; Wang, Xiangxi; Yin, Weidong; Zhang, Yanjun; Qin, ChuanScience (Washington, DC, United States) (2020), 369 (6499), 77-81CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in an unprecedented public health crisis. Because of the novelty of the virus, there are currently no SARS-CoV-2-specific treatments or vaccines available. Therefore, rapid development of effective vaccines against SARS-CoV-2 are urgently needed. Here, we developed a pilot-scale prodn. of PiCoVacc, a purified inactivated SARS-CoV-2 virus vaccine candidate, which induced SARS-CoV-2-specific neutralizing antibodies in mice, rats, and nonhuman primates. These antibodies neutralized 10 representative SARS-CoV-2 strains, suggesting a possible broader neutralizing ability against other strains. Three immunizations using two different doses, 3 or 6μg per dose, provided partial or complete protection in macaques against SARS-CoV-2 challenge, resp., without observable antibody-dependent enhancement of infection. These data support the clin. development and testing of PiCoVacc for use in humans.
- 10Liu, C.; Mendonæa, L.; Yang, Y.; Gao, Y.; Shen, C.; Liu, J.; Ni, T.; Ju, B.; Liu, C.; Tang, X. The Architecture of Inactivated SARS-CoV-2 with Postfusion Spikes Revealed by Cryo-EM and Cryo-ET. Structure 2020, 28, 1218, DOI: 10.1016/j.str.2020.10.001[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVylsb3I&md5=79c3780b2fb235c85d39a7cc855ada50The Architecture of Inactivated SARS-CoV-2 with Postfusion Spikes Revealed by Cryo-EM and Cryo-ETLiu, Chuang; Mendonca, Luiza; Yang, Yang; Gao, Yuanzhu; Shen, Chenguang; Liu, Jiwei; Ni, Tao; Ju, Bin; Liu, Congcong; Tang, Xian; Wei, Jinli; Ma, Xiaomin; Zhu, Yanan; Liu, Weilong; Xu, Shuman; Liu, Yingxia; Yuan, Jing; Wu, Jing; Liu, Zheng; Zhang, Zheng; Liu, Lei; Wang, Peiyi; Zhang, PeijunStructure (Oxford, United Kingdom) (2020), 28 (11), 1218-1224.e4CODEN: STRUE6; ISSN:0969-2126. (Elsevier Ltd.)The ongoing global pandemic of coronavirus disease 2019 (COVID-19) resulted from the outbreak of SARS-CoV-2 in Dec. 2019. Currently, multiple efforts are being made to rapidly develop vaccines and treatments to fight COVID-19. Current vaccine candidates use inactivated SARS-CoV-2 viruses; therefore, it is important to understand the architecture of inactivated SARS-CoV-2. We have genetically and structurally characterized β-propiolactone-inactivated viruses from a propagated and purified clin. strain of SARS-CoV-2. We obsd. that the virus particles are roughly spherical or moderately pleiomorphic. Although a small fraction of prefusion spikes are found, most spikes appear nail shaped, thus resembling a postfusion state, where the S1 protein of the spike has disassocd. from S2. Cryoelectron tomog. and subtomogram averaging of these spikes yielded a d. map that closely matches the overall structure of the SARS-CoV postfusion spike and its corresponding glycosylation site. Our findings have major implications for SARS-CoV-2 vaccine design, esp. those using inactivated viruses.
- 11Zost, S. J.; Gilchuk, P.; Case, J. B.; Binshtein, E.; Chen, R. E.; Nkolola, J. P.; Schäfer, A.; Reidy, J. X.; Trivette, A.; Nargi, R. S. Potently Neutralizing and Protective Human Antibodies against SARS-CoV-2. Nature 2020, 584 (7821), 443– 449, DOI: 10.1038/s41586-020-2548-6[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsF2ntLrM&md5=d39be143814010bd4eacc7b69f2e85cbPotently neutralizing and protective human antibodies against SARS-CoV-2Zost, Seth J.; Gilchuk, Pavlo; Case, James Brett; Binshtein, Elad; Chen, Rita E.; Nkolola, Joseph P.; Schafer, Alexandra; Reidy, Joseph X.; Trivette, Andrew; Nargi, Rachel S.; Sutton, Rachel E.; Suryadevara, Naveenchandra; Martinez, David R.; Williamson, Lauren E.; Chen, Elaine C.; Jones, Taylor; Day, Samuel; Myers, Luke; Hassan, Ahmed O.; Kafai, Natasha M.; Winkler, Emma S.; Fox, Julie M.; Shrihari, Swathi; Mueller, Benjamin K.; Meiler, Jens; Chandrashekar, Abishek; Mercado, Noe B.; Steinhardt, James J.; Ren, Kuishu; Loo, Yueh-Ming; Kallewaard, Nicole L.; McCune, Broc T.; Keeler, Shamus P.; Holtzman, Michael J.; Barouch, Dan H.; Gralinski, Lisa E.; Baric, Ralph S.; Thackray, Larissa B.; Diamond, Michael S.; Carnahan, Robert H.; Crowe Jr, James E.Nature (London, United Kingdom) (2020), 584 (7821), 443-449CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The ongoing pandemic of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major threat to global health and the medical countermeasures available so far are limited. Moreover, we currently lack a thorough understanding of the mechanisms of humoral immunity to SARS-CoV-2. We analyze a large panel of human monoclonal antibodies that target the spike (S) glycoprotein, and identify several that exhibit potent neutralizing activity and fully block the receptor-binding domain of the S protein (SRBD) from interacting with human angiotensin-converting enzyme 2 (ACE2). Using competition-binding, structural, and functional studies, we show that the monoclonal antibodies can be clustered into classes that recognize distinct epitopes on the SRBD, as well as distinct conformational states of the S trimer. Two potently neutralizing monoclonal antibodies, COV2-2196 and COV2-2130, which recognize non-overlapping sites, bound simultaneously to the S protein and neutralized wild-type SARS-CoV-2 virus in a synergistic manner. In 2 mouse models of SARS-CoV-2 infection, passive transfer of COV2-2196, COV2-2130, or a combination of both of these antibodies protected mice from wt. loss and reduced the viral burden and levels of inflammation in the lungs. In addn., passive transfer of either of 2 of the most potent ACE2-blocking monoclonal antibodies (COV2-2196 or COV2-2381) as monotherapy protected rhesus macaques from SARS-CoV-2 infection. These results identify protective epitopes on the SRBD and provide a structure-based framework for rational vaccine design and the selection of robust immunotherapeutic agents.
- 12Robbiani, D. F.; Gaebler, C.; Muecksch, F.; Lorenzi, J. C. C.; Wang, Z.; Cho, A.; Agudelo, M.; Barnes, C. O.; Gazumyan, A.; Finkin, S. Convergent Antibody Responses to SARS-CoV-2 in Convalescent Individuals. Nature 2020, 584 (7821), 437– 442, DOI: 10.1038/s41586-020-2456-9[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFegtb3P&md5=952577f69ffe8282d0b2451b17a5ecadConvergent antibody responses to SARS-CoV-2 in convalescent individualsRobbiani, Davide F.; Gaebler, Christian; Muecksch, Frauke; Lorenzi, Julio C. C.; Wang, Zijun; Cho, Alice; Agudelo, Marianna; Barnes, Christopher O.; Gazumyan, Anna; Finkin, Shlomo; Hagglof, Thomas; Oliveira, Thiago Y.; Viant, Charlotte; Hurley, Arlene; Hoffmann, Hans-Heinrich; Millard, Katrina G.; Kost, Rhonda G.; Cipolla, Melissa; Gordon, Kristie; Bianchini, Filippo; Chen, Spencer T.; Ramos, Victor; Patel, Roshni; Dizon, Juan; Shimeliovich, Irina; Mendoza, Pilar; Hartweger, Harald; Nogueira, Lilian; Pack, Maggi; Horowitz, Jill; Schmidt, Fabian; Weisblum, Yiska; Michailidis, Eleftherios; Ashbrook, Alison W.; Waltari, Eric; Pak, John E.; Huey-Tubman, Kathryn E.; Koranda, Nicholas; Hoffman, Pauline R.; West Jr, Anthony P.; Rice, Charles M.; Hatziioannou, Theodora; Bjorkman, Pamela J.; Bieniasz, Paul D.; Caskey, Marina; Nussenzweig, Michel C.Nature (London, United Kingdom) (2020), 584 (7821), 437-442CODEN: NATUAS; ISSN:0028-0836. (Nature Research)During the coronavirus disease-2019 (COVID-19) pandemic, severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has led to the infection of millions of people and has claimed hundreds of thousands of lives. The entry of the virus into cells depends on the receptor-binding domain (RBD) of the spike (S) protein of SARS-CoV-2. Although there is currently no vaccine, it is likely that antibodies will be essential for protection. However, little is known about the human antibody response to SARS-CoV-2. We report on 149 COVID-19-convalescent individuals. Plasma samples collected an av. of 39 days after the onset of symptoms had variable half-maximal pseudovirus neutralizing titers; titers were <50 in 33% of samples, <1000 in 79% of samples, and only 1% of samples had titers >5000. Antibody sequencing revealed the expansion of clones of RBD-specific memory B cells that expressed closely related antibodies in different individuals. Despite low plasma titers, antibodies to 3 distinct epitopes on the RBD neutralized the virus with half-maximal inhibitory concns. (IC50 values) as low as 2 ng/mL. In conclusion, most convalescent plasma samples obtained from individuals who recover from COVID-19 do not contain high levels of neutralizing activity. Nevertheless, rare but recurring RBD-specific antibodies with potent antiviral activity were found in all individuals tested, suggesting that a vaccine designed to elicit such antibodies could be broadly effective.
- 13Du, S.; Cao, Y.; Zhu, Q.; Yu, P.; Qi, F.; Wang, G.; Du, X.; Bao, L.; Deng, W.; Zhu, H. Structurally Resolved SARS-CoV-2 Antibody Shows High Efficacy in Severely Infected Hamsters and Provides a Potent Cocktail Pairing Strategy. Cell 2020, 183, 1013, DOI: 10.1016/j.cell.2020.09.035[Crossref], [PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFGqsLvJ&md5=230461817cefd12ef9eccbe6d6aa6a73Structurally resolved SARS-CoV-2 antibody shows high efficacy in severely infected hamsters and provides a potent cocktail pairing strategyDu, Shuo; Cao, Yunlong; Zhu, Qinyu; Yu, Pin; Qi, Feifei; Wang, Guopeng; Du, Xiaoxia; Bao, Linlin; Deng, Wei; Zhu, Hua; Liu, Jiangning; Nie, Jianhui; Zheng, Yinghui; Liang, Haoyu; Liu, Ruixue; Gong, Shuran; Xu, Hua; Yisimayi, Ayijiang; Lv, Qi; Wang, Bo; He, Runsheng; Han, Yunlin; Zhao, Wenjie; Bai, Yali; Qu, Yajin; Gao, Xiang; Ji, Chenggong; Wang, Qisheng; Gao, Ning; Huang, Weijin; Wang, Youchun; Xie, X. Sunney; Su, Xiao-dong; Xiao, Junyu; Qin, ChuanCell (Cambridge, MA, United States) (2020), 183 (4), 1013-1023.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Understanding how potent neutralizing antibodies (NAbs) inhibit SARS-CoV-2 is crit. for effective therapeutic development. We previously described BD-368-2, a SARS-CoV-2 NAb with high potency; however, its neutralization mechanism is largely unknown. Here, we report the 3.5-Å cryo-EM structure of BD-368-2/trimeric-spike complex, revealing that BD-368-2 fully blocks ACE2 recognition by occupying all three receptor-binding domains (RBDs) simultaneously, regardless of their "up" or "down" conformations. Also, BD-368-2 treats infected adult hamsters at low dosages and at various administering windows, in contrast to placebo hamsters that manifested severe interstitial pneumonia. Moreover, BD-368-2's epitope completely avoids the common binding site of VH3-53/VH3-66 recurrent NAbs, evidenced by tripartite co-crystal structures with RBDs. Pairing BD-368-2 with a potent recurrent NAb neutralizes SARS-CoV-2 pseudovirus at pM level and rescues mutation-induced neutralization escapes. Together, our results rationalized a new RBD epitope that leads to high neutralization potency and demonstrated BD-368-2's therapeutic potential in treating COVID-19.
- 14Barnes, C. O.; Jette, C. A.; Abernathy, M. E.; Dam, K.-M. A.; Esswein, S. R.; Gristick, H. B.; Malyutin, A. G.; Sharaf, N. G.; Huey-Tubman, K. E.; Lee, Y. E. SARS-CoV-2 Neutralizing Antibody Structures Inform Therapeutic Strategies. Nature 2020, 1– 9, DOI: 10.1038/s41586-020-2852-1
- 15Brouwer, P. J. M.; Caniels, T. G.; van der Straten, K.; Snitselaar, J. L.; Aldon, Y.; Bangaru, S.; Torres, J. L.; Okba, N. M. A.; Claireaux, M.; Kerster, G. Potent Neutralizing Antibodies from COVID-19 Patients Define Multiple Targets of Vulnerability. Science 2020, eabc5902 DOI: 10.1126/science.abc5902
- 16Folegatti, P. M.; Ewer, K. J.; Aley, P. K.; Angus, B.; Becker, S.; Belij-Rammerstorfer, S.; Bellamy, D.; Bibi, S.; Bittaye, M.; Clutterbuck, E. A.; Safety and Immunogenicity of the ChAdOx1 NCoV-19 Vaccine against SARS-CoV-2: A Preliminary Report of a Phase 1/2, Single-Blind, Randomised Controlled Trial. Lancet 2020, 0 (0). DOI: 10.1016/S0140-6736(20)31604-4 .
- 17van Doremalen, N.; Lambe, T.; Spencer, A.; Belij-Rammerstorfer, S.; Purushotham, J. N.; Port, J. R.; Avanzato, V. A.; Bushmaker, T.; Flaxman, A.; Ulaszewska, M. ChAdOx1 NCoV-19 Vaccine Prevents SARS-CoV-2 Pneumonia in Rhesus Macaques. Nature 2020, 1– 5, DOI: 10.1038/s41586-020-2608-y
- 18Graham, S. P.; McLean, R. K.; Spencer, A. J.; Belij-Rammerstorfer, S.; Wright, D.; Ulaszewska, M.; Edwards, J. C.; Hayes, J. W. P.; Martini, V.; Thakur, N. Evaluation of the Immunogenicity of Prime-Boost Vaccination with the Replication-Deficient Viral Vectored COVID-19 Vaccine Candidate ChAdOx1 NCoV-19. npj Vaccines 2020, 5 (1), 1– 6, DOI: 10.1038/s41541-020-00221-3
- 19Ramasamy, M. N.; Minassian, A. M.; Ewer, K. J.; Flaxman, A. L.; Folegatti, P. M.; Owens, D. R.; Voysey, M.; Aley, P. K.; Angus, B.; Babbage, G.; Safety and Immunogenicity of ChAdOx1 NCoV-19 Vaccine Administered in a Prime-Boost Regimen in Young and Old Adults (COV002): A Single-Blind, Randomised, Controlled, Phase 2/3 Trial. Lancet 2020, 0 (0). DOI: 10.1016/s0140-6736(20)32466-1 .
- 20Sebastian, S.; Flaxman, A.; Cha, K. M.; Ulaszewska, M.; Gilbride, C.; Sharpe, H.; Wright, E.; Spencer, A. J.; Dowall, S.; Hewson, R.; A Multi-Filovirus Vaccine Candidate: Co-Expression of Ebola, Sudan, and Marburg Antigens in a Single Vector. Vaccines 2020, 8 (2). DOI: 10.3390/vaccines8020241 .
- 21Zhang, L.; Jackson, C. B.; Mou, H.; Ojha, A.; Peng, H.; Quinlan, B. D.; Rangarajan, E. S.; Pan, A.; Vanderheiden, A.; Suthar, M. S. SARS-CoV-2 Spike-Protein D614G Mutation Increases Virion Spike Density and Infectivity. Nat. Commun. 2020, 11 (1), 1– 9, DOI: 10.1038/s41467-020-19808-4
- 22Pallesen, J.; Wang, N.; Corbett, K. S.; Wrapp, D.; Kirchdoerfer, R. N.; Turner, H. L.; Cottrell, C. A.; Becker, M. M.; Wang, L.; Shi, W. Immunogenicity and Structures of a Rationally Designed Prefusion MERS-CoV Spike Antigen. Proc. Natl. Acad. Sci. U. S. A. 2017, 114 (35), E7348– E7357, DOI: 10.1073/pnas.1707304114[Crossref], [PubMed], [CAS], Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlWmsrfI&md5=3054f8054d852ed4192fd8f1fb6c870dImmunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigenPallesen, Jesper; Wang, Nianshuang; Corbett, Kizzmekia S.; Wrapp, Daniel; Kirchdoerfer, Robert N.; Turner, Hannah L.; Cottrell, Christopher A.; Becker, Michelle M.; Wang, Lingshu; Shi, Wei; Kong, Wing-Pui; Andres, Erica L.; Kettenbach, Arminja N.; Denison, Mark R.; Chappell, James D.; Graham, Barney S.; Ward, Andrew B.; McLellan, Jason S.Proceedings of the National Academy of Sciences of the United States of America (2017), 114 (35), E7348-E7357CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Middle East respiratory syndrome coronavirus (MERS-CoV) is a lineage C betacoronavirus that since its emergence in 2012 has caused outbreaks in human populations with case-fatality rates of ∼36%. As in other coronaviruses, the spike (S) glycoprotein of MERS-CoV mediates receptor recognition and membrane fusion and is the primary target of the humoral immune response during infection. Here we use structure-based design to develop a generalizable strategy for retaining coronavirus S proteins in the antigenically optimal prefusion conformation and demonstrate that our engineered immunogen is able to elicit high neutralizing antibody titers against MERS-CoV. We also detd. high-resoln. structures of the trimeric MERS-CoV S ectodomain in complex with G4, a stem-directed neutralizing antibody. The structures reveal that G4 recognizes a glycosylated loop that is variable among coronaviruses and they define four conformational states of the trimer wherein each receptor-binding domain is either tightly packed at the membrane-distal apex or rotated into a receptor-accessible conformation. Our studies suggest a potential mechanism for fusion initiation through sequential receptor-binding events and provide a foundation for the structure-based design of coronavirus vaccines.
- 23Walls, A. C.; Tortorici, M. A.; Snijder, J.; Xiong, X.; Bosch, B. J.; Rey, F. A.; Veesler, D. Tectonic Conformational Changes of a Coronavirus Spike Glycoprotein Promote Membrane Fusion. Proc. Natl. Acad. Sci. U. S. A. 2017, 114 (42), 11157– 11162, DOI: 10.1073/pnas.1708727114[Crossref], [PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1SksLjI&md5=a5b64abf0a761b46c0098e63f57369c4Tectonic conformational changes of a coronavirus spike glycoprotein promote membrane fusionWalls, Alexandra C.; Tortorici, M. Alejandra; Snijder, Joost; Xiong, Xiaoli; Bosch, Berend-Jan; Rey, Felix A.; Veesler, DavidProceedings of the National Academy of Sciences of the United States of America (2017), 114 (42), 11157-11162CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The tremendous pandemic potential of coronaviruses was demonstrated twice in the past few decades by 2 global outbreaks of deadly pneumonia. The coronavirus spike (S) glycoprotein initiates infection by promoting fusion of the viral and cellular membranes through conformational changes that remain largely uncharacterized. Here, we report the cryo-electron microscopy (cryo-EM) structure of a coronavirus S glycoprotein in the post-fusion state, showing large-scale secondary, tertiary, and quaternary rearrangements compared with the pre-fusion trimer and rationalizing the free-energy landscape of this conformational machine. We also biochem. characterized the mol. events assocd. with refolding of the metastable pre-fusion S glycoprotein to the post-fusion conformation using limited proteolysis, mass spectrometry, and single-particle EM. The obsd. similarity between post-fusion coronavirus S and paramyxovirus F structures demonstrated that a conserved refolding trajectory mediates entry of these viruses and supports the evolutionary relatedness of their fusion subunits. Finally, our data provide a structural framework for understanding the mode of neutralization of antibodies targeting the fusion machinery and for engineering next-generation subunit vaccines or inhibitors against this medically important virus family.
- 24Berger, I.; Schaffitzel, C. The SARS-CoV-2 Spike Protein: Balancing Stability and Infectivity. Cell Research; Springer Nature: November 2, 2020; pp 1059– 1060. DOI: 10.1038/s41422-020-00430-4 .
- 25Himes, B. A.; Zhang, P. EmClarity: Software for High-Resolution Cryo-Electron Tomography and Subtomogram Averaging. Nat. Methods 2018, 15 (11), 955– 961, DOI: 10.1038/s41592-018-0167-z[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvF2lsbrE&md5=1358ebf4bdbb9e14df75ffafec9495ebemClarity: software for high-resolution cryo-electron tomography and subtomogram averagingHimes, Benjamin A.; Zhang, PeijunNature Methods (2018), 15 (11), 955-961CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)Macromol. complexes are intrinsically flexible and often challenging to purify for structure detn. by single-particle cryo-electron microscopy (cryo-EM). Such complexes can be studied by cryo-electron tomog. (cryo-ET) combined with subtomogram alignment and classification, which in exceptional cases achieves subnanometer resoln., yielding insight into structure-function relationships. However, it remains challenging to apply this approach to specimens that exhibit conformational or compositional heterogeneity or are present in low abundance. To address this, we developed emClarity (https://github.com/bHimes/emClarity/wiki), a GPU-accelerated image-processing package featuring an iterative tomog. tilt-series refinement algorithm that uses subtomograms as fiducial markers and a 3D-sampling-function-compensated, multi-scale principal component anal. classification method. We demonstrate that our approach offers substantial improvement in the resoln. of maps and in the sepn. of different functional states of macromol. complexes compared with current state-of-the-art software.
- 26Turoňová, B.; Sikora, M.; Schürmann, C.; Hagen, W.; Welsch, S.; Blanc, F.; von Bülow, S.; Gecht, M.; Bagola, K.; Hörner, C.; In Situ Structural Analysis of SARS-CoV-2 Spike Reveals Flexibility Mediated by Three Hinges. bioRxiv 2020, DOI: 10.1101/2020.06.26.173476 .
- 27Yao, H.; Song, Y.; Chen, Y.; Wu, N.; Xu, J.; Sun, C.; Zhang, J.; Weng, T.; Zhang, Z.; Wu, Z. Molecular Architecture of the SARS-CoV-2 Virus. Cell 2020, 183, 730, DOI: 10.1016/j.cell.2020.09.018[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFKjs7nM&md5=7bd110334cb42931034a4eb5f9d2ff00Molecular Architecture of the SARS-CoV-2 VirusYao, Hangping; Song, Yutong; Chen, Yong; Wu, Nanping; Xu, Jialu; Sun, Chujie; Zhang, Jiaxing; Weng, Tianhao; Zhang, Zheyuan; Wu, Zhigang; Cheng, Linfang; Shi, Danrong; Lu, Xiangyun; Lei, Jianlin; Crispin, Max; Shi, Yigong; Li, Lanjuan; Li, SaiCell (Cambridge, MA, United States) (2020), 183 (3), 730-738.e13CODEN: CELLB5; ISSN:0092-8674. (Cell Press)SARS-CoV-2 is an enveloped virus responsible for the COVID-19 pandemic. Despite recent advances in the structural elucidation of SARS-CoV-2 proteins, the detailed architecture of the intact virus remains to be unveiled. Here we report the mol. assembly of the authentic SARS-CoV-2 virus using cryoelectron tomog. (cryo-ET) and subtomogram averaging (STA). Native structures of the S proteins in pre- and postfusion conformations were detd. to av. resolns. of 8.7-11 Å. Compns. of the N-linked glycans from the native spikes were analyzed by mass spectrometry, which revealed overall processing states of the native glycans highly similar to that of the recombinant glycoprotein glycans. The native conformation of the ribonucleoproteins (RNPs) and their higher-order assemblies were revealed. Overall, these characterizations revealed the architecture of the SARS-CoV-2 virus in exceptional detail and shed light on how the virus packs its ∼30-kb-long single-segmented RNA in the ∼80-nm-diam. lumen.
- 28Ke, Z.; Oton, J.; Qu, K.; Cortese, M.; Zila, V.; McKeane, L.; Nakane, T.; Zivanov, J.; Neufeldt, C. J.; Cerikan, B. Structures and Distributions of SARS-CoV-2 Spike Proteins on Intact Virions. Nature 2020, 1– 5, DOI: 10.1038/s41586-020-2665-2
- 29Toelzer, C.; Gupta, K.; Yadav, S. K. N.; Borucu, U.; Davidson, A. D.; Kavanagh Williamson, M.; Shoemark, D. K.; Garzoni, F.; Staufer, O.; Milligan, R. Free Fatty Acid Binding Pocket in the Locked Structure of SARS-CoV-2 Spike Protein. Science 2020, 370 (6517), 725– 730, DOI: 10.1126/science.abd3255[Crossref], [PubMed], [CAS], Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1Kqsr7I&md5=ab0610e4c61dd1cbf7500004cee9bd30Free fatty acid binding pocket in the locked structure of SARS-CoV-2 spike proteinToelzer, Christine; Gupta, Kapil; Yadav, Sathish K. N.; Borucu, Ufuk; Davidson, Andrew D.; Kavanagh Williamson, Maia; Shoemark, Deborah K.; Garzoni, Frederic; Staufer, Oskar; Milligan, Rachel; Capin, Julien; Mulholland, Adrian J.; Spatz, Joachim; Fitzgerald, Daniel; Berger, Imre; Schaffitzel, ChristianeScience (Washington, DC, United States) (2020), 370 (6517), 725-730CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents a global crisis. Key to SARS-CoV-2 therapeutic development is unraveling the mechanisms that drive high infectivity, broad tissue tropism, and severe pathol. Our 2.85-angstrom cryo-electron microscopy structure of SARS-CoV-2 spike (S) glycoprotein reveals that the receptor-binding domains tightly bind the essential free fatty acid linoleic acid (LA) in 3 composite binding pockets. A similar pocket also appears to be present in the highly pathogenic severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). LA binding stabilizes a locked S conformation, resulting in reduced angiotensin-converting enzyme 2 (ACE2) interaction in vitro. In human cells, LA supplementation synergizes with the COVID-19 drug remdesivir, suppressing SARS-CoV-2 replication. Our structure directly links LA and S, setting the stage for intervention strategies that target LA binding by SARS-CoV-2.
- 30Watanabe, Y.; Bowden, T. A.; Wilson, I. A.; Crispin, M. Exploitation of Glycosylation in Enveloped Virus Pathobiology. Biochim. Biophys. Acta, Gen. Subj. 2019, 1863 (10), 1480– 1497, DOI: 10.1016/j.bbagen.2019.05.012[Crossref], [PubMed], [CAS], Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtVynsbvE&md5=118babe3b318937a35757e0cd5b57521Exploitation of glycosylation in enveloped virus pathobiologyWatanabe, Yasunori; Bowden, Thomas A.; Wilson, Ian A.; Crispin, MaxBiochimica et Biophysica Acta, General Subjects (2019), 1863 (10), 1480-1497CODEN: BBGSB3; ISSN:0304-4165. (Elsevier B.V.)A review. Glycosylation is a ubiquitous post-translational modification responsible for a multitude of crucial biol. roles. As obligate parasites, viruses exploit host-cell machinery to glycosylate their own proteins during replication. Viral envelope proteins from a variety of human pathogens including HIV-1, influenza virus, Lassa virus, SARS, Zika virus, dengue virus, and Ebola virus have evolved to be extensively glycosylated. These host-cell derived glycans facilitate diverse structural and functional roles during the viral life-cycle, ranging from immune evasion by glycan shielding to enhancement of immune cell infection. In this review, we highlight the imperative and auxiliary roles glycans play, and how specific oligosaccharide structures facilitate these functions during viral pathogenesis. We discuss the growing efforts to exploit viral glycobiol. in the development of anti-viral vaccines and therapies.
- 31Cao, L.; Pauthner, M.; Andrabi, R.; Rantalainen, K.; Berndsen, Z.; Diedrich, J. K.; Menis, S.; Sok, D.; Bastidas, R.; Park, S.-K. R. Differential Processing of HIV Envelope Glycans on the Virus and Soluble Recombinant Trimer. Nat. Commun. 2018, 9 (1), 3693, DOI: 10.1038/s41467-018-06121-4[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c3ot1aksA%253D%253D&md5=d4daee53f36dffe65dff73789b3bf39eDifferential processing of HIV envelope glycans on the virus and soluble recombinant trimerCao Liwei; Diedrich Jolene K; Park Sung-Kyu Robin; Delahunty Claire M; He Lin; Yates John R 3rd; Paulson James C; Cao Liwei; Pauthner Matthias; Andrabi Raiees; Menis Sergey; Sok Devin; Bastidas Raiza; Guenaga Javier; Wyatt Richard T; Schief William R; Burton Dennis R; Paulson James C; Cao Liwei; Pauthner Matthias; Andrabi Raiees; Rantalainen Kimmo; Berndsen Zachary; Menis Sergey; Sok Devin; Bastidas Raiza; Guenaga Javier; Wyatt Richard T; Schief William R; Ward Andrew B; Burton Dennis R; Paulson James C; Rantalainen Kimmo; Berndsen Zachary; Ward Andrew B; Burton Dennis RNature communications (2018), 9 (1), 3693 ISSN:.As the sole target of broadly neutralizing antibodies (bnAbs) to HIV, the envelope glycoprotein (Env) trimer is the focus of vaccination strategies designed to elicit protective bnAbs in humans. Because HIV Env is densely glycosylated with 75-90 N-glycans per trimer, most bnAbs use or accommodate them in their binding epitope, making the glycosylation of recombinant Env a key aspect of HIV vaccine design. Upon analysis of three HIV strains, we here find that site-specific glycosylation of Env from infectious virus closely matches Envs from corresponding recombinant membrane-bound trimers. However, viral Envs differ significantly from recombinant soluble, cleaved (SOSIP) Env trimers, strongly impacting antigenicity. These results provide a benchmark for virus Env glycosylation needed for the design of soluble Env trimers as part of an overall HIV vaccine strategy.
- 32Cao, L.; Diedrich, J. K.; Kulp, D. W.; Pauthner, M.; He, L.; Park, S.-K. R.; Sok, D.; Su, C. Y.; Delahunty, C. M.; Menis, S. Global Site-Specific N-Glycosylation Analysis of HIV Envelope Glycoprotein. Nat. Commun. 2017, 8, 14954, DOI: 10.1038/ncomms14954[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlt12nu7w%253D&md5=eb9e80327cbc6e95a3c8ee602e7c14cdGlobal site-specific N-glycosylation analysis of HIV envelope glycoproteinCao, Liwei; Diedrich, Jolene K.; Kulp, Daniel W.; Pauthner, Matthias; He, Lin; Park, Sung-Kyu Robin; Sok, Devin; Su, Ching Yao; Delahunty, Claire M.; Menis, Sergey; Andrabi, Raiees; Guenaga, Javier; Georgeson, Erik; Kubitz, Michael; Adachi, Yumiko; Burton, Dennis R.; Schief, William R.; Yates, John R., III; Paulson, James C.Nature Communications (2017), 8 (), 14954CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)HIV-1 envelope glycoprotein (Env) is the sole target for broadly neutralizing antibodies (bnAbs) and the focus for design of an antibody-based HIV vaccine. The Env trimer is covered by ∼90N-linked glycans, which shield the underlying protein from immune surveillance. BNAbs to HIV develop during infection, with many showing dependence on glycans for binding to Env. The ability to routinely assess the glycan type at each glycosylation site may facilitate design of improved vaccine candidates. Here we present a general mass spectrometry-based proteomics strategy that uses specific endoglycosidases to introduce mass signatures that distinguish peptide glycosites that are unoccupied or occupied by high-mannose/hybrid or complex-type glycans. The method yields >95% sequence coverage for Env, provides semi-quant. anal. of the glycosylation status at each glycosite. We find that most glycosites in recombinant Env trimers are fully occupied by glycans, varying in the proportion of high-mannose/hybrid and complex-type glycans.
- 33Struwe, W. B.; Chertova, E.; Allen, J. D.; Seabright, G. E.; Watanabe, Y.; Harvey, D. J.; Medina-Ramirez, M.; Roser, J. D.; Smith, R.; Westcott, D. Site-Specific Glycosylation of Virion-Derived HIV-1 Env Is Mimicked by a Soluble Trimeric Immunogen. Cell Rep. 2018, 24 (8), 1958– 1966, e5 DOI: 10.1016/j.celrep.2018.07.080[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFGqtLzL&md5=2113b0b4d0702bc1cb54ade8dfc1b0bbSite-Specific Glycosylation of Virion-Derived HIV-1 Env Is Mimicked by a Soluble Trimeric ImmunogenStruwe, Weston B.; Chertova, Elena; Allen, Joel D.; Seabright, Gemma E.; Watanabe, Yasunori; Harvey, David J.; Medina-Ramirez, Max; Roser, James D.; Smith, Rodman; Westcott, David; Keele, Brandon F.; Bess, Julian W. Jr.; Sanders, Rogier W.; Lifson, Jeffrey D.; Moore, John P.; Crispin, MaxCell Reports (2018), 24 (8), 1958-1966.e5CODEN: CREED8; ISSN:2211-1247. (Cell Press)Many broadly neutralizing antibodies (bnAbs) against HIV-1 recognize and/or penetrate the glycan shield on native, virion-assocd. envelope glycoprotein (Env) spikes. The same bnAbs also bind to recombinant, sol. trimeric immunogens based on the SOSIP design. While SOSIP trimers are close structural and antigenic mimics of virion Env, the extent to which their glycan structures resemble ones on infectious viruses is undefined. Here, we compare the overall glycosylation of gp120 and gp41 subunits from BG505 (clade A) virions produced in a lymphoid cell line with those from recombinant BG505 SOSIP trimers, including CHO-derived clin. grade material. We also performed detailed site-specific analyses of gp120. Glycans relevant to key bnAb epitopes are generally similar on the recombinant SOSIP and virion-derived Env proteins, although the latter do contain hotspots of elevated glycan processing. Knowledge of native vs. recombinant Env glycosylation will guide vaccine design and manufg. programs.
- 34Watanabe, Y.; Berndsen, Z. T.; Raghwani, J.; Seabright, G. E.; Allen, J. D.; Pybus, O. G.; McLellan, J. S.; Wilson, I. A.; Bowden, T. A.; Ward, A. B. Vulnerabilities in Coronavirus Glycan Shields despite Extensive Glycosylation. Nat. Commun. 2020, 11 (1), 2688, DOI: 10.1038/s41467-020-16567-0[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVCmu77L&md5=2c0ce9b10f2a8d10d6889e975e9e09b3Vulnerabilities in coronavirus glycan shields despite extensive glycosylationWatanabe, Yasunori; Berndsen, Zachary T.; Raghwani, Jayna; Seabright, Gemma E.; Allen, Joel D.; Pybus, Oliver G.; McLellan, Jason S.; Wilson, Ian A.; Bowden, Thomas A.; Ward, Andrew B.; Crispin, MaxNature Communications (2020), 11 (1), 2688CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: Severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) coronaviruses (CoVs) are zoonotic pathogens with high fatality rates and pandemic potential. Vaccine development focuses on the principal target of the neutralizing humoral immune response, the spike (S) glycoprotein. Coronavirus S proteins are extensively glycosylated, encoding around 66-87 N-linked glycosylation sites per trimeric spike. Here, we reveal a specific area of high glycan d. on MERS S that results in the formation of oligomannose-type glycan clusters, which were absent on SARS and HKU1 CoVs. We provide a comparison of the global glycan d. of coronavirus spikes with other viral proteins including HIV-1 envelope, Lassa virus glycoprotein complex, and influenza hemagglutinin, where glycosylation plays a known role in shielding immunogenic epitopes. Overall, our data reveal how organization of glycosylation across class I viral fusion proteins influence not only individual glycan compns. but also the immunol. pressure across the protein surface.
- 35Zhao, P.; Praissman, J. L.; Grant, O. C.; Chen, B.; Brief, I.; Cai, Y.; Xiao, T.; Rosenbalm, K. E.; Aoki, K.; Kellman, B. P. Virus-Receptor Interactions of Glycosylated SARS-CoV-2 Spike and Human ACE2 Receptor. Cell Host Microbe 2020, 28, 1– 16, DOI: 10.1016/j.chom.2020.08.004
- 36Brun, J.; Vasiljevic, S.; Gangadharan, B.; Hensen, M.; Chandran, A. V.; Hill, M. L.; Kiappes, J. L.; Dwek, R. A.; Alonzi, D. S.; Struwe, W. B.; Analysis of SARS-CoV-2 Spike Glycosylation Reveals Shedding of a Vaccine Candidate. bioRxiv . bioRxiv November 16, 2020; DOI: 10.1101/2020.11.16.384594 .
- 37Morris, S. J.; Sebastian, S.; Spencer, A. J.; Gilbert, S. C. Simian Adenoviruses as Vaccine Vectors. Future Virology; Future Medicine Ltd: September 1, 2016; pp 649– 659. DOI: 10.2217/fvl-2016-0070 .
- 38Dicks, M. D. J.; Spencer, A. J.; Coughlan, L.; Bauza, K.; Gilbert, S. C.; Hill, A. V. S.; Cottingham, M. G. Differential Immunogenicity between HAdV-5 and Chimpanzee Adenovirus Vector ChAdOx1 Is Independent of Fiber and Penton RGD Loop Sequences in Mice. Sci. Rep. 2015, 5 (1), 1– 15, DOI: 10.1038/srep16756
- 39Essalmani, R.; Jain, J.; Susan-Resiga, D.; Andréo, U.; Evagelidis, A.; Derbali, R. M.; Huynh, D. N.; Dallaire, F.; Laporte, M.; Delpal, A.; Furin Cleaves SARS-CoV-2 Spike-Glycoprotein at S1/S2 and S2’ for Viral Fusion/Entry: Indirect Role of TMPRSS2. bioRxiv 2020, DOI: 10.1101/2020.12.18.423106 .
- 40Uhlen, M.; Fagerberg, L.; Hallstrom, B. M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; Asplund, A. Tissue-Based Map of the Human Proteome. Science 2015, 347 (6220), 1260419– 1260419, DOI: 10.1126/science.1260419[Crossref], [PubMed], [CAS], Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2MvptFakug%253D%253D&md5=95510888806e5feb6b8024ee2a417d1cProteomics. Tissue-based map of the human proteomeUhlen Mathias; Fagerberg Linn; Oksvold Per; Sivertsson ¡ÑÜAsa; Lundberg Emma; Odeberg Jacob; Alm Tove; Nilsson Peter; Schwenk Jochen M; von Feilitzen Kalle; Forsberg Mattias; Persson Lukas; Johansson Fredric; Zwahlen Martin; Hallstrom Bjorn M; Lindskog Cecilia; Kampf Caroline; Asplund Anna; Olsson IngMarie; Djureinovic Dijana; Edqvist Per-Henrik; Ponten Fredrik; Mardinoglu Adil; Sjostedt Evelina; Edlund Karolina; Navani Sanjay; Szigyarto Cristina Al-Khalili; Takanen Jenny Ottosson; Hober Sophia; Berling Holger; Tegel Hanna; Rockberg Johan; Hamsten Marica; Mulder Jan; von Heijne Gunnar; Nielsen JensScience (New York, N.Y.) (2015), 347 (6220), 1260419 ISSN:.Resolving the molecular details of proteome variation in the different tissues and organs of the human body will greatly increase our knowledge of human biology and disease. Here, we present a map of the human tissue proteome based on an integrated omics approach that involves quantitative transcriptomics at the tissue and organ level, combined with tissue microarray-based immunohistochemistry, to achieve spatial localization of proteins down to the single-cell level. Our tissue-based analysis detected more than 90% of the putative protein-coding genes. We used this approach to explore the human secretome, the membrane proteome, the druggable proteome, the cancer proteome, and the metabolic functions in 32 different tissues and organs. All the data are integrated in an interactive Web-based database that allows exploration of individual proteins, as well as navigation of global expression patterns, in all major tissues and organs in the human body.
- 41Hamming, I.; Timens, W.; Bulthuis, M. L. C.; Lely, A. T.; Navis, G. J.; van Goor, H. Tissue Distribution of ACE2 Protein, the Functional Receptor for SARS Coronavirus. A First Step in Understanding SARS Pathogenesis. J. Pathol. 2004, 203 (2), 631– 637, DOI: 10.1002/path.1570[Crossref], [PubMed], [CAS], Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXlsVWktbY%253D&md5=0e7e65a40dd1d6cc50c0d1d28acf41cfTissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesisHamming, I.; Timens, W.; Bulthuis, M. L. C.; Lely, A. T.; Navis, G. J.; van Goor, H.Journal of Pathology (2004), 203 (2), 631-637CODEN: JPTLAS; ISSN:0022-3417. (John Wiley & Sons Ltd.)Severe acute respiratory syndrome (SARS) is an acute infectious disease that spreads mainly via the respiratory route. A distinct coronavirus (SARS-CoV) has been identified as the etiol. agent of SARS. Recently, a metallopeptidase named angiotensin-converting enzyme 2 (ACE2) has been identified as the functional receptor for SARS-CoV. Although ACE2 mRNA is known to be present in virtually all organs, its protein expression is largely unknown. Since identifying the possible route of infection has major implications for understanding the pathogenesis and future treatment strategies for SARS, the present study investigated the localization of ACE2 protein in various human organs (oral and nasal mucosa, nasopharynx, lung, stomach, small intestine, colon, skin, lymph nodes, thymus, bone marrow, spleen, liver, kidney, and brain). The most remarkable finding was the surface expression of ACE2 protein on lung alveolar epithelial cells and enterocytes of the small intestine. Furthermore, ACE2 was present in arterial and venous endothelial cells and arterial smooth muscle cells in all organs studied. In conclusion, ACE2 is abundantly present in humans in the epithelia of the lung and small intestine, which might provide possible routes of entry for the SARS-CoV. This epithelial expression, together with the presence of ACE2 in vascular endothelium, also provides a first step in understanding the pathogenesis of the main SARS disease manifestations.
- 42Sungnak, W.; Huang, N.; Bécavin, C.; Berg, M.; Queen, R.; Litvinukova, M.; Talavera-López, C.; Maatz, H.; Reichart, D.; Sampaziotis, F. SARS-CoV-2 Entry Factors Are Highly Expressed in Nasal Epithelial Cells Together with Innate Immune Genes. Nat. Med. 2020, 26 (5), 681– 687, DOI: 10.1038/s41591-020-0868-6[Crossref], [PubMed], [CAS], Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXotVCjurY%253D&md5=cc313df9c190333b98af43951a2146faSARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genesSungnak, Waradon; Huang, Ni; Becavin, Christophe; Berg, Marijn; Queen, Rachel; Litvinukova, Monika; Talavera-Lopez, Carlos; Maatz, Henrike; Reichart, Daniel; Sampaziotis, Fotios; Worlock, Kaylee B.; Yoshida, Masahiro; Barnes, Josephine L.; HCA Lung Biological NetworkNature Medicine (New York, NY, United States) (2020), 26 (5), 681-687CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)We investigated SARS-CoV-2 potential tropism by surveying expression of viral entry-assocd. genes in single-cell RNA-sequencing data from multiple tissues from healthy human donors. We co-detected these transcripts in specific respiratory, corneal and intestinal epithelial cells, potentially explaining the high efficiency of SARS-CoV-2 transmission. These genes are co-expressed in nasal epithelial cells with genes involved in innate immunity, highlighting the cells' potential role in initial viral infection, spread and clearance. The study offers a useful resource for further lines of inquiry with valuable clin. samples from COVID-19 patients and we provide our data in a comprehensive, open and user-friendly fashion at www.covid19cellatlas.org.
- 43Kornfeld, R.; Kornfeld, S. Assembly of Asparagine-Linked Oligosaccharides. Annu. Rev. Biochem. 1985, 54 (1), 631– 664, DOI: 10.1146/annurev.bi.54.070185.003215[Crossref], [PubMed], [CAS], Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaL2M3nvVSjsA%253D%253D&md5=e4e0de7e37f61bbe1c7bb100927460ceAssembly of asparagine-linked oligosaccharidesKornfeld R; Kornfeld SAnnual review of biochemistry (1985), 54 (), 631-64 ISSN:0066-4154.There is no expanded citation for this reference.
- 44Gagneux, P.; Aebi, M.; Varki, A. Evolution of Glycan Diversity; Cold Spring Harbor Laboratory Press: 2015. DOI: 10.1101/GLYCOBIOLOGY.3E.020 .
- 45Mastronarde, D. N. Automated Electron Microscope Tomography Using Robust Prediction of Specimen Movements. J. Struct. Biol. 2005, 152 (1), 36– 51, DOI: 10.1016/j.jsb.2005.07.007[Crossref], [PubMed], [CAS], Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2MrjtVSrtw%253D%253D&md5=16b6502753c0f1f63ff559a784bce8f0Automated electron microscope tomography using robust prediction of specimen movementsMastronarde David NJournal of structural biology (2005), 152 (1), 36-51 ISSN:1047-8477.A new method was developed to acquire images automatically at a series of specimen tilts, as required for tomographic reconstruction. The method uses changes in specimen position at previous tilt angles to predict the position at the current tilt angle. Actual measurement of the position or focus is skipped if the statistical error of the prediction is low enough. This method allows a tilt series to be acquired rapidly when conditions are good but falls back toward the traditional approach of taking focusing and tracking images when necessary. The method has been implemented in a program, SerialEM, that provides an efficient environment for data acquisition. This program includes control of an energy filter as well as a low-dose imaging mode, in which tracking and focusing occur away from the area of interest. The program can automatically acquire a montage of overlapping frames, allowing tomography of areas larger than the field of the CCD camera. It also includes tools for navigating between specimen positions and finding regions of interest.
- 46Hagen, W. J. H.; Wan, W.; Briggs, J. A. G. Implementation of a Cryo-Electron Tomography Tilt-Scheme Optimized for High Resolution Subtomogram Averaging. J. Struct. Biol. 2017, 197 (2), 191– 198, DOI: 10.1016/j.jsb.2016.06.007[Crossref], [PubMed], [CAS], Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2s7gvFGksw%253D%253D&md5=e729954381b30719b7f07eaff9eaa496Implementation of a cryo-electron tomography tilt-scheme optimized for high resolution subtomogram averagingHagen Wim J H; Wan William; Briggs John A GJournal of structural biology (2017), 197 (2), 191-198 ISSN:.Cryo-electron tomography (cryoET) allows 3D structural information to be obtained from cells and other biological samples in their close-to-native state. In combination with subtomogram averaging, detailed structures of repeating features can be resolved. CryoET data is collected as a series of images of the sample from different tilt angles; this is performed by physically rotating the sample in the microscope between each image. The angles at which the images are collected, and the order in which they are collected, together are called the tilt-scheme. Here we describe a "dose-symmetric tilt-scheme" that begins at low tilt and then alternates between increasingly positive and negative tilts. This tilt-scheme maximizes the amount of high-resolution information maintained in the tomogram for subsequent subtomogram averaging, and may also be advantageous for other applications. We describe implementation of the tilt-scheme in combination with further data-collection refinements including setting thresholds on acceptable drift and improving focus accuracy. Requirements for microscope set-up are introduced, and a macro is provided which automates the application of the tilt-scheme within SerialEM.
- 47Zheng, S. Q.; Palovcak, E.; Armache, J. P.; Verba, K. A.; Cheng, Y.; Agard, D. A. MotionCor2: Anisotropic Correction of Beam-Induced Motion for Improved Cryo-Electron Microscopy. Nat. Methods 2017, 14 (4), 331– 332, DOI: 10.1038/nmeth.4193[Crossref], [PubMed], [CAS], Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjt1ags7g%253D&md5=5f4e225ef8123dacd8475d526175e1d2MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopyZheng, Shawn Q.; Palovcak, Eugene; Armache, Jean-Paul; Verba, Kliment A.; Cheng, Yifan; Agard, David A.Nature Methods (2017), 14 (4), 331-332CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)A review on anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Here we describe MotionCor2, a software tool for anisotropic correction of beam-induced motion. Overall, MotionCor2 is extremely robust and sufficiently accurate at correcting local motions so that the very time-consuming and computationally intensive particle polishing in RELION can be skipped, importantly, it also works on a wide range of data sets, including cryo tomog. tilt series.
- 48Kremer, J. R.; Mastronarde, D. N.; McIntosh, J. R. Computer Visualization of Three-Dimensional Image Data Using IMOD. J. Struct. Biol. 1996, 116 (1), 71– 76, DOI: 10.1006/jsbi.1996.0013[Crossref], [PubMed], [CAS], Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK28zmtFyqtg%253D%253D&md5=b6ab779bdec60f8644e254124c4dbbe7Computer visualization of three-dimensional image data using IMODKremer J R; Mastronarde D N; McIntosh J RJournal of structural biology (1996), 116 (1), 71-6 ISSN:1047-8477.We have developed a computer software package, IMOD, as a tool for analyzing and viewing three-dimensional biological image data. IMOD is useful for studying and modeling data from tomographic, serial section, and optical section reconstructions. The software allows image data to be visualized by several different methods. Models of the image data can be visualized by volume or contour surface rendering and can yield quantitative information.
- 49Pettersen, E. F.; Goddard, T. D.; Huang, C. C.; Couch, G. S.; Greenblatt, D. M.; Meng, E. C.; Ferrin, T. E. UCSF Chimera - A Visualization System for Exploratory Research and Analysis. J. Comput. Chem. 2004, 25 (13), 1605– 1612, DOI: 10.1002/jcc.20084[Crossref], [PubMed], [CAS], Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmvVOhsbs%253D&md5=944b175f440c1ff323705987cf937ee7UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 50Shevchenko, A.; Tomas, H.; Havli, J.; Olsen, J. V.; Mann, M. In-Gel Digestion for Mass Spectrometric Characterization of Proteins and Proteomes. Nat. Protoc. 2006, 1 (6), 2856– 2860, DOI: 10.1038/nprot.2006.468[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtFGjt7nM&md5=7c82516bf8197dff95fe73a371af2f0cIn-gel digestion for mass spectrometric characterization of proteins and proteomesShevchenko, Andrej; Tomas, Henrik; Havlis, Jan; Olsen, Jesper V.; Mann, MatthiasNature Protocols (2006), 1 (6), 2856-2860CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)In-gel digestion of proteins isolated by gel electrophoresis is a cornerstone of mass spectrometry (MS)-driven proteomics. The 10-yr-old recipe by Shevchenko et al. has been optimized to increase the speed and sensitivity of anal. The protocol is for the in-gel digestion of both silver and Coomassie-stained protein spots or bands and can be followed by MALDI-MS or LC-MS/MS anal. to identify proteins at sensitivities better than a few femtomoles of protein starting material.
- 51Emsley, P.; Crispin, M. Structural Analysis of Glycoproteins: Building N-Linked Glycans with Coot. Acta Crystallogr. Sect. D Struct. Biol. 2018, 74 (4), 256– 263, DOI: 10.1107/S2059798318005119[Crossref], [PubMed], [CAS], Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1KhsL8%253D&md5=b6324f66f05fbd26eed4d6b1cc6eb89eStructural analysis of glycoproteins: building N-linked glycans with CootEmsley, Paul; Crispin, MaxActa Crystallographica, Section D: Structural Biology (2018), 74 (4), 256-263CODEN: ACSDAD; ISSN:2059-7983. (International Union of Crystallography)Coot is a graphics application that is used to build or manipulate macromol. models; its particular forte is manipulation of the model at the residue level. The model-building tools of Coot have been combined and extended to assist or automate the building of N-linked glycans. The model is built by the addn. of monosaccharides, placed by variation of internal coordinates. The subsequent model is refined by real-space refinement, which is stabilized with modified and addnl. restraints. It is hoped that these enhanced building tools will help to reduce building errors of N-linked glycans and improve our knowledge of the structures of glycoproteins.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscentsci.1c00080.
Supplementary Figures 1–5 and Table 1; FACS gating strategy, tomographic slices, cryo-EM images, site-specific glycosylation comparisons between recombinant proteins, and values for glycoforms observed on S proteins derived from ChAdOx1 nCoV-19 (PDF)
Movie 1: Cryo-electron tomography of U2OS cells infected with ChAdOx1 nCoV-19 (MP4)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.