Sisi Shan’s research while affiliated with Tsinghua University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (27)


Binding, neutralizing, germline and phylogenetic properties of isolated nanobodies against SARS-CoV-1 and SARS-CoV-2 BA.4/5 variant
a. Binding activity is indicated by the percent (%) of spike positive cells analyzed by flow cytometry with red being the highest, followed by orange, and green. Neutralizing activity is shown by IC50 (μg/mL) and colored in red, orange, yellow, green, and gray, with red being the strongest and gray failed to reach IC50 at the highest concentration tested (10μg/ml). The five nanobodies highlighted in red in the column of mAb ID are the ones selected for downstream evaluation. All results were calculated from at least two independent experiments. The germline variable gene segment (V), diversity gene segment (D), and junction gene (J) as well as CDR3 sequences of each nanobody are colored for clarity. b. Phylogenetic analysis of nanobodies isolated in this study indicated by their names at the tip of the branches, compared with those isolated previously by our group (blue line) [28] and randomly selected from CoV-AbDab database (https://opig.stats.ox.ac.uk/webapps/covabdab/) (black line).
Neutralization breadth and potency of top 5 nanobodies against 25 diverse sarbecoviruses
a. Nanobody concentrations (μg/ml) required to achieve 50% (IC50) and 90% (IC90) reduction in viral infection. For clarity, neutralizing activity of each nanobody and control antibody is also colored with decreasing sequence from red, orange, yellow, green, to gray. Those in gray failed to reach IC50 at the highest concentration (10μg/ml) tested. A few representative escape mutations found in SARS-CoV-2 variants, and the GenBank accession numbers of SARS-CoV-1 and hACE2-dependent bat and pangolin coronaviruses are indicated under each strain tested. The complete set of mutations in each of the SARS-CoV-2 variant are indicated in the methods section. b. Actual neutralizing curve of nanobodies and control antibodies against the 25 diverse pseudoviruses, from which the IC50 and IC90 are estimated. The results shown are representatives of two independent experiments and presented as mean ± SEM.
Inhibition of diverse spike-mediated cell-cell fusion by isolated nanobodies
a. Cartoon illustration of a dually-split protein (DSP) assay to quantitatively measure cell-cell fusion by luciferase and GFP activity between two reporter cell lines: 293T cells expressing spike and nRL-nGFP (N-Renilla Luciferase-GFP1-7) and Huh-7 cells expressing the endogenous ACE2 and cRL-cGFP (C-N-Renilla Luciferase-GFP8-11). The cartoon was created with BioRender.com. b. Comparison of fusion activity of diverse spike of SARS-CoV-2 variants. c. Correlation between luciferase- and GFP-measured fusion activity. d. The potency and breadth of the top 5 isolated nanobodies and control antibodies in inhibiting cell-cell fusion mediated by a panel of 20 spike variants. Numbers indicate the nanobody and antibody concentrations (μg/ml) required to achieve 50% (IC50) reduction in cell-cell fusion. For clarity, inhibitory activity of each nanobody and control antibody is colored with decreasing sequence from red, orange, yellow, green, to gray. Those in gray failed to reach IC50 at the highest concentration tested (100μg/ml). A few representative mutations that potentially facilitate viral escape from antibodies are indicated below each of variants tested. The complete set of mutations in each of the variant are indicated in the methods section. e. Inhibitory activity of the best nanobody Tnb04 on cell-cell fusion shown by fluorescence imaging, captured and measured by the Opera Phenix Plus High-Content Screening System (PerkinElmer). Fluorescence intensity against each variant was first normalized against internal control mCherry before comparison was performed. Three concentrations of nanobody Tnb04 (100μg/ml, 1μg/ml, 0.05μg/ml) were tested against each and every variant listed.
Broadly neutralizing activity of Tnb04-1 against authentic SARS-CoV-1 and SARS-CoV-2 variants
a. Actual neutralization curve of Tnb04-1 against diverse clinical isolates of SARS-CoV-1, SARS-CoV-2 WT, Delta, and various Omicron subvariants BA.1, BA.2, BA.5.2, BQ.1.1, XBB, XBB.1.5, and EG.5.1, using focus reduction neutralization test (FRNT). b. The estimated IC50 and IC90 based on neutralization curves in the left panel. The results shown are representatives of two independent experiments and presented as mean ± SEM.
Structure basis and mechanism of action for the broadly neutralization by Tnb04-1
a. Overall crystal structure of the Tnb04-1 (green), SARS-CoV-2 RBD (cyan), and 1F11 fab (yellow and pink) complex. b. The epitope and paratope of Tnb04-1, highlighted in light gray in the context of RBD (cyan) and Tnb04-1 (green) and the involved residues are indicated. c. Molecular interaction between Tnb04-1 and SARS-CoV-2 RBD. The hydrogen bond interaction is shown on the left and the hydrophobic interaction on the right. The residues involved are indicated. d. The binding model of Tnb04-1 (green), Tnb03 (3-2A2-4) (gray), S309 (dark salmon), LY-CoV1404 (yellow), and ACE2 (purple) to SARS-CoV-2 RBD (cyan). e. The footprints of Tnb04-1 (green) on RBD, relative to that of ACE2 (purple) and control antibodies. Residue mutations found in BA.2.86 variant are highlighted in red. f Superimpose of variable and conserved residues on RBD structure derived from over 16 million sequences in the GISAID database collected from December 2019 to January 2024. Conserved residues are in white and variable residues in purple, and extent of which are in indicated by color gradient. Tnb04-1 gained two highly conserved epitope residues E340 and P337 (green) while excluded two relatively variable residues S371 and F374 (purple), compared to control Tnb03 (3-2A2-4). g. The epitope comparison between Tnb04-1, Tnb03 (3-2A2-4) and the five classes antibodies reported by Xiang et al [47] h. The simulated model of three Tnb04-1 bind to partially open SARS-CoV-2 S trimer (one RBD is open). Tnb04-1 is in green, RBD in cyan, NTD in dark salmon, and rest of the spike in dark gray. i. Detection of proteinase K-resistant core on a western blot probed with a rabbit anti-S2 polyclonal antibody. Various experimented conditions are indicated above. j. The integrated density of proteinase K-resistant core under each experimental condition detected and calculated by ImageJ.

+2

Super broad and protective nanobodies against Sarbecoviruses including SARS-CoV-1 and the divergent SARS-CoV-2 subvariant KP.3.1.1
  • Article
  • Full-text available

November 2024

·

34 Reads

Haodi Dong

·

Runhong Zhou

·

Jing Chen

·

[...]

·

The ongoing evolution and immune escape of SARS-CoV-2, alongside the potential threat of SARS-CoV-1 and other sarbecoviruses, underscore the urgent need for effective strategies against their infection and transmission. This study highlights the discovery of nanobodies from immunized alpacas, which demonstrate exceptionally broad and potent neutralizing capabilities against the recently emerged and more divergent SARS-CoV-2 Omicron subvariants including JD.1.1, JN.1, KP.3, KP.3.1.1, as well as SARS-CoV-1 and coronaviruses from bats and pangolins utilizing receptor ACE2. Among these, Tnb04-1 emerges as the most broad and potent, binding to a conserved hydrophobic pocket in the spike’s receptor-binding domain, distinct from the ACE2 binding site. This interaction disrupts the formation of a proteinase K-resistant core, crucial for viral-cell fusion. Notably, intranasal administration of Tnb04-1 in Syrian hamsters effectively prevented respiratory infection and transmission of the authentic Omicron XBB.1.5 subvariant. Thus, Thb04-1 holds promise in combating respiratory acquisition and transmission of diverse sarbecoviruses.

Download

Better Prior Distribution for Antibody Complementarity-Determining Regions Design

November 2024

Designing antibodies with desired binding specificity and affinity is vital for various fields, including pharmaceutical and biological research. While recent advances leverage diffusion-based models for co-designing the sequences and structures of the Complementarity-Determining Regions (CDRs), they are still confronted with three challenges: non-informative prior distribution, incompatibility with discrete amino acid types, and impractical computational cost in large-scale sampling. To address these pitfalls, we proposed FlowDesign, a novel approach for sequence-structure co-design based on Flow Matching with the following merits: (1) Flexible selection of prior distributions; (2) Direct matching of discrete distributions; (3) Enhanced computational efficiency for large-scale sampling. By leveraging various prior distributions, we first identified data-driven structural models as the most informative prior. Subsequently, evaluations against strong baselines illustrated the superiority of our model across diverse metrics including Amino Acid Recovery (AAR), RMSD, and Rosetta energy. We also demonstrated the utility of FlowDesign in designing antibodies to target HIV-1 cellular receptor CD4. Through Biolayer interferometry (BLI) and pseudovirus neutralization evaluation, we obtained regenerated antibodies with better binding affinity and neutralizing potency compared with the state-of-the-art HIV antibody Ibalizumab across multiple HIV mutants at the cellular level. These results highlight the power of our FlowDesign model in antibody design and its potential application to engineering other protein molecules.


Figure 5. B8-dIgA1 and B8-dIgA2 enhance SARS-CoV-2 infection via CD209. (A) The effects of B8-dIgA2 on SARS-CoV-2 infection in the MucilAir TM model, consisting of primary human nasal epithelial cells but no DCs. B8-mIgA2 or B8-dIgA2 were pre-incubated at doses of 10, 100, and 1000 ng/ml, respectively, in the apical compartment with or without mucus for 1 hour, before adding 10 4 PFU of SARS-CoV-2 (BetaCoV/France/IDF00372/2020) for 4 hours. The viral RNA loads were measured by RT-PCR in both the apical and basal compartments and are shown in log-transformed units; (B) Representative confocal images (400×) of olfactory epithelium in NT showed the expression of CD209 (DC-SIGN) in green and ACE2 in magenta by immunohistochemical staining of experimental hamsters treated with B8-dIgA2 without (left) or with (middle and right) SARS-CoV-2 infection. Color-coding indicates specific antibodies used for double staining. Infected CD209 + cells are visualized in yellow as indicated by arrows (right); (C) The CD209 or CD299 overexpressed-HEK 293 T cells were pre-treated for 6 hours with 10 ng/ml of B8-dIgA1 or B8-dIgA2 or control dIgA1 or control dIgA2 or PBS, respectively, prior to SARS-CoV-2 infection (MOI: 0.05). Each treatment was performed in triplicate. Two days after infection, SARS-CoV-2 NP expression (green) was quantified by the mean fluorescence intensity (MFI) after anti-NP IF staining. Statistics were generated using student-t tests. *p < 0.05; **p < 0.01; ***p < 0.001L; (D) The effects of B8 antibodies on cell-cell fusion. 293 T cells co-transfected with SARS-CoV-2 spike and GFP were pre-treated with 100× the IC 90 dose of B8-IgG1, B8-mIgA1, B8-mIgA2, B8-dIgA1, B8-dIgA2 or and IgG isotypic control for 1 hour, respectively. Vero-E6 cells transfected with TMPRSS2 were then added to the treated 293T-spike-GFP cells and co-cultured for 48 hours. Cell-cell fusion was imaged under a fluorescence confocal microscope at the 50× magnification.
Figure 6. B8-dIgA1 promotes MoDC-mediated transmission of SARS-CoV-2 pseudovirus via CD209. Human MoDCs were treated with 25 ng/ml of different hIgAs for 1 hour. (A) MoDCs were then subjected to flow cytometry analysis. Representative plots showing hIgA-expressing live MoDCs (left) and the expression of CD209 on hIgA positive cells (right). (B) Quantified results depict the frequencies of hIgA positive MoDCs. (C) hIgA-treated MoDCs were subjected to confocal imaging with 20× objective. CD209: green, human IgAs (red) and nucleus (blue). Scale bar: 10 µm. (D) Schedule of cell-to-cell transmission assay. MoDCs were pretreated with different hIgAs (1) prior to SARS-CoV-2 pseudovirus infection (2). Infected MoDCs were co-cultured with Vero-E6-TMPRSS2 cells (3). (E) Luciferase activity of infected MoDCs with without hIgAs treatments. Luciferase activity of co-culture of MoDCs and Vero-E6 TMPRSS2 with hIgA1 (F) and hIgA2 (G) treatments. Each symbol represents an individual donor with a line indicating the change after different hIgAs treatments.
SARS-CoV-2 hijacks neutralizing dimeric IgA for nasal infection and injury in Syrian hamsters

August 2023

·

134 Reads

·

6 Citations

Prevention of robust severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in nasal turbinate (NT) requires in vivo evaluation of IgA neutralizing antibodies. Here, we report the efficacy of receptor binding domain (RBD)-specific monomeric B8-mIgA1 and B8-mIgA2, and dimeric B8-dIgA1, B8-dIgA2 and TH335-dIgA1 against intranasal SARS-CoV-2 challenge in Syrian hamsters. These antibodies exhibited comparable neutralization potency against authentic virus by competing with human angiotensin converting enzyme-2 (ACE2) receptor for RBD binding. While reducing viral loads in lungs significantly, prophylactic intranasal B8-dIgA led to high amount of infectious viruses and extended damage in NT than controls. Mechanistically, while B8-dIgA failed inhibition of SARS-CoV-2 cell-to-cell transmission, virus might hijack B8-dIgA through dendritic cell-mediated trans-infection of NT epithelia with robust nasal infection. Cryo-EM further revealed B8 as a class II antibody binding trimeric RBDs in 3-up or 2-up/1-down conformation. Neutralizing dIgA, therefore, may engage an unexpected mode for SARS-CoV-2 nasal infection and injury.


Structural basis of α1A-adrenergic receptor activation and recognition by an extracellular nanobody

June 2023

·

362 Reads

·

20 Citations

The α1A-adrenergic receptor (α1AAR) belongs to the family of G protein-coupled receptors that respond to adrenaline and noradrenaline. α1AAR is involved in smooth muscle contraction and cognitive function. Here, we present three cryo-electron microscopy structures of human α1AAR bound to the endogenous agonist noradrenaline, its selective agonist oxymetazoline, and the antagonist tamsulosin, with resolutions range from 2.9 Å to 3.5 Å. Our active and inactive α1AAR structures reveal the activation mechanism and distinct ligand binding modes for noradrenaline compared with other adrenergic receptor subtypes. In addition, we identified a nanobody that preferentially binds to the extracellular vestibule of α1AAR when bound to the selective agonist oxymetazoline. These results should facilitate the design of more selective therapeutic drugs targeting both orthosteric and allosteric sites in this receptor family.


Identification and characterization of SWT-specific antibodies from convalescent patients in the early pandemic
a, Initial screening for SWT-specific antibodies from nine convalescent patients using ELISA. Of 771 antibodies produced in the culture supernatant, 476 (61.7%) had strong binding with optical density (OD) values above the cut-off of 0.2, more than threefold higher than the background. b, Epitope specificity of the 476 antibodies identified in a. The patient number and the number of antibodies obtained from each patient are shown in the inner circle. Each slice is colored according to its epitope specificity and is proportional to the total antibodies obtained. c, Initial screening of the 476 antibodies as in a for their neutralizing activity against wild-type SARS-CoV-2 using pseudovirus-based neutralization assay. The cut-off value of neutralization was 10,000 ng ml⁻¹. d, Proportions of nAbs among those binding to RBD (40 of 132, 30%), S2 (1 of 214, 0.5%) and others (17 of 130, 13%). Results are representative of at least two independent experiments. IC50, half-maximum inhibitory concentration.
Source data
Neutralizing activity, epitope characterization and gene family analysis of the top 40 RBD-specific antibodies
Neutralization activity against a panel of 17 pseudoviruses carrying the S proteins of wild-type and various VOCs and VOIs, indicated by antibody concentration required to achieve 50% reduction in viral infectivity (IC50, ng ml⁻¹). The neutralizing activity is colored from red, orange, yellow, green to gray, with red being the strongest, while gray failed to reach IC50 at the highest concentration tested (10,000 ng ml⁻¹). A few representative mutations that potentially facilitate viral escape from antibodies are indicated below each of the VOCs and VOIs. The complete set of mutations for each of the VOCs and VOIs can be found in the Methods. Epitope specificity was determined by competition with typical class 1 (P2C-1F11 and REGN10933), class 2 (P2B-2F6) and class 3 (REGN10987 and S309) antibodies measured by surface plasmon resonance (SPR). All results were calculated from at least two independent experiments. The germline gene usage (IGHV, IGKV, IGLV), the length of complementarity determining region (CDR) 3 and the proportion of somatic hypermutation (SHM) were estimated using the IMGT program. Antibodies highlighted in red text are those competed with typical class 1 antibody P2C-1F11 and which preferentially used germline IGHV3-53/66. For clarity, the five antibodies that had their crystal or cryo-EM structures resolved in complex with RBD or S trimer are labeled with either red (class 1) or black (other classes) background. G1 to G5, group 1 to group 5; VOIs, variants of interest.
Preferred germline gene usage among the RBD-specific and the S-specific antibodies
a–d, Germline heavy and light gene usage and pairing among the top 40 RBD-specific (a,b) and the 476 S-specific antibodies (c,d), presented in chord diagrams (a,c) and heatmaps (b,d). In the chord diagrams, each of the paired germline heavy and light chains are linked by arcs, the sizes of which are proportional to the total antibodies identified. The number and the color in the heatmaps represent the number of antibodies identified with their germline heavy and light chains indicated along the vertical and horizontal axes. The preferred germline usages are highlighted by colored background in both chord diagrams and heatmaps.
Source data
P2-1B1, P5S-2B10, P5-1H1 and P2S-2E9 prophylaxis protects K18-hACE2 mice from infection with SARS-CoV-2 omicron BA.1 or beta
a–c, Survival percentage and body weight recorded daily post infection with BA.1 until death occurred or at the end point of experiments at 14 dpi in P2-1B1 (a), P5S-2B10 (b) and P5-1H1 (c) groups. d–f, Lung viral titers and brain viral titers in mice killed at 3 dpi for BA.1 in P2-1B1 (d), P5S-2B10 (e) and P5-1H1 (f) groups. g, Survival percentage and body weight recorded daily post infection with beta until death occurred or at the end point of experiments at 14 dpi in P2S-2E9 group. h, Lung viral titers and brain viral titers in mice killed at 4 dpi for beta in P2S-2E9 group. Weight change and PFU per organ are presented as mean ± s.e.m. The significance was estimated by a two-tailed, unpaired t-test. *P < 0.05; ***P < 0.001; NS, not significant; ND, not detected. i–l, H&E and immunohistochemistry staining of lung tissue from P2-1B1 (i), P5S-2B10 (j), P5-1H1 (k) or P2S-2E9 (l) intraperitoneally treated and corresponding untreated mice at day 3 (BA.1) or day 4 (beta) post infection. Dark brown, cells positive for SARS-CoV-2 N protein. Scale bars, 50 µm. Images were derived from one representative mouse in each group. The P2-1B1 experiment shared the same negative control mice as previously published⁵⁰. P5S-2B10 and P5-1H1 experiments shared the same negative control mice in this study. dpi, days post infection; H&E, hematoxylin and eosin.
Source data
Binding mode and epitope specificity of five bnAbs to SARS-CoV-2
a–e, Crystal or cryo-EM structures of five Fab fragments complexed with RBDs derived from wild-type, beta or omicron BA.1. All RBDs are colored in cyan whereas P5S-2B10 (a) is in red, P5-1H1 (b) is in green, P2S-2E9 (c) is in magenta, P5S-3B11 (d) is in orange and P2-1B1 (e) is in purple. f, Fab fragments of P5S-2B10, P5-1H1, P2S-2E9, P5S-3B11 and P2-1B1 complexed with RBDs superimposed into one composite together with receptor ACE2 (brown). g, The footprints of P5S-2B10, P5-1H1 and P2-1B1 Fabs, together with those of S2E12 and ACE2, shown on the surface of the SARS-CoV-2 RBD. The epitope residues are indicted just below each structure, with the mutation sites found in omicron BA.1 indicated in red. h, Comparison of the epitope residues of the P5S-2B10, P5-1H1, P2-1B1, P2S-2E9 and P5S-3B11 antibodies with published representative antibodies and the receptor ACE2 along the linear RBD sequence. A logo plot of RBD sequences was created based on all tested SARS-CoV-2 variants. The numbering system follows that in the GISAID database. i,j, The footprints of P2S-2E9 (i) and P5S-3B11 (j) Fabs, together with those of representative antibodies and ACE2, shown on the surface of the SARS-CoV-2 RBD. The epitope residues are indicted just below each structure, with the mutation sites found in omicron BA.1 indicated in red. The highly conserved N-glycosylation residue N343 among the sarbecoviruses is indicated.
Infection with wild-type SARS-CoV-2 elicits broadly neutralizing and protective antibodies against omicron subvariants

March 2023

·

242 Reads

·

18 Citations

Nature Immunology

The omicron variants of SARS-CoV-2 have substantial ability to escape infection- and vaccine-elicited antibody immunity. Here, we investigated the extent of such escape in nine convalescent patients infected with the wild-type SARS-CoV-2 during the first wave of the pandemic. Among the total of 476 monoclonal antibodies (mAbs) isolated from peripheral memory B cells, we identified seven mAbs with broad neutralizing activity to all variants tested, including various omicron subvariants. Biochemical and structural analysis indicated the majority of these mAbs bound to the receptor-binding domain, mimicked the receptor ACE2 and were able to accommodate or inadvertently improve recognition of omicron substitutions. Passive delivery of representative antibodies protected K18-hACE2 mice from infection with omicron and beta SARS-CoV-2. A deeper understanding of how the memory B cells that produce these antibodies could be selectively boosted or recalled can augment antibody immunity against SARS-CoV-2 variants.


Broadly neutralizing and protective nanobodies against SARS-CoV-2 Omicron subvariants BA.1, BA.2, and BA.4/5 and diverse sarbecoviruses

December 2022

·

347 Reads

·

42 Citations

As SARS-CoV-2 Omicron and other variants of concern (VOCs) continue spreading worldwide, development of antibodies and vaccines to confer broad and protective activity is a global priority. Here, we report on the identification of a special group of nanobodies from immunized alpaca with potency against diverse VOCs including Omicron subvariants BA.1, BA.2 and BA.4/5, SARS-CoV-1, and major sarbecoviruses. Crystal structure analysis of one representative nanobody, 3-2A2-4, discovers a highly conserved epitope located between the cryptic and the outer face of the receptor binding domain (RBD), distinctive from the receptor ACE2 binding site. Cryo-EM and biochemical evaluation reveal that 3-2A2-4 interferes structural alteration of RBD required for ACE2 binding. Passive delivery of 3-2A2-4 protects K18-hACE2 mice from infection of authentic SARS-CoV-2 Delta and Omicron. Identification of these unique nanobodies will inform the development of next generation antibody therapies and design of pan-sarbecovirus vaccines. The authors identify nanobodies from immunized alpaca with broadly neutralizing activity against SARS-CoV-1, SARS-CoV-2 variants, and major sarbecoviruses. One representative nanobody binds to a highly conserved epitope on RBD and protects K18-hACE2 mice from Omicron and Delta infection.


Fig. 4 Structural basis for the potent and broad neutralization. a Overall structure of Omicron-RBD-BA.1 bound with F61 and CDRs involved in binding are labeled. On the RBD, the footprint of F61 is represented by magenta surface and the footprint of REGN10933 (PDB ID: 6XDG) is circled by yellow line. Omicron-RBD-BA.1 residues recognized by F61 are listed and mutated N417, N477, K478, R493 and H505 in BA.1 are colored red. Additional mutations sites in BA.2 (D405 and R408), BA.3 (D405), BA.4 (D405, R408 and F486) and BA.5 (D405, R408 and F486) are colored blue. b Overall structure of Omicron-RBD-BA.1 bound with D2 and CDRs involved in binding are labeled. On the RBD, the footprint of D2 is represented by orange surface and the footprints of P2B-2F6 (PDB ID: 7BWJ) and S309 (PDB ID: 6WPS) are circled by purple and violet lines, respectively. Omicron-RBD-BA.1 residues recognized by D2 are listed and mutated S446 in BA.1 is colored red. Additional mutation site in BA.1.1 (R346) and only L452 mutation site in BA.4 and BA.5 are colored blue. c The detailed interactions between residues mutated in above Omicron sublineages and F61. F61 footprint is circled by a magenta line. Interfacing residues of Omicron-RBD are shown as cyan sticks and F61 are shown as magenta sticks. Hydrogen bonds and salt bridges are represented by dashed lines and solid lines, respectively. d The detailed interactions between residues mutated in above Omicron sublineages and D2. D2 footprint is circled by an orange line. Interfacing residues of Omicron-RBD are shown as cyan sticks and D2 shown as orange sticks. Hydrogen bonds and salt bridges are represented by dashed lines and solid lines, respectively.
Structural basis of a two-antibody cocktail exhibiting highly potent and broadly neutralizing activities against SARS-CoV-2 variants including diverse Omicron sublineages

September 2022

·

111 Reads

·

17 Citations

Cell Discovery

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs), especially the latest Omicron, have exhibited severe antibody evasion. Broadly neutralizing antibodies with high potency against Omicron are urgently needed for understanding the working mechanisms and developing therapeutic agents. In this study, we characterized the previously reported F61, which was isolated from convalescent patients infected with prototype SARS-CoV-2, as a broadly neutralizing antibody against all VOCs including Omicron BA.1, BA.1.1, BA.2, BA.3 and BA.4 sublineages by utilizing antigen binding and cell infection assays. We also identified and characterized another broadly neutralizing antibody D2 with epitope distinct from that of F61. More importantly, we showed that a combination of F61 with D2 exhibited synergy in neutralization and protecting mice from SARS-CoV-2 Delta and Omicron BA.1 variants. Cryo-Electron Microscopy (Cryo-EM) structures of the spike-F61 and spike-D2 binary complexes revealed the distinct epitopes of F61 and D2 at atomic level and the structural basis for neutralization. Cryo-EM structure of the Omicron-spike-F61-D2 ternary complex provides further structural insights into the synergy between F61 and D2. These results collectively indicated F61 and F61-D2 cocktail as promising therapeutic antibodies for combating SARS-CoV-2 variants including diverse Omicron sublineages.


Structural basis of a two-antibody cocktail exhibiting highly potent and broadly neutralizing activities against SARS-CoV-2 variants including diverse Omicron sublineages

May 2022

·

71 Reads

SARS-CoV-2 variants of concern (VOCs), especially the latest Omicron, have exhibited severe antibody evasion. Broadly neutralizing antibodies with high potency against Omicron are urgently needed for understanding working mechanisms and developing therapeutic agents. In this study, we characterized previously reported F61, which was isolated from convalescent patients infected with prototype SARS-CoV-2, as a broadly neutralizing antibody against all VOCs including Omicron BA.1, BA.1.1, BA.2, BA.3 and BA.4 sublineages by utilizing antigen binding and cell infection assays. We also identified and characterized another broadly neutralizing antibody D2 with epitope distinct from that of F61. More importantly, we showed that a combination of F61 with D2 exhibited synergy in neutralization and protecting mice from SARS-CoV-2 Delta and Omicron BA.1 variants. Cryo-EM structures of the spike-F61 and spike-D2 binary complexes revealed the distinct epitopes of F61 and D2 at atomic level and the structural basis for neutralization. Cryo-EM structure of the Omicron-spike-F61-D2 ternary complex provides further structural insights into the synergy between F61 and D2. These results collectively indicated F61 and F61-D2 cocktail as promising therapeutic antibodies for combating SARS-CoV-2 variants including diverse Omicron sublineages.


Figure S2, related to Figure 2. Neutralizing activity of isolated nanobodies against SARSCoV-2 variants. Serial dilutions of each nanobody were evaluated against pseudoviruses carrying spike protein of prototype and variants of SARS-CoV-2. Neutralizing activity was defined as the percent reduction in luciferase activities compared to no antibody controls. Results presented are representatives of three independent experiments.
Figure S3, related to Figure 2. Neutralizing activity of isolated nanobodies against hACE2-dependent sarbecoviruses. Serial dilutions of each nanobody were tested against pseudoviruses carrying spike protein of various sarbecoviruses. Neutralizing activity was defined as the percent reduction in luciferase activities compared to no antibody controls. Results presented are representatives of three independent experiments.
Figure S4, related to Figure 3. Proposed binding models of the three representative nanobodies to various RBD conformations in the context of prototype SARS-CoV-2 spike trimer. Crystal structures of 1-2C7, Nb70, and 3-2A2-4 bound to RBD are aligned to SARSCoV-2 spike trimer in four different conformations: 1) 3up RBD-S trimer (PDB: 7KMS); 2) 2up RBD-S trimer (PDB: 7A93); 3) 1up RBD-S trimer (PDB: 6VYB); and 4) 0 up RBD-S trimer (PDB: 6VXX). The spike trimers are shown as a molecular surface, with up RBD colored in cyan, down RBD in light blue, NTD and S2 in grey. 1-2C7, Nb70, and 3-2A2-4 are colored in orange, green, and purple, respectively.
Figure S5. Structural illustration of SARS-CoV-1 and SARS-CoV-2 cross-neutralizing epitopes recognized by the three representative nanobodies and various published antibodies. Cross-neutralizing epitopes on the top face of RBD is recognized by S2X146, inner face by Nb70 and 1-2C7, outer face by S309 and BG10-19, cliff face by S2H97 and 6D6, and escarpment face by 47D11 and 3-2A2-4. The glycosylation site at position 343 (N343), conserved across the sarbecovirus subgenus, is colored in dark blue. The ACE2-binding site is outlined in black.
Broadly neutralizing and protective nanobodies against diverse sarbecoviruses

April 2022

·

154 Reads

·

2 Citations

As SARS-CoV-2 Omicron and other variants of concern continue spreading around the world, development of antibodies and vaccines to confer broad and protective activity is a global priority. Here, we report on the identification of a special group of nanobodies from immunized alpaca with exceptional breadth and potency against diverse sarbecoviruses including SARS-CoV-1, Omicron BA.1, and BA.2. Crystal structure analysis of one representative nanobody, 3-2A2-4, revealed a highly conserved epitope between the cryptic and the outer face of the receptor binding domain (RBD). The epitope is readily accessible regardless of RBD in up or down conformation and distinctive from the receptor ACE2 binding site. Passive delivery of 3-2A2-4 protected K18-hACE2 mice from infection of authentic SARS-CoV-2 Delta and Omicron. This group of nanobodies and the epitope identified should provide invaluable reference for the development of next generation antibody therapies and vaccines against wide varieties of SARS-CoV-2 infection and beyond.


Deep learning guided optimization of human antibody against SARS-CoV-2 variants with broad neutralization

March 2022

·

227 Reads

·

96 Citations

Proceedings of the National Academy of Sciences

Significance SARS-CoV-2 continues to evolve through emerging variants, more frequently observed with higher transmissibility. Despite the wide application of vaccines and antibodies, the selection pressure on the Spike protein may lead to further evolution of variants that include mutations that can evade immune response. To catch up with the virus’s evolution, we introduced a deep learning approach to redesign the complementarity-determining regions (CDRs) to target multiple virus variants and obtained an antibody that broadly neutralizes SARS-CoV-2 variants.


Citations (22)


... This impressive in vitro activity translated into robust protection against both contact and respiratory transmission of the Omicron XBB.1.5 subvariant in a Syrian hamster model of SARS-CoV-2 infection [48,51]. Crystal structure analysis of Tnb04-1 revealed a highly conserved epitope consisting of a hydrophobic pocket between the cryptic and outer face of the RBD, distinct from the ACE2 binding site. ...

Reference:

Super broad and protective nanobodies against Sarbecoviruses including SARS-CoV-1 and the divergent SARS-CoV-2 subvariant KP.3.1.1
SARS-CoV-2 hijacks neutralizing dimeric IgA for nasal infection and injury in Syrian hamsters

... The de novo discovery of antibodies that activate signaling with a similar efficacy as the endogenous agonist ligand is notoriously difficult. Indeed, despite multiple reports describing positive allosteric modulating nanobodies that increase agonist affinity or signaling potency or efficacy at low ligand tonus [19][20][21][22][23] , only a few are reported to agonize the GPCR in the absence of any ligand. Some of the reported agonistic antibodies have been designed by ligand tethering (genetically fusing the agonist ligand motifs to the C-or N-terminus of the fusion partner 24,25 or grafting agonist ligand motifs into the antibody hypervariable complementarity determining region (CDR)) 26 or by structure-informed engineering 27 . ...

Structural basis of α1A-adrenergic receptor activation and recognition by an extracellular nanobody

... Since the initial outbreak of the COVID-19 pandemic, both our team and other researchers have concentrated on analyzing antibody responses in infected humans and immunized animals [9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24][25][26]. Through single-B-cell and antibody-library based strategies, a large number of human monoclonal antibodies (mAbs) and nanobodies (nbs) have been isolated and characterized for their structure and function properties . ...

Infection with wild-type SARS-CoV-2 elicits broadly neutralizing and protective antibodies against omicron subvariants

Nature Immunology

... Their single-domain structure endows them with high stability, deep tissue penetration, and ease of production, making them strong candidates for a range of therapeutic applications [2,3]. Amidst the global onslaught of the novel coronavirus, research on nanobodies has accelerated, with successful generation of effective nanobodies in biological experiments for the diagnosis of SARS-CoV-2 and treatment of COVID-19 [4,5,6,7,8,9]. Looking ahead, nanobodies are poised to become a significant innovation in modern biomedicine, aiding in the treatment and prevention of diseases [10,11]. ...

Broadly neutralizing and protective nanobodies against SARS-CoV-2 Omicron subvariants BA.1, BA.2, and BA.4/5 and diverse sarbecoviruses

... Interestingly, the results of another research group showed that more RBD non-RBM specific antibodies with neutralizing properties are produced exclusively in vaccinated patients with COVID-19 than in vaccinees alone [75]. The RBD non-RBM nAbs described by several studies could inhibit the interaction of spike with ACE2 directly [76][77][78] or indirectly [71,76,[79][80][81], when utilized in antigen binding and cell infection assays. ...

Structural basis of a two-antibody cocktail exhibiting highly potent and broadly neutralizing activities against SARS-CoV-2 variants including diverse Omicron sublineages

Cell Discovery

... The large number of mutations (15 mutations in BA.1 RBD) in Omicron variants abolished the binding of nanobody mNb6 and thus the cross-linking between mNb6(108FFY) and Omicron RBD (Figure S9). We therefore used a different nanobody Nb70 capable of binding with Omicron J o u r n a l P r e -p r o o f variants, contacting two RBDs in their up-state38 . We decided to incorporate FSY at site G56, Y103, or D115 of nanobody Nb70 based on the crystal structure38 to target the K386, Y369, or K378 of the Spike RBD, respectively (Figure S10A). ...

Broadly neutralizing and protective nanobodies against diverse sarbecoviruses

... Modeling approaches, such as phylogenetic analysis, structural modeling, and machine and deep learning, offer valuable insights into understanding viral behavior, assessing antibody and vaccine efficacy, and predicting the impact of mutations. Recent studies have successfully modeled the temporal and geographic evolution of SARS-CoV-2, determined phyletic lineages of SARS-CoV-2 variants, and predicted the impact of mutations on ACE2 binding [12][13][14] . The arrival of new influenza strains each year has also driven the development of models that predict antigenic variation of influenza, helping aid the creation of annual flu vaccines [15][16][17] . ...

Deep learning guided optimization of human antibody against SARS-CoV-2 variants with broad neutralization

Proceedings of the National Academy of Sciences

... The identification of epitopes recognized by antibodies from vaccinated subjects involved in virus neutralization is crucial not only for understanding the mechanism of the action of existing vaccines but also for the development of refined active vaccination and passive immunization strategies against COVID-19 [11,12]. Most of the current vaccines are focused on obtaining an S-specific antibody response with special attention to RBD-specific antibody response, because it has been seen that RBD-specific antibodies are correlated with high neutralizing activity against SARS-CoV-2 [13][14][15]. However, the number of mutations acquired by each new variant of concern (VOC), indicates the importance of identifying epitopes conserved among VOCs targeted by neutralizing antibodies which are located not only in RBD but also outside of RBD [16]. ...

A Potent and Protective Human Neutralizing Antibody Against SARS-CoV-2 Variants

... μg/mL,上皮细胞 IC 50 为 0.42±0.02 μg/mL [21] 。Zhu 等人 筛选出另一株人源中和单抗 1D8 [42] ,其在 B 细胞和上皮细胞感染模型中的 IC 50 分别为 0.238 μg/mL 和 0.123 μg/mL。Chen 等人近期筛选获得六株靶向 gH/gL 蛋白的人源中和单抗(769A7、 ...

A potent and protective human neutralizing antibody targeting a novel vulnerable site of Epstein-Barr virus

... The comparison of P4A2 with other known broadly neutralizing antibodies shows that epitopes of 87G7, 510A5, Cov2-2196, NCV2SG48, NCV2SG53, S2E12, S2K146 and ZWD12 show varying degrees of overlap with that of P4A2. Among these mAbs, P4A2 is the only one that forms multiple interactions with its cognate epitope on spike-RBD which is composed of residues that are critical for interaction with ACE2 (S11 Fig and S2 Table) [8][9][10][11][12][13][14][15][16][17][18][19][20][21]. Overall, our data suggests that P4A2 may represent a viable therapeutic option which is required to reduce the impact of the COVID19 pandemic on human health across the globe. ...

SARS-CoV-2 hijacks neutralizing dimeric IgA for enhanced nasal infection and injury