24
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Characterization of SARS-CoV-2 Omicron spike RBD reveals significantly decreased stability, severe evasion of neutralizing-antibody recognition but unaffected engagement by decoy ACE2 modified for enhanced RBD binding

      letter

      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

          Dear Editor, The recent-emerging Omicron variant (B.1.1.529 lineage) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has raised serious public concern because of its rapid regional- and global-transmission. As of 11th January 2022, the Omicron variant has spread to 140 countries, territories or areas through infected air travelers, and the number is continuously increasing. 1 Currently, Omicron has outcompeted the Delta variant (B.1.617.2 lineage) in many countries (e.g., USA, United Kingdom, France, Italy, etc.), becoming the dominant circulating variant and causing surges in weekly infections. 1 Therefore, it is an urgent issue to re-evaluate and/or re-develop effective agents to combat the potential Omicron pandemic. The Omicron variant accumulated unusual large number of mutations (over 60 amino-acid substitutions/deletions/insertions) in its genome-encoded proteins. Among these proteins, the surface-located spike (S) that determines viral infectivity and antigenicity, carries 30 amino-acid substitutions, 6 residue deletions, and 3 residue insertions. Most importantly, the receptor-binding domain in spike (S-RBD), which is the main target for therapeutic antibodies and the key component of prophylactic vaccines, harbors 15 substitutions, including G339D, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, and Y505H. Most of the substitutions are near or located on the human angiotensin-converting enzyme 2 (ACE2) binding interface, and all of the substitutions could be mapped to one or more of the known antigenic sites in S-RBD (Fig. 1a), suggesting that S-RBD of the Omicron variant might behave differently from that of the original SARS-CoV-2 strain when interacting with the ACE2 receptor and the therapeutic antibodies. Fig. 1 Antibody-escape profile, receptor-binding capacity, and biochemical property of Omicron variant S-RBD. a Multiple previously identified antigenic sites were mapped on original strain S-RBD (PDB code: 6XC4). The RBS-A, RBS-B, RBS-C, RBS-D were circled on left panel, the CR3022 site and S309 site were circled on right panel. Amino-acid mutations of Omicron variant S-RBD were marked on both panels. b The binding affinities between SARS-CoV-2 S-RBD (original strain and Omicron variant) and each representative antibody (in scFv form) calculated by SPR. The dissociation constant (K D) values and associated affinity-fold decrease [K D (Omicron/Original] were individually shown. The antibodies that have been approved for clinical use were highlighted by shadowing in orange. The related real-time binding profiles were demonstrated in Supplementary Fig. S2. c–h The interaction between SARS-CoV-2 S-RBD (original strain and Omicron variant) and ACE2 proteins [wild-type or affinity-enhanced ACE2 mutants] characterized by SPR. The real-time binding profiles and calculated kinetic parameters are shown. i A DSF assay characterizing the thermostability of original strain and Omicron variant S-RBDs. The fluorescence-unit curve and melting temperature (Tm) for each S-RBD were shown. j, k Protease-digestion assays with fivefold serially diluted Trypsin (j) or Chymotrypsin (k) towards original strain S-RBD and Omicron variant S-RBD In order to evaluate the impact of the Omicron mutations, we targeted the multiple previously identified antigenic sites in S-RBD [including RBS-A, RBS-B, RBS-C, RBS-D, CR3022 site, and S309 site (Fig. 1a)], 2 selected a series of representative neutralizing antibodies for each site (CB6, CC12.3 and P2C-1F11 for RBS-A, CV07-250, 2-4 and REGN10933 for RBS-B, CV07-270, BD-368-2 and LY-CoV555 for RBS-C, REGN10987 for RBD-D, EY6A, S2A4 and H014 for CR3022 site, S309 and C135 for S309 site), and prepared the antibody protein in the single-chain variable fragment (scFv) form by in-vitro refolding method (Supplementary Fig. S1a). For each antibody, its affinities towards SARS-CoV-2 S-RBDs of the original strain and the Omicron variant (Supplementary Fig. S1b) were individually determined via surface plasmon resonance (SPR) for quantitative comparison of the binding capacity difference. Expectedly, all the antibodies tested readily bound to original strain S-RBD, showing nano-molar affinities (Fig. 1b and Supplementary Fig. S2). Towards Omicron S-RBD, however, only EY6A targeting CR3022 site and S309 targeting S309 site retained comparable binding. The remaining antibodies, especially those targeting the RBS-A, -B, -C and -D sites, were either inert in S-RBD recognition or showed significantly reduced binding capacity (decreased >400 folds), demonstrating significant escape of neutralizing-antibody recognition for Omicron S-RBD. It is notable that of the antibodies tested, CB6 (Etesevimab), LY-CoV555 (Bamlanivimab), P2C-1F11 (Amubarvimab), REGN10933 (Casirivimab), REGN10987 (Imdevimab), and S309 (Sotrovimab) have been approved for clinical use. We also evaluated another pair of clinically used antibodies [AZD8895 (Tixagevimab) and AZD1061 (Cilgavimab)], which also showed severely impaired binding towards Omicron S-RBD (Fig. 1b and Supplementary Fig. S2). These results raised concerns over their therapeutic efficacies against the Omicron virus. Noted that the binding of S309 to S-RBD was only marginally affected by the Omicron-specific mutations, we believe this clinically approved antibody should remain effective in combating Omicron. Considering that the Omicron variant has a potential to cause another global SARS-CoV-2 pandemic, evaluation of the binding capacity between Omicron variant S-RBD and its receptor ACE2 has become another focus. Thus, we subsequently side-by-side determined the binding affinities of original strain S-RBD and Omicron variant S-RBD to human ACE2 (Supplementary Fig. S1c). With SPR, the kinetic-affinity values were determined to be 87.9 nM for original strain S-RBD and 40.8 nM for Omicron variant S-RBD, respectively (Fig. 1c, d). These values highlight ~2.2-fold-enhanced receptor-binding with Omicron S-RBD. In light of the previous report showing that soluble ectodomain protein of ACE2 could function as a “neutralizing decoy” to block SARS-CoV-2 entry, 3 we thus further measured the binding capability between Omicron S-RBD and our affinity-enhanced ACE2 decoy proteins (ACE2[W19/Y330] and ACE2[W27/Y330]). As expected, towards the original strain and Omicron variant S-RBDs, ACE2[W19/Y330] and ACE2[W27/Y330] both showed apparently higher binding capacities than wild-type ACE2 (ACE2/WT) (Fig. 1e–h). The calculated affinities revealed ~10.2-fold increase for ACE2[W19/Y330] and ~6.5-fold increase for ACE2[W27/Y330] towards Omicron S-RBD, respectively. These observations highlighted that the improved binding between SARS-CoV-2 S-RBD and the affinity-enhanced decoy ACE2s were not affected by the Omicron mutations, forming stark contrast to the tested antibodies, which showed high degree of immune evasion. Since the safety of recombinant ACE2 have been verified in humans, 4 the affinity-enhanced decoy ACE2s might be developed as weapons to fight against the Omicron variant and/or other emerging SARS-CoV-2 variants that use human ACE2 as the functional receptor. As SARS-CoV-2 S-RBD is the most important component in vaccine preparation, we thus further evaluated the thermal stability and protease-digestion sensitivity of the Omicron variant S-RBD protein. The melting temperatures (Tms) of both SARS-CoV-2 original strain and Omicron variant S-RBDs were determined in parallel using differential scanning fluorimetry (DSF). Unexpectedly, the calculated Tm value for the Omicron variant S-RBD protein was significantly lower than that of the original strain S-RBD (decreased by ~8.3 °C) (Fig. 1i), demonstrating impaired protein thermal stability due to the mutations in Omicron S-RBD. We also performed the incomplete-digestion assay using trypsin and chymotrypsin to investigate the susceptibility of both S-RBDs to protease digestion. In comparison to S-RBD of the original strain, its Omicron counterpart was indeed much more easily digested by both proteases (Fig. 1j, k). Such observation was well consistent with the introduction of several basic amino acids (N440K, T478K, Q493R, Q498R, and Y505H for trypsin digestion) and aromatic/hydrophobic residues (S371L, S375F, and N501Y for chymotrypsin digestion) in the Omicron S-RBD primary amino-acid sequence. The results also suggested that more attention should be paid to Omicron S-RBD to prevent its potential denaturation and/or degradation when developing RBD-based Omicron vaccines. In addition, it has been shown that the SARS-CoV-2 virus could infect individuals through the digestive tracts, and further cause symptoms in the situs. 5 Since both trypsin and chymotrypsin are a class of digestion-related proteases, they are very common and abundant in human digestive tracts. The decreased thermal stability and increased digestion susceptibility observed with Omicron S-RBD seem to coincide with the less virulence of the Omicron virus, at least along the digestive tract. In conclusion, our study revealed that the multiple mutations on Omicron S-RBD have significantly compromised antibody recognition. Nevertheless, the Omicron-specific mutations did not affect S-RBD binding by affinity-enhanced decoy ACE2 proteins, highlighting future development of decoy ACE2 and its affinity-enhanced proteins for potential treatment of the Omicron infections. Supplementary information Supplementary Materials

          Related collections

          Most cited references4

          • Record: found
          • Abstract: found
          • Article: not found

          Evidence for Gastrointestinal Infection of SARS-CoV-2

          Since the novel coronavirus (SARS-CoV-2) was identified in Wuhan, China, at the end of 2019, the virus has spread to 32 countries, infecting more than 80,000 people and causing more than 2600 deaths globally. The viral infection causes a series of respiratory illnesses, including severe respiratory syndrome, indicating that the virus most likely infects respiratory epithelial cells and spreads mainly via respiratory tract from human to human. However, viral target cells and organs have not been fully determined, impeding our understanding of the pathogenesis of the viral infection and viral transmission routes. According to a recent case report, SARS-CoV-2 RNA was detected in a stool specimen, 1 raising the question of viral gastrointestinal infection and a fecal-oral transmission route. It has been proven that SARS-CoV-2 uses angiotensin-converting enzyme (ACE) 2 as a viral receptor for entry process. 2 ACE2 messenger RNA is highly expressed and stabilized by B0AT1 in gastrointestinal system, 3 , 4 providing a prerequisite for SARS-CoV-2 infection. To further investigate the clinical significance of SARS-CoV-2 RNA in feces, we examined the viral RNA in feces from 71 patients with SARS-CoV-2 infection during their hospitalizations. The viral RNA and viral nucleocapsid protein were examined in gastrointestinal tissues from 1 of the patients. Methods From February 1 to 14, 2020, clinical specimens, including serum, nasopharyngeal, and oropharyngeal swabs; urine; stool; and tissues from 73 hospitalized patients infected with SARS-CoV-2 were obtained in accordance with China Disease Control and Prevention guidelines and tested for SARS-CoV-2 RNA by using the China Disease Control and Prevention–standardized quantitative polymerase chain reaction assay. 5 Clinical characteristics of the 73 patients are shown in Supplementary Table 1. The esophageal, gastric, duodenal, and rectal tissues were obtained from 1 of the patients by using endoscopy. The patient’s clinical information is described in the Supplementary Case Clinical Information and Supplementary Table 2. Histologic staining (H&E) as well as viral receptor ACE2 and viral nucleocapsid staining were performed as described in the Supplementary Methods. The images of fluorescent staining were obtained by using laser scanning confocal microscopy (LSM880, Carl Zeiss MicroImaging, Oberkochen, Germany) and are shown in Figure 1 . This study was approved by the Ethics Committee of The Fifth Affiliated Hospital, Sun Yat-sen University, and all patients signed informed consent forms. Figure 1 Images of histologic and immunofluorescent staining of gastrointestinal tissues. Shown are images of histologic and immunofluorescent staining of esophagus, stomach, duodenum, and rectum. The scale bar in the histologic image represents 100 μm. The scale bar in the immunofluorescent image represents 20 μm. Results From February 1 to 14, 2020, among all of the 73 hospitalized patients infected with SARS-CoV-2, 39 (53.42%), including 25 male and 14 female patients, tested positive for SARS-CoV-2 RNA in stool, as shown in Supplementary Table 1. The age of patients with positive results for SARS-CoV-2 RNA in stool ranged from 10 months to 78 years old. The duration time of positive stool results ranged from 1 to 12 days. Furthermore, 17 (23.29%) patients continued to have positive results in stool after showing negative results in respiratory samples. Gastrointestinal endoscopy was performed on a patient as described in the Supplementary Case Clinical Information. As shown in Figure 1, the mucous epithelium of esophagus, stomach, duodenum, and rectum showed no significant damage with H&E staining. Infiltrate of occasional lymphocytes was observed in esophageal squamous epithelium. In lamina propria of the stomach, duodenum, and rectum, numerous infiltrating plasma cells and lymphocytes with interstitial edema were seen. Importantly, viral host receptor ACE2 stained positive mainly in the cytoplasm of gastrointestinal epithelial cells (Figure 1). We observed that ACE2 is rarely expressed in esophageal epithelium but is abundantly distributed in the cilia of the glandular epithelia. Staining of viral nucleocapsid protein was visualized in the cytoplasm of gastric, duodenal, and rectum glandular epithelial cell, but not in esophageal epithelium. The positive staining of ACE2 and SARS-CoV-2 was also observed in gastrointestinal epithelium from other patients who tested positive for SARS-CoV-2 RNA in feces (data not shown). Discussion In this article, we provide evidence for gastrointestinal infection of SARS-CoV-2 and its possible fecal-oral transmission route. Because viruses spread from infected to uninfected cells, 6 viral-specific target cells or organs are determinants of viral transmission routes. Receptor-mediated viral entry into a host cell is the first step of viral infection. Our immunofluorescent data showed that ACE2 protein, which has been proven to be a cell receptor for SARS-CoV-2, is abundantly expressed in the glandular cells of gastric, duodenal, and rectal epithelia, supporting the entry of SARS-CoV-2 into the host cells. ACE2 staining is rarely seen in esophageal mucosa, probably because the esophageal epithelium is mainly composed of squamous epithelial cells, which express less ACE2 than glandular epithelial cells. Our results of SARS-CoV-2 RNA detection and intracellular staining of viral nucleocapsid protein in gastric, duodenal, and rectal epithelia demonstrate that SARS-CoV-2 infects these gastrointestinal glandular epithelial cells. Although viral RNA was also detected in esophageal mucous tissue, absence of viral nucleocapsid protein staining in esophageal mucosa indicates low viral infection in esophageal mucosa. After viral entry, virus-specific RNA and proteins are synthesized in the cytoplasm to assemble new virions, 7 which can be released to the gastrointestinal tract. The continuous positive detection of viral RNA from feces suggests that the infectious virions are secreted from the virus-infected gastrointestinal cells. Recently, we and others have isolated infectious SARS-CoV-2 from stool (unpublished data), confirming the release of the infectious virions to the gastrointestinal tract. Therefore, fecal-oral transmission could be an additional route for viral spread. Prevention of fecal-oral transmission should be taken into consideration to control the spread of the virus. Our results highlight the clinical significance of testing viral RNA in feces by real-time reverse transcriptase polymerase chain reaction (rRT-PCR) because infectious virions released from the gastrointestinal tract can be monitored by the test. According to the current Centers for Disease Control and Prevention guidance for the disposition of patients with SARS-CoV-2, the decision to discontinue transmission-based precautions for hospitalized patients with SARS-CoV-2 is based on negative results rRT-PCR testing for SARS-CoV-2 from at least 2 sequential respiratory tract specimens collected ≥24 hours apart. 8 However, in more than 20% of patients with SARS-CoV-2, we observed that the test result for viral RNA remained positive in feces, even after test results for viral RNA in the respiratory tract converted to negative, indicating that the viral gastrointestinal infection and potential fecal-oral transmission can last even after viral clearance in the respiratory tract. Therefore, we strongly recommend that rRT-PCR testing for SARS-CoV-2 from feces should be performed routinely in patients with SARS-CoV-2 and that transmission-based precautions for hospitalized patients with SARS-CoV-2 should continue if feces test results are positive by rRT-PCR testing.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome

            Background Renin-angiotensin system (RAS) signaling and angiotensin-converting enzyme 2 (ACE2) have been implicated in the pathogenesis of acute respiratory distress syndrome (ARDS). We postulated that repleting ACE2 using GSK2586881, a recombinant form of human angiotensin-converting enzyme 2 (rhACE2), could attenuate acute lung injury. Methods We conducted a two-part phase II trial comprising an open-label intrapatient dose escalation and a randomized, double-blind, placebo-controlled phase in ten intensive care units in North America. Patients were between the ages of 18 and 80 years, had an American-European Consensus Criteria consensus diagnosis of ARDS, and had been mechanically ventilated for less than 72 h. In part A, open-label GSK2586881 was administered at doses from 0.1 mg/kg to 0.8 mg/kg to assess safety, pharmacokinetics, and pharmacodynamics. Following review of data from part A, a randomized, double-blind, placebo-controlled investigation of twice-daily doses of GSK2586881 (0.4 mg/kg) for 3 days was conducted (part B). Biomarkers, physiological assessments, and clinical endpoints were collected over the dosing period and during follow-up. Results Dose escalation in part A was well-tolerated without clinically significant hemodynamic changes. Part B was terminated after 39 of the planned 60 patients following a planned futility analysis. Angiotensin II levels decreased rapidly following infusion of GSK2586881, whereas angiotensin-(1–7) and angiotensin-(1–5) levels increased and remained elevated for 48 h. Surfactant protein D concentrations were increased, whereas there was a trend for a decrease in interleukin-6 concentrations in rhACE2-treated subjects compared with placebo. No significant differences were noted in ratio of partial pressure of arterial oxygen to fraction of inspired oxygen, oxygenation index, or Sequential Organ Failure Assessment score. Conclusions GSK2586881 was well-tolerated in patients with ARDS, and the rapid modulation of RAS peptides suggests target engagement, although the study was not powered to detect changes in acute physiology or clinical outcomes. Trial registration ClinicalTrials.gov, NCT01597635. Registered on 26 January 2012. Electronic supplementary material The online version of this article (doi:10.1186/s13054-017-1823-x) contains supplementary material, which is available to authorized users.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Structural and functional ramifications of antigenic drift in recent SARS-CoV-2 variants

              Neutralizing antibodies (nAbs) elicited against the receptor-binding site (RBS) of the spike protein of wild-type SARS-CoV-2 are generally less effective against recent variants of concern. RBS residues E484, K417 and N501 are mutated in variants first described in South Africa (B.1.351) and Brazil (P.1). We analyzed their effects on ACE2 binding and K417N and E484K mutations on nAbs isolated from COVID-19 patients. Binding and neutralization of the two most frequently elicited antibody families (IGHV3-53/3-66 and IGHV1-2), which can both bind the RBS in alternate binding modes, are abrogated by K417N, E484K, or both. These effects can be structurally explained by their extensive interactions with RBS nAbs. However, nAbs to the more conserved, cross-neutralizing CR3022 and S309 sites were largely unaffected. The results have implications for next-generation vaccines and antibody therapies.
                Bookmark

                Author and article information

                Contributors
                lugw@scu.edu.cn
                Journal
                Signal Transduct Target Ther
                Signal Transduct Target Ther
                Signal Transduction and Targeted Therapy
                Nature Publishing Group UK (London )
                2095-9907
                2059-3635
                21 February 2022
                21 February 2022
                2022
                : 7
                : 56
                Affiliations
                [1 ]GRID grid.13291.38, ISNI 0000 0001 0807 1581, West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, , Sichuan University, ; 610041 Chengdu, Sichuan China
                [2 ]GRID grid.13291.38, ISNI 0000 0001 0807 1581, Disaster Medicine Center, West China Hospital, , Sichuan University, ; 610041 Chengdu, Sichuan China
                Author information
                http://orcid.org/0000-0001-7568-592X
                Article
                914
                10.1038/s41392-022-00914-2
                8860268
                35190526
                9bdc2552-f807-4ea2-81a0-56f54a93e7a7
                © The Author(s) 2022

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 20 January 2022
                : 27 January 2022
                Funding
                Funded by: This work was supported by the National Key Research and Development Program of China (Grant no. 2021YFC2301400), the National Natural Science Foundation of China (Grant no. 82041042), the special research fund on COVID-19 of West China Hospital, Sichuan University (Grant no. HX-2019-nCoV-004), and the 1.3.5 project for disciplines of excellence, West China Hospital, Sichuan University (Grant no. ZYYC20008).
                Categories
                Letter
                Custom metadata
                © The Author(s) 2022

                vaccines,infectious diseases
                vaccines, infectious diseases

                Comments

                Comment on this article