3
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Competitive SARS-CoV-2 Serology Reveals Most Antibodies Targeting the Spike Receptor-Binding Domain Compete for ACE2 Binding

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          With the emergence and continued spread of the SARS-CoV-2 virus, and of the associated disease, coronavirus disease 2019 (COVID-19), there is an urgent need for improved understanding of how the body mounts an immune response to the virus. Here, we developed a competitive SARS-CoV-2 serological assay that can simultaneously determine whether an individual has developed antibodies against the SARS-CoV-2 Spike protein receptor-binding domain (RBD) and measure the proportion of these antibodies that block interaction with the human angiotensin-converting enzyme 2 (ACE2) required for viral entry. Using this assay and 144 SARS-CoV-2 patient serum samples, we found that a majority of anti-RBD antibodies compete for ACE2 binding. These results not only highlight the need to design vaccines to generate such blocking antibodies but also demonstrate the utility of this assay to rapidly screen patient sera for potentially neutralizing antibodies.

          ABSTRACT

          As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread around the world, there is an urgent need for new assay formats to characterize the humoral response to infection. Here, we present an efficient, competitive serological assay that can simultaneously determine an individual’s seroreactivity against the SARS-CoV-2 Spike protein and determine the proportion of anti-Spike antibodies that block interaction with the human angiotensin-converting enzyme 2 (ACE2) required for viral entry. In this approach based on the use of enzyme-linked immunosorbent assays (ELISA), we present natively folded viral Spike protein receptor-binding domain (RBD)-containing antigens via avidin-biotin interactions. Sera are then competed with soluble ACE2-Fc, or with a higher-affinity variant thereof, to determine the proportion of ACE2 blocking anti-RBD antibodies. Assessment of sera from 144 SARS-CoV-2 patients ultimately revealed that a remarkably consistent and high proportion of antibodies in the anti-RBD pool targeted the epitope responsible for ACE2 engagement (83% ± 11%; 50% to 107% signal inhibition in our largest cohort), further underscoring the importance of tailoring vaccines to promote the development of such antibodies.

          IMPORTANCE With the emergence and continued spread of the SARS-CoV-2 virus, and of the associated disease, coronavirus disease 2019 (COVID-19), there is an urgent need for improved understanding of how the body mounts an immune response to the virus. Here, we developed a competitive SARS-CoV-2 serological assay that can simultaneously determine whether an individual has developed antibodies against the SARS-CoV-2 Spike protein receptor-binding domain (RBD) and measure the proportion of these antibodies that block interaction with the human angiotensin-converting enzyme 2 (ACE2) required for viral entry. Using this assay and 144 SARS-CoV-2 patient serum samples, we found that a majority of anti-RBD antibodies compete for ACE2 binding. These results not only highlight the need to design vaccines to generate such blocking antibodies but also demonstrate the utility of this assay to rapidly screen patient sera for potentially neutralizing antibodies.

          Related collections

          Most cited references15

          • Record: found
          • Abstract: found
          • Article: found

          Emerging coronaviruses: Genome structure, replication, and pathogenesis

          Abstract The recent emergence of a novel coronavirus (2019‐nCoV), which is causing an outbreak of unusual viral pneumonia in patients in Wuhan, a central city in China, is another warning of the risk of CoVs posed to public health. In this minireview, we provide a brief introduction of the general features of CoVs and describe diseases caused by different CoVs in humans and animals. This review will help understand the biology and potential risk of CoVs that exist in richness in wildlife such as bats.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2

            Summary We have previously provided the first genetic evidence that angiotensin converting enzyme 2 (ACE2) is the critical receptor for severe acute respiratory syndrome coronavirus (SARS-CoV), and ACE2 protects the lung from injury, providing a molecular explanation for the severe lung failure and death due to SARS-CoV infections. ACE2 has now also been identified as a key receptor for SARS-CoV-2 infections, and it has been proposed that inhibiting this interaction might be used in treating patients with COVID-19. However, it is not known whether human recombinant soluble ACE2 (hrsACE2) blocks growth of SARS-CoV-2. Here, we show that clinical grade hrsACE2 reduced SARS-CoV-2 recovery from Vero cells by a factor of 1,000–5,000. An equivalent mouse rsACE2 had no effect. We also show that SARS-CoV-2 can directly infect engineered human blood vessel organoids and human kidney organoids, which can be inhibited by hrsACE2. These data demonstrate that hrsACE2 can significantly block early stages of SARS-CoV-2 infections.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Convergent Antibody Responses to SARS-CoV-2 in Convalescent Individuals

              During the COVID-19 pandemic, SARS-CoV-2 infected millions of people and claimed hundreds of thousands of lives. Virus entry into cells depends on the receptor binding domain (RBD) of the SARS-CoV-2 spike protein (S). Although there is 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 1–5 . Here we report on 149 COVID-19 convalescent individuals. Plasmas collected an average of 39 days after the onset of symptoms had variable half-maximal pseudovirus neutralizing titers: less than 1:50 in 33% and below 1:1000 in 79%, while only 1% showed titers >1:5000. Antibody sequencing revealed expanded clones of RBD-specific memory B cells expressing closely related antibodies in different individuals. Despite low plasma titers, antibodies to three distinct epitopes on RBD neutralized at half-maximal inhibitory concentrations (IC50s) as low as single digit ng/mL. Thus, most convalescent plasmas 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.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                mSphere
                mSphere
                msph
                msph
                mSphere
                mSphere
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2379-5042
                16 September 2020
                Sep-Oct 2020
                16 September 2020
                : 5
                : 5
                : e00802-20
                Affiliations
                [a ]Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
                [b ]Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, USA
                [c ]Department of Neurology, University of California, San Francisco, San Francisco, California, USA
                [d ]Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California, USA
                [e ]Department of Medicine, University of California, San Francisco, San Francisco, California, USA
                [f ]Chan Zuckerberg Biohub, San Francisco, California, USA
                [g ]Department of Medicine, Stanford University Medical School, Stanford, California, USA
                [h ]Department of Microbiology and Immunology, Stanford University Medical School, Stanford, California, USA
                [i ]Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California, USA
                National Institute of Allergy and Infectious Diseases
                Author notes
                Address correspondence to James A. Wells, jim.wells@ 123456ucsf.edu .

                Citation Byrnes JR, Zhou XX, Lui I, Elledge SK, Glasgow JE, Lim SA, Loudermilk RP, Chiu CY, Wang TT, Wilson MR, Leung KK, Wells JA. 2020. Competitive SARS-CoV-2 serology reveals most antibodies targeting the Spike receptor-binding domain compete for ACE2 binding. mSphere 5:e00802-20. https://doi.org/10.1128/mSphere.00802-20.

                Author information
                https://orcid.org/0000-0003-0297-1209
                https://orcid.org/0000-0002-8198-2856
                https://orcid.org/0000-0002-6171-5443
                https://orcid.org/0000-0002-9621-3881
                https://orcid.org/0000-0001-6699-8983
                https://orcid.org/0000-0003-2136-2732
                https://orcid.org/0000-0001-9970-5112
                https://orcid.org/0000-0003-2915-2094
                https://orcid.org/0000-0002-3894-685X
                https://orcid.org/0000-0002-8705-5084
                https://orcid.org/0000-0002-2087-4974
                https://orcid.org/0000-0001-8267-5519
                Article
                mSphere00802-20
                10.1128/mSphere.00802-20
                7494835
                32938700
                2203cf53-275e-4178-aa4d-39099bb53681
                Copyright © 2020 Byrnes et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 7 August 2020
                : 8 September 2020
                Page count
                supplementary-material: 10, Figures: 4, Tables: 0, Equations: 0, References: 18, Pages: 11, Words: 7538
                Funding
                Funded by: Charles and Helen Schwab Foundation;
                Award Recipient :
                Funded by: Rachleff Family Endowment;
                Award Recipient :
                Funded by: CEND COVID Catalyst Fund;
                Award Recipient :
                Funded by: Harry and Dianna Hind Endowed Professorship in Pharmaceutical Sciences;
                Award Recipient :
                Funded by: HHS | NIH | National Cancer Institute (NCI), https://doi.org/10.13039/100000054;
                Award ID: 5F32CA239417
                Award Recipient :
                Funded by: HHS | NIH | National Institute of Allergy and Infectious Diseases (NIAID), https://doi.org/10.13039/100000060;
                Award ID: R33-129077
                Award Recipient :
                Funded by: HHS | NIH | National Institute of Allergy and Infectious Diseases (NIAID), https://doi.org/10.13039/100000060;
                Award ID: U19AI111825
                Award ID: R01AI139119
                Award Recipient :
                Funded by: National Science Foundation (NSF), https://doi.org/10.13039/100000001;
                Award ID: 1650113
                Award Recipient : Award Recipient :
                Funded by: HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI), https://doi.org/10.13039/100000050;
                Award ID: R01-HL105704
                Award Recipient :
                Funded by: Helen Hay Whitney Foundation (HHWF), https://doi.org/10.13039/100005237;
                Award Recipient :
                Funded by: Chan Zuckerberg Initiative (CZI), https://doi.org/10.13039/100014989;
                Award Recipient :
                Funded by: Fast Grants, https://doi.org/10.13039/100016127;
                Award Recipient :
                Funded by: Damon Runyon Cancer Research Foundation (DRCRF), https://doi.org/10.13039/100001021;
                Award Recipient :
                Categories
                Research Article
                Clinical Science and Epidemiology
                Custom metadata
                September/October 2020

                covid-19,sars-cov-2,angiotensin-converting enzyme 2,immunoserology,neutralizing antibodies,receptor-binding domain,serology

                Comments

                Comment on this article