31
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Bimodular effects of D614G mutation on the spike glycoprotein of SARS-CoV-2 enhance protein processing, membrane fusion, and viral infectivity

      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, By the end of June 2020, the pandemic of coronavirus diseases 2019 (COVID-19) had resulted in more than 10 million individuals, all over the world, being infected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). 1 The high contagiousness of SARS-CoV-2 virus was largely attributed to the relatively unique sequence composition in its spike (S) glycoprotein, which is in charge of the host entry by interacting with its cellular receptor angiotensin-converting enzyme 2 (ACE2). 2,3 The S glycoprotein can be processed into an N terminal S1 fragment that is responsible for receptor binding, and a C terminal S2 fragment that functions to promote membrane fusion. 4 SARS-CoV-2 is a type of single-stranded positive-sense ribonucleic acid (RNA) virus, and an increasing number of mutations were identified across the SARS-CoV-2 genome, including the region encoding S glycoprotein. Nevertheless, the functional implications of these mutations remain largely unknown, we here reported the mutation analysis of S gene and a functional exploration of the dominant D614G mutation. In total, 9002 S gene sequences were extracted from the high-quality complete genomes deposited in the GISAID EpiCoV database as of April 28, 2020. Sequence alignment identified 6253 non-synonymous mutations at 82 different sites of the S gene (Supplementary Table S1 and Fig. 1a). While the majority of the mutations (for 81 sites) were in low frequency (<0.6%), the D614G stood out as a prominent mutation, accounting for about 90% (5583/6253) mutations identified (Supplementary Table S1), with a site-specific mutation frequency of >62% (5583/9002) (Fig. 1a), suggesting potential impacts on the function of S protein. Fig. 1 Enhanced infectivity of D614G mutant of SARS-CoV-2 virus. a The mutation profile of SARS-CoV-2 spike glycoprotein. From a set of 9002 SARS-CoV-2 genome sequences, mutations were identified at 82 positions across the gene encoding S glycoprotein with the D614G being the dominant mutation (>62%); for the plot of mutation counts (the left Y axis), only those showing more than 5 counts were displayed. b Three-dimensional structure modeling of the D614 (upper) and G614 (lower) S glycoprotein colored by chain. The α subunit in red is on the left, the neighboring β subunit in green is on the right. The D614, G614, and T859 residues are displayed in the style of scaled ball and stick with simulated surfaces in yellow. c Detection of the expression of the indicated proteins by Western blot. The spike protein was blotted by an antibody against the S2 region. d, e Altered expression of spike trimer in G614 mutant. d Expression of two spike proteins in the absence and presence of ACE2 in 293T cells at the time points of 12 and 48-h post transfection by Western blot. e Quantification of the relative expression of spike trimer and monomer with the respective D614 expression in the absence of ACE2 as the reference. All results were normalized by the expression of β-actin. Data are mean ± SD of triple quantification. f, g Quantification of syncytia formation upon expression of the indicated S glycoprotein in 293T-ACE2 cells (f) and Hela-ACE2 cells (g). Data are the mean ± SD of results from 4–5 fields (20x objective lens). ***p < 0.001; ****p < 0.0001. More than three replicates were done for the experiment. h, i The expression of the luciferase reporter in 293T-ACE2 cells (h) and Hela-ACE2 cells (i) upon infection of viruses pseudotyped with D614 or G614 S glycoproteins as indicated. Data are the mean ± SD of the results of quadruplicate. Fold changes between D614 and G614 genotypes are displayed. More than three replicates were done for the experiment To explore the potential effects of the D614G mutation on the spatial structure of S glycoprotein, we performed three-dimensional (3D) modeling with the template of published SARS-CoV-2 S structure (6vxx.pdb) by the Modeling algorithm at SWISS-MODEL. The native S protein trimer was comprised of 3 identical subunits, the Aspartic acid residue at position 614 (D614) of wild type S protein located at a region interfacing with the neighboring subunit (Fig. 1b, left α subunit in red), and was spatially close to and potentially interacted with the T859 residue of neighboring subunit by an inter-subunit hydrogen bond (Fig. 1b, left β subunit in green). This potential inter-subunit interaction mediated by D614-T859 was disrupted by the D to G mutation due to the loss of the side chain, which eliminated the inter-subunit hydrogen bond and may increase the flexibility of the main chain, as indicated by the surface simulation of T859, V860, L861 residues of the neighboring subunit (Fig. 1b, left β subunit in yellow). Meanwhile, the backbone amine of the glycine (G) residue shortened the hydrogen bond with the carboxyl group of neighboring amino acids (A647) within the same subunit and strengthened it, which might locally stabilize the S protein. Thus, the D614G mutation may conceivably affect the stability of the S protein trimer and its related functions, such as host entry and viral infectivity. In order to examine the potential functional outcomes of the D614G mutation, we made two constructs expressing the wild type (D614) and mutant (G614) SARS-CoV-2 S glycoprotein, respectively. Transfection of the same amount of these two constructs into 293T cells expressing ACE2 (293T-ACE2) effectively expressed full length (FL) S protein (Fig. 1c) at both 24 and 48-h post-transfection as detected by an antibody against the S2 region. Whereas, increased S2 production, which is resulted from protease-mediated cleavage of S protein, was observed in the G614 mutant as compared with that in the wild type D614 transfectant, the difference was even more obvious for transfectants of 48-h post-transfection (Fig. 1c), suggesting that the D614G mutation enhanced S protein processing, which is consistent with the idea that the D614G mutation may affect the stability of the S protein trimer as suggested above based on structural analysis (Fig. 1b). Interestingly, the expression of the super-shifted band above the full length S protein, representing for the trimer, was higher in G614 than D614 in the absence of ACE2 at both 12 and 48-h post-transfection, but was lower in G614 than D614 in the presence of ACE2 at 12-h post-transfection, and the difference disappeared at 48-h post-transfection, probably due to the saturated effects from ACE2 which could clearly promote the accumulation of overexpressed S protein (Fig. 1d, e). The results fit well with a bimodular effect of D614G mutation on S-trimer stability depending on ACE2 engagement, i.e., G614 mutation increased S-trimer stability on virion in the absence of ACE2, but promoted S-trimer dissociation, upon engaged with ACE2, to expose the S2 fragment (Fig. 1c), which conceivably contributed to high contagiousness. Since SARS-CoV-2 infection could induce membrane fusion (Supplementary Fig. S1) via S expression, the increased S2 production may predict the enhanced ability of the D614G mutant in inducing membrane fusion. To test this hypothesis, we took the advantages of syncytium formation assay as a surrogate of S protein-induced membrane fusion. As shown in Supplementary Fig. S2, a number of syncytia were formed in 293T-ACE2 cells 24-h post-transfection of wild type D614 S construct, while the G614 mutant exhibited significantly stronger induction of syncytium formation as indicated by higher fusion index calculated as the percentage of nuclei in syncytia (Fig. 1f), the difference maintained at 48-h post-transfection, when extensive syncytium formation had occurred (Supplementary Fig. S2 and 1f). the effect of D614G on syncytium formation was also confirmed in Hela cells engineered to stably express ACE2 cells (Fig. 1g). These results indicate that the D614G mutation may endow enhanced fusion ability to the SARS-CoV-2 S glycoprotein. To examine whether the enhanced fusion ability could be translated into enhanced virus infection, we made viruses pseudotyped with wild type and G614 S proteins, and infected 293T-ACE2 cells and Hela-ACE2 cells with the same amount of virus particles. As shown in Fig. 1h, i, the viruses pseudotyped with G614 mutant displayed luciferase activities about 3.1 ~ 4.2 times higher than those pseudotyped with wild type (D614) S protein by the time of 24-h post infection; and the fold changes increased to 5.2 ~ 5.6 by the time of 48-h post infection. Moreover, the luciferase activities positively correlated with membrane fusion indexes (Fig. 1f–i). Thus, these data are consistent with the notion that the D614G mutation enhanced SARS-CoV-2 infectivity via membrane fusion-mediated host entry. Interestingly, the Hela-ACE2 cells, but not 293T-ACE2 cells, expressed considerable amounts of TMPRSS2, which was also involved in the phenotypes mediated by S-G614 as the treatment of Camostat, a TMPRSS2 inhibitor, compromised S-G614-induced phenotypes (Supplementary Fig. S3). Consistent with our finding, a recent study by Kober et al. 5 reported that the SARS-CoV-2 variant containing the D614G mutation, initially beginning spreading in Europe, rapidly became a dominant form in regions where it was introduced. And the recurrent pattern of the increase occurred at multiple geographic levels, indicating high efficacy of transmission. Together with our findings, it is proposed that the SARS-CoV-2 variant of G614 genotype is highly transmissible, which is probably attributed to an increased S2 production associated with bi-modularly altered S-trimer stability, leading to enhanced membrane fusion and host entry. Therefore, particular attention should be paid to the emergence of this variant in a region to help prevent recurrent COVID-19 outbreak. Supplementary information Supplemenary files

          Related collections

          Most cited references3

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

          SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor

          Summary The recent emergence of the novel, pathogenic SARS-coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread pose a global health emergency. Cell entry of coronaviruses depends on binding of the viral spike (S) proteins to cellular receptors and on S protein priming by host cell proteases. Unravelling which cellular factors are used by SARS-CoV-2 for entry might provide insights into viral transmission and reveal therapeutic targets. Here, we demonstrate that SARS-CoV-2 uses the SARS-CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming. A TMPRSS2 inhibitor approved for clinical use blocked entry and might constitute a treatment option. Finally, we show that the sera from convalescent SARS patients cross-neutralized SARS-2-S-driven entry. Our results reveal important commonalities between SARS-CoV-2 and SARS-CoV infection and identify a potential target for antiviral intervention.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Tracking changes in SARS-CoV-2 Spike: evidence that D614G increases infectivity of the COVID-19 virus

            Summary A SARS-CoV-2 variant carrying the Spike protein amino acid change D614G has become the most prevalent form in the global pandemic. Dynamic tracking of variant frequencies revealed a recurrent pattern of G614 increase at multiple geographic levels: national, regional and municipal. The shift occurred even in local epidemics where the original D614 form was well established prior to the introduction of the G614 variant. The consistency of this pattern was highly statistically significant, suggesting that the G614 variant may have a fitness advantage. We found that the G614 variant grows to higher titer as pseudotyped virions. In infected individuals G614 is associated with lower RT-PCR cycle thresholds, suggestive of higher upper respiratory tract viral loads, although not with increased disease severity. These findings illuminate changes important for a mechanistic understanding of the virus, and support continuing surveillance of Spike mutations to aid in the development of immunological interventions.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found

              Structure, Function, and Evolution of Coronavirus Spike Proteins

              Fang Li (2016)
              The coronavirus spike protein is a multifunctional molecular machine that mediates coronavirus entry into host cells. It first binds to a receptor on the host cell surface through its S1 subunit and then fuses viral and host membranes through its S2 subunit. Two domains in S1 from different coronaviruses recognize a variety of host receptors, leading to viral attachment. The spike protein exists in two structurally distinct conformations, prefusion and postfusion. The transition from prefusion to postfusion conformation of the spike protein must be triggered, leading to membrane fusion. This article reviews current knowledge about the structures and functions of coronavirus spike proteins, illustrating how the two S1 domains recognize different receptors and how the spike proteins are regulated to undergo conformational transitions. I further discuss the evolution of these two critical functions of coronavirus spike proteins, receptor recognition and membrane fusion, in the context of the corresponding functions from other viruses and host cells.
                Bookmark

                Author and article information

                Contributors
                hhongy1999@126.com
                sunq@bmi.ac.cn
                Journal
                Signal Transduct Target Ther
                Signal Transduct Target Ther
                Signal Transduction and Targeted Therapy
                Nature Publishing Group UK (London )
                2095-9907
                2059-3635
                17 November 2020
                17 November 2020
                2020
                : 5
                : 268
                Affiliations
                [1 ]GRID grid.43555.32, ISNI 0000 0000 8841 6246, Beijing Institute of Biotechnology, ; 20 Dongda Street, Beijing, 100071 China
                [2 ]GRID grid.414367.3, Department of Oncology, , Beijing Shijitan Hospital of Capital Medical University, ; 10 TIEYI Road, Beijing, 100038 China
                [3 ]GRID grid.73113.37, ISNI 0000 0004 0369 1660, Department of Microbiology, , Second Military Medical University, ; Shanghai, 200433 China
                Author information
                http://orcid.org/0000-0002-5342-0637
                Article
                392
                10.1038/s41392-020-00392-4
                7670837
                33203835
                928a9929-aef7-46de-8e54-e482fd3b40bb
                © The Author(s) 2020

                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
                : 8 July 2020
                : 12 October 2020
                : 15 October 2020
                Funding
                Funded by: the National Key Research & Development Program of China (2019YFA09003801 to QS, 2018YFA0900804 to YZ)
                Categories
                Letter
                Custom metadata
                © The Author(s) 2020

                infectious diseases,microbiology
                infectious diseases, microbiology

                Comments

                Comment on this article