Expression of S2 Proteins
Six proline mutations were introduced in DNA encoding the S2 subunit regions of SARS-CoV-2 (YP_009724390.1, residues 686–1208), RsSHC014 (AGZ48806.1, residues 669–1191), and SARS-CoV-1 (P59594, residues 668–1190) based on the SARS-COV-2 HexaPro spike protein.26 Additional mutations were made to residues to eliminate the S2’ protease cut site of the SARS-CoV-2 (K814G and R815G)14, RsSHC014 (T796S, K797G, and R798G), and SARS-CoV-1 (T792S, K793G, and R794G) S2 regions. Each of these DNA segments was cloned into pcDNA3.1 (-) with the addition of an N-terminal mouse Ig Kappa signal peptide, C-terminal T4 fibritin trimerization domain, AviTag, and his-tag between the Ncol and Xhol restriction sites by Gene Universal, Inc. (Newark, DE). These three plasmids were transfected into Expi293F cells (RRID: CVCL_D615) per manufacturer protocol using the ExpiFectamine Transfection Kit (Thermo Fisher Scientific). Cultures were centrifuged at 6,000xg for 10 minutes 5 days post-transfection. The supernatant was collected and dialyzed into PBS for 2 hours, after which it was dialyzed in fresh 1X PBS for an additional 2 hours. The supernatant was then mixed with 1 mL of HisPure Ni-NTA resin (Thermo Fisher Scientific) and incubated overnight on a stir plate. The rest of the purification was done as described previously.12,14,25The supernatant resin mixture was then loaded into a gravity flow column (G-Biosciences). After the column was washed with 90 mL of binding buffer (150 mM Tris, 150 mM NaCl, 20 mM Imidazole, pH 8), the resin was incubated for 5 minutes with 3 mL of elution buffer (150 mM Tris, 150 mM NaCl, 400 mM Imidazole, pH 8). Eluate was collected after the incubation period and the elution process was repeated two more times resulting in a total volume of 9 mL. A 10 kDa MWCO spin filter (Millipore Sigma) was used to concentrate down the eluate, which was then further purified using a Superdex 200 Increase 10/30 column in PBS.12,14,25 Protein concentration was quantified using the bicinchoninic acid assay (BCA) assay (Thermo Scientific).
Expression and Purification of MS2
The expression and purification of MS2 have been described previously.12,14,24,25 In brief, DNA encoding a single chain dimer of the MS2 coat protein was cloned into pET-28b between the NdeI and XhoI restriction sites by GenScript Biotech Corporation (Piscataway, NJ) with the additional insertion of an AviTag between the 14th and 15th residues of the first coat protein monomer. This plasmid was co-transformed with a BirA-biotin-protein ligase expressing plasmid into BL21(DE3) competent E. coli (New England Biolabs) per manufacturer protocol. A 5 mL 2xYT media starter culture was made using the transformed E. coli, which were incubated at 37°C overnight in a rotating incubator.12,14,24,25The starter culture was then added to 1 L of 2xYT and further grown at 37°C in a shaking incubator at 225 rpm until it reached an optical density of 0.6–0.8, after which the culture was induced by adding 1 mL of of 1 M IPTG (Fisher BioReagents). Simultaneously, 50 mM D-biotin was added to the culture, and the incubation temperature was reduced to 30°C.12,14,24,25The culture was centrifuged at 7000xg for 7 minutes after overnight incubation. The cell pellet was resuspended in 25 mL of lysis buffer (20 mM Tris base, 0.5 mg/mL lysozyme, 125 units of EMD Millipore benzonase, a quarter of a SigmaFast EDTA-free protease inhibitor cocktail tablet, pH 8) and incubated at 4°C on a rocker for 20 minutes. Sodium deoxycholate (Alfa Aesar) was then added to the lysate to reach a final concentration of 0.1% (w/v). The lysate was sonicated on ice at 35% amplitude with 3-second pulses for 3 minutes (Sonifier S-450, Branson Ultrasonics).12,14,24,25 The lysate was sonicated again after being allowed to rest on ice for 5 minutes. The lysate was centrifuged at 19,000xg for 30 minutes, the supernatant was collected and centrifuged again. The supernatant was collected a second time and diluted with 20 mM Tris Base, pH 8 to reach a total volume of 100 mL.12,14,24,25 The supernatant was loaded onto four HiScreen Capto Core columns (Cytvia) in series using an Akta Start system. The columns were washed with 5 column volumes (CVs) of 20 mM Tris Base, pH 8 to elute out the MS2 protein. Collected MS2 fractions were combined and concentrated down using a 10 kDa MWCO spin filter (Millipore Sigma) before being further purified using a Superdex 200 Increase 10/30 column in 20 mM Tris, 20 mM NaCl, pH 8.12,14,24,25 Protein concentration was quantified using the BCA assay (Thermo Scientific).
In vitro Biotinylation of AviTagged MS2 and S2 proteins
The S2 and MS2 proteins were biotinylated using a BirA biotin-protein ligase standard reaction kit (Avidity LLC). A mixture of Biomix B (ATP, biotin, magnesium acetate) and BirA were added to the protein solutions, which were then mixed overnight at 4°C. The following day, the protein-BirA solution was further supplemented with Biomix B and mixed at 37°C for 2 hours. After 2 hours, additional Biomix B was added to the solution and left to mix overnight at 4°C. The biotinylated proteins were then purified using a Superdex 200 Increase 10/30 column in 1X PBS to remove the excess BirA and biotin. The final protein concentration was quantified using the BCA assay (Thermo Scientific).12,14,25
Expression, Refolding, and Purification of Streptavidin
The expression, refolding, and purification of streptavidin (SA) have been extensively described previously.12,14,24,25,38,39 SA encoding plasmid (Addgene plasmid #46367, a gift from Mark Howarth), was transformed into BL21(DE3) cells (New England Biolabs) per manufacturer protocol. The following protocol describes SA expression, refolding, and purification on a 2 L scale. Two 5mL 2xYT starter cultures were made using the transformed BL21(DE3) cells. 12,14,24,25,38,39 The cultures were grown overnight in a 37°C rotation incubator. The next morning, each starter culture was added to 1 L of 2xYT media and grown further on a shaking incubator until the OD reached 0.6–1.0, after which the cultures were induced by the addition of 1 mL of 1 M IPTG (Fisher BioReagents).12,14,24,25,38,39 The temperature was reduced to 30°C, and the cultures were incubated overnight. The next day, the cultures were combined and centrifuged at 7000 ×g for 7 minutes. The resulting cell pellet was resuspended with 50 mL of resuspension buffer (50 mM Tris, 100 mM NaCl, pH 8.0) supplemented with 1 mg/mL lysozyme (Alfa Aesar) and 500 units of benzonase (EMD Millipore).12,14,24,25,38,39 The lysate was incubated on a rocker at 4°C for 1 hour and homogenized for 30 seconds. Sodium deoxycholate (Alfa Aesar) was then added to the lysate to reach a final concentration of 0.1% (w/v), after which the lysate was sonicated at 35% amplitude for 3 minutes with 3-second pulses. The sonicated lysate was then centrifuged at 27000 ×g for 15 minutes.12,14,24,25,38,39 The supernatant was discarded, and the above-mentioned lysis process was repeated, except benzonase was excluded in the lysis buffer and the 4°C incubation period was reduced to 30 minutes. Following the second centrifugation, the pellet was resuspended in 50 mL of wash buffer #1 (50 mM Tris, 100 mM NaCl, 100 mM EDTA, 0.5% (v/v) Triton X-100, pH 8.0), homogenized for 30 seconds, and then sonicated for 30 seconds at 35% amplitude. The lysate was centrifuged at 27,000×g for 15 min, and this wash process was repeated two more times.12,14,24,25,38,39 The pellet was then washed with a second wash buffer (50 mM Tris, 10 mM EDTA, pH 8.0), homogenized for 30 seconds, sonicated for 30 seconds at 35% amplitude, and centrifuged at 15,000×g for 15 min. This wash process was repeated one more time. The pellet was then resuspended in 10 mL of resuspension buffer.12,14,24,25,38,39 Guanidine hydrochloride was added to the mixture to reach a final concentration of 7.12 M then mixed at room temperature for 1 hour. The mixture was then centrifuged at 12,000xg for 12 minutes, transferred to a syringe, loaded onto a syringe pump, and allowed to drip at a rate of 30 mL/hour into 1 L of chilled, rapidly stirring PBS. The solution was mixed overnight at 4°C then centrifuged the next day at 17000xg for 15 minutes to remove insoluble protein.12,14,24,25,38,39 The supernatant was then filtered through a 0.45-µm bottle-top filter. Ammonium sulfate was slowly added to the filtrate while it was rapidly stirring to a final concentration of 1.9 M. The solution was mixed further for 3 hours at 4°C, centrifuged again at 17000xg for 15 minutes, then filtered again. 12,14,24,25,38,39 More ammonium sulfate was added to the filtrate while it was stirring vigorously to a final concentration of 3.68 M. The mixture was stirred overnight at 4°C, and the SA was pelleted out by centrifuging it at 17000xg for 15 minutes. The SA pellet was resuspended using 20 mL of Iminobiotin Affinity Chromatography (IBAC) binding buffer (50 mM Sodium Borate, 300 mM NaCl, pH 11.0). Afterward, 5 mL of Pierce Iminobiotin Agarose (Thermo Scientific) was added to a gravity flow column (G-Biosciences) and then equilibrated with 25 mL of IBAC binding buffer.12,14,24,25,38,39 The resuspended SA was loaded onto the column and then washed with 50 mL of IBAC binding buffer. The SA was eluted with 20 mL of IBAC elution buffer (20 mM Potassium Phosphate, pH 2.2) and dialyzed in PBS overnight. The SA was then concentrated using a 10 kDa MWCO centrifugal filter (Millipore Sigma), and the concentration was measured by absorbance at 280 nm.12,14,24,25,38,39
Assembly and Purification of MS2-SA VLPs
MS2-SA VLPs were generated as described previously.12,14,24,25 A concentrated solution of 20x molar excess of SA (at a minimum concentration of 30 mg/mL) was stirred rapidly in a small glass vial. Biotinylated MS2 (at a maximum concentration of 700 µg/mL) was added in 2.5 µL increments to the SA. The MS2-SA mixture was then loaded onto a Superdex 200 Increase 10/30 column in PBS to separate the MS2-SA VLPs and excess SA. The MSA-SA samples were quantified via SDS polyacrylamide gel electrophoresis (SDS-PAGE). The MS2-SA samples were mixed with Nu-PAGE lithium dodecyl sulfate (LDS) sample buffer (Invitrogen) and then heated for 30 minutes at 90°C. Afterward, the MS2-SA samples were loaded onto a polyacrylamide gel with SA standard samples of known concentrations. The SA band intensities of the MS2-SA samples were compared to the band intensities of the SA standard samples to determine the SA concentration of the MS2-SA samples.12,14,24,25
Preparation of VLP-S2
VLPs were generated as described previously.12,14,24,25 The optimal ratio of biotinylated S2 to MS2-SA was determined using analytical SEC. Mixtures of 8 µg of the S2 antigens and varying amounts of MS2-SA were loaded onto a Superdex 200 increase 10/300 SEC column (Cytiva). The optimal ratio was determined to be the ratio that included the least amount of MS2-SA in the mixture and also resulted in a chromatogram without a peak corresponding to excess S2 antigen.12,14,24,25
SDS-PAGE
Protein samples were deglycosylated with PNGase F (New England BioLabs), then diluted with 2 µL of 2-Mercaptoethanol and 5 µL of Nu-PAGE lithium dodecyl sulfate (LDS) sample buffer (Invitrogen) as described previously.14 Samples were heated at 98°C for 30 minutes then loaded alongside a PageRuler Plus Prestained Protein Ladder (Thermo Scientific) into a 4–12% Bis-Tris gel (Invitrogen). The gel was run at 120 V for 50 minutes in MES-SDS buffer. The gels were imaged with ChemiDoc MP imaging system and Image Lab 5.2.1 software (Bio-Rad) after being stained with Imperial Protein Stain for 30 minutes and destained overnight.14
Expression of S2P6 Antibody
The S2P6 antibody40 was expressed as described previously.12 The heavy and light chains were cloned into TGEX-HC and TGEX-LC vectors (Antibody Design Labs), respectively, and expressed in Expi293F cells using the ExpiFectamine Transfection Kit (Thermo Fisher Scientific). After a 6-day 37°C incubation, cells were centrifuged at 6000 xg for 15 minutes, and the supernatant was diluted in MabSelect Binding Buffer (20 mM sodium phosphate, 150 mM NaCl, pH 7.2. The diluted supernatant was then injected onto a 1-mL MabSelect SuRe column (Cytiva) connected to an ÄKTA Start to purify the protein. The protein was then further purified by SEC. Fractions with the protein were collected, dialyzed in PBS, and the protein concentration was determined using the BCA assay. Transfection and MabSelect purification were done according to manufacturer instructions.
S2 and VLP-S2 ELISA
ELISA characterization was done as described previously.12,14,24,25 Protein samples were diluted in PBS to a concentration of 1 µg of S2 per 1 mL. For each well of a Nunc Maxisorp 96-well plate, 100 µL of diluted protein solution was added (total 0.1 µg of S2 per well) and incubated for 1 hour. The protein solution was removed from the plate, which was then blocked with 200 µL of a 5% BSA (EMD Millipore) in PBST (0.05% Tween-20) and allowed to incubate for another hour. Afterward, the BSA solution was discarded, and the wells were washed three times with 100 µL of PBST. A stock solution of the primary S2P6 antibody (at 1.9 mg/mL) was diluted 1:30000 in 1% BSA in PBST and 100 µL of this solution was added to the wells. After 1 hr, the solution was removed, and the plate was washed again three times with PBST, after which 100 µL of horseradish peroxidase-conjugated anti-human IgG Fc goat antibody (MP Biomedical, catalog #674171) diluted 1:5000 in 1% BSA in PBST was added to the wells.12,14,24,25 After incubating for an hour, the solution was removed, and the plates were washed again three times with PBST. 100 µL of TMB (Thermo Scientific) was added to each well, and after 3 minutes, 160 mM sulfuric acid was added to stop development. The plates were read using a Synergy H4 plate reader (BioTek) with Gen5 2.07 software (BioTek) at 450 nm.12,14,24,25
Dynamic Light Scattering
Measurements were taken using a Zetasizer Nano (Malvern). S2 and VLP-S2 proteins were diluted so there was 5 µg of S2 per 100 µL of PBS, and MS2-SA was diluted so there was approximately 2 µg in 100 µL. 100 µL of diluted protein solution was added to a UVette (Eppendorf), and 13 acquisitions per sample were collected at 25°C. Results were displayed as % volume.
Analytical SEC
Analytical SEC was done as described previously.12,14,24,25 S2 and VLP-S2 proteins were diluted so there was 8 µg of S2 per 950 µL of PBS. They were loaded into a Superdex 200 Increase 10/300 Column (Cytiva) using the Unicorn 7 control system (Cytiva). They were eluted with a full column volume of PBS flowing at 0.65 mL/min, while the absorbance was monitored at 210 nm.
Amino acid Identity and Phylogenetic Trees
Phylogenetic trees and identity tables were generated as described previously.12 All spike amino acid sequences were retrieved from GenBank. Clustal Omega 1.2.3 (Conway Institute, UCD Dublin) was used to align sequences. These alignments were used to calculate the percent identity of amino acids for each paired comparison. PhyML 3.3.20220408 (Stephane Guindon, University of Montpellier) was used to generate the Maximum likelihood phylogenetic trees. This program used the LG amino acid substitution model and a maximum parsimony starting tree. TreeViewer 2.0.1 (Giorgio Bianchini, University of Bristol) was used to visualize the phylogenetic trees.
Negative-stain Transmission Electron Microscopy
We performed conventional negative-stain transmission electron microscopy (NS-TEM) on VLP-S variants, as described previously.12,14,25 Briefly, 4 µl of the diluted samples were applied onto glow-discharged 200-mesh copper grids (CF200-Cu; Electron Microscopy Sciences, PA). The grids were washed with drops of distilled water (3X), drops of staining solution of 1% phosphotungstic acid (PTA, pH 6 ~ 7) (2X), followed by 1 minute of staining drop incubation. Excess stain was removed from the grids with backside blotting with filter paper, the grids were then allowed to air-dry. The grids were imaged with a low dose of 50 ~ 60 e-/Å2, under a nominal magnification of 73 kx (pixel size of 2.0 Å), defocus of -0.5 to -2 µm, on a Talos L120C transmission electron microscope (TEM, ThermoFisher Scientific, Hillsboro, OR), operating at 120 kV. Images were captured on a 4K x 4K Ceta CMOS camera (ThermoFisher Scientific, Hillsboro, OR) using the SerialEM 3.841 software package.
Biosafety and Containment for Coronaviruses
Research with sarbecoviruses was performed under biosafety level 3 agriculture (BSL-3 AG) containment at the Influenza Research Institute with an approved protocol reviewed by the University of Wisconsin-Madison’s Institutional Biosafety Committee. The laboratory is designed to meet and exceed the standards outlined in Biosafety in Microbiological and Biomedical Laboratories (6th edition).
Approval of Animal Studies
Immunization and challenge studies at the University of Wisconsin-Madison were performed under an approved protocol (Protocol Number: V006426) reviewed by the Institutional Animal Care and Use Committee. To minimize pain, virus infections were performed under isoflurane anesthesia. Mouse immunizations for B cell characterization were performed with the approval of the Weill Cornell Medical College Institutional Animal Care and Use Committee (Protocol Number: 2021-0024). Group sizes were determined based on previous S2-antigen vaccine studies, and no sample-size calculations were performed prior to the study in order to determine the power of these studies. Animal groups were not blinded to the researchers.
Cell Lines and Viruses
All virus stocks were propagated on Vero E6 TMPRSS2 cells (National Institute of Infectious Diseases, Japan) which were maintained in high glucose Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum (FBS) and antibiotic/antimycotic solution along with G418 (1 mg/ml). For consistency, virus titrations of tissue samples were performed on Vero E6 TMPRSS2-T2A-ACE2 cells (NIAID Vaccine Research Center; Dr. Barney Graham). This cell line was maintained in DMEM supplemented with 10% FBS, 10 mM HEPES (pH 7.3) and antibiotic/antimycotic solution along with puromycin (10 µg/ml). Both cells are tested monthly for mycoplasma contamination by PCR and are confirmed to be mycoplasma-free.
In the mouse challenge studies, the following viruses were used, SARS-CoV-2 mouse-adapted strain (MA10 variant), XBB (hCoV-19/Japan/TY41-795/2022 (Accession ID: EPI_ISL_16355653), Banal-20-236/Laos/2020 (GenBank: MZ937003), WIV1 (GenBank: KF367457), RsSHC014 (GenBank KC881005), and BetaCoV/pangolin/Guandong/1/2019 (Accession ID: EPI_ISL_410721). For the live virus neutralization assay, the early isolate SARS-CoV-2/UT-HP095-1N/Human/2020/Tokyo (HP095) was used.
Mouse Immunizations and Challenge Studies
C57BL/6, K18-hACE2, and BALB/c mice were purchased from Jackson Laboratories while FcgR knockout mice and the wild-type BALB/c control mice were purchased from Taconic. All mice were females and at the age of 10–12 weeks at the start of the studies. Mice were immunized by subcutaneous injection with a total volume of 250 µl of an equal mixture of VLP vaccine preparations (7.5 µg of each S2 antigen) and adjuvant mixture (AddaS03 with poly I:C [InvivoGen]). Additional mice were immunized with Pfizer’s BNT162b2 mRNA virus at a dose of 1.0 µg by intramuscular inoculation. Under anesthesia (isoflurane), mice were infected intranasally with 105 plaque-forming units (pfu) of challenge virus in 30 µl of total volume of diluted virus. Animals were humanely sacrificed by an overdose of isoflurane three days after infection to collect lung tissue to measure the amount of virus.
Anti-CD8 Antibody Treatment
To deplete CD8 + T cells, mice were inoculated intraperitoneally with 500 µl of PBS with the anti-CD8 depleting antibody (Harlan Sprague-Dawley, clone 2.43, lot # 70088).28 Treatments started three days and one day prior to immunization (day − 3 and day − 1), on the day of immunization (day 0), and after immunization (day + 1, +3, + 6, +9 and + 12).
B Cell Characterization Studies
The following B cell characterization studies were performed using cells isolated from mouse spleen samples that were collected four weeks after immunization.
Probe Generation for B Cell Characterization
Subunit proteins (RBD, NTD, and S2) and HexaPro or 2P spike proteins (SARS-CoV-2 XBB.1.5, SARS-CoV-1, MERS-CoV, OC43, HKU1, and NL63) were biotinylated for 30 min on room temperature using EZ-Link Sulfo-NHS-Biotin, No-Weigh Format (Thermo Fisher) according to the manufacturer’s instructions. Unreacted biotin was removed by passage through a 7K MWCO desalting column (Zeba spin, Thermo Fisher). Biotinylated proteins were then conjugated to Biolegend TotalSeq PE streptavidin (PE-SA), APC streptavidin (APC-SA) oligos at a 0.72:1 molar ratio of antigen to PE-SA or APC-SA. The amount of antigen was chosen based on a fixed amount of 0.5 µg PE-SA or APC-SA and diluted in a final volume of 10 µL. PE-SA or APC-SA was then added gradually to 10 µL biotinylated proteins 5 times on ice, 1 µL PE-SA or APC-SA (0.1 mg/ml stock) every 20 minutes for a total of 5 µL (0.5 µg). The reaction was then quenched with 5 µL 4mM Pierce biotin (Thermo Fisher) for 30 minutes for a total probe volume of 20 µL. Probes were then used immediately for staining.
Antigen-specific B Cell Sorting
For antigen-specific B cell sorting, B cells were enriched using EasySep Mouse Pan-B Cell Isolation Kit (STEMCELL). B cells were stained with anti-mouse B220 FITC (BioLegend), anti-mouse IgM PE-Cyanine7 (Thermo Fisher), anti-mouse IgD APC-Cyanine7 (BioLegend), and antigen probes (PE or APC) for 30 min on ice in 1× phosphate-buffered saline (PBS) supplemented with 0.2% bovine serum albumin (BSA) and 2 mM Pierce biotin. Cells were subsequently washed with 1× PBS with 0.2% BSA and resuspended at a maximum of 10 million cells/ml in 1× PBS supplemented with 0.2% BSA and 2 mM Pierce biotin for downstream cell sorting using the Aurora CS Cell Sorter (Cytek Biosciences). BB220+/IgM-/IgD-/antigen-PE-positive cells were sorted as probe positive. Cells were then collected from the sorter and used for downstream processing with a chromium controller (10X Genomics).
Single-cell RNA-seq and B cell Receptor Sequencing
The mouse B cell V(D)J, 5′ gene expression, feature barcode libraries were prepared according to the manufacturer’s instructions. Libraries were pooled and sequenced using an Illumina NextSeq1000. Raw sequence processing, sample demultiplexing, barcode processing, single-cell 5′ transcript counting, and B cell receptor repertoire sequence assembly were performed as previously described.42
Monoclonal Antibody Production.
Antibody heavy and light chain genes obtained by 10X Genomics V(D)J sequencing analysis were synthesized by Integrated DNA Technologies. The synthesized fragments for heavy and light chains with 5′ and 3′ Gibson overhangs were then cloned into human IgG1 and human kappa or lambda light chain expression vectors by Gibson assembly as previously described.43 The heavy and light chains of the corresponding mAb were co-transfected into HEK293T cells. After 4 days, mAbs secreted into the medium supernatant were harvested and purified using protein A-agarose beads (Thermo Fisher).
Enzyme-linked Immunosorbent Assay (ELISA) for mAbs and Mouse Serum.
High-protein-binding microtiter plates (Costar) were coated with 50 µl of subunit or spike proteins at 2 µg/ml in a 1× PBS solution overnight at 4°C. The plates were washed 3 times the next day with 1× PBS supplemented with 0.05% Tween 20 and blocked with 200 µl of 1× PBS containing 20% fetal bovine serum (FBS) for 1 h at 37°C. mAbs were serially diluted 1:3 starting at 10 µg/ml while mouse sera were serially diluted 1:2 starting at 1:20; both were incubated for 1 h at 37°C. The plates were then washed 3 times and incubated with horseradish peroxidase (HRP)-conjugated goat anti-human IgG antibody for mAbs or anti-mouse IgG antibody for mouse serum (Jackson ImmunoResearch) diluted 1:5,000 for 1 h at 37°C, and plates were subsequently developed with the Super AquaBlue enzyme-linked immunosorbent assay (ELISA) substrate (eBioscience) for 10 min. The absorbance was measured at 405 nm on a microplate spectrophotometer (Bio-Rad). All mAbs were tested in duplicate, and each experiment was performed twice, while mouse serum was tested in singlet from groups of four immunized mice.
Monoclonal Antibody Pseudovirus Neutralization Assay.
Virus neutralization assays were performed with SARS-CoV-2 spike pseudotyped virus that generated with the recombinant replication-deficient vesicular stomatitis virus (VSV) containing luciferase instead of the VSV-glycoprotein (VSV-G) gene (Kerafast). The 50% tissue culture infectious dose (TCID50) of the pseudotyped virus is calculated as previously described.44 Vero E6-ACE2-TMPRSS2 cells (BEI) were seeded in 96-well white, flat-bottom plates (Corning) at 40,000 cells/well in culture medium and cultured overnight at 37°C with 5% CO2. Each mAb was four-fold serially diluted in culture medium from 100 µg/mL (100–0.39 µg/ml). mAb dilutions were mixed 1:1 with a certain amount (325–1,300 TCID50/ml) of pseudovirus for 30 minutes at 37°C prior to addition to Vero E6-ACE2-TMPRSS2 cell monolayers and incubation at 37°C with 5% CO2 for 24 hours. Supernatants were removed, and the cells were lysed with luciferase reagent (Promega). Luminescence was measured on a Spectramax M5 (Molecular Devices), and neutralization titers (IC50) were calculated using an Excel macro.44
Mouse Serum Focus Reduction Neutralization Test (FRNT).
Serial dilutions of sera from immunized mice starting at a final concentration of 1:20 were mixed with ~ 1000 focus-forming units (FFU) of an early virus isolate (HP095) for each well and incubated for 1 h at 37°C. Pooled serum from mice immunized with VLP only served as a control. The antibody-virus mixture was inoculated onto Vero E6/TMPRSS2 cells in 96-well plates and incubated for 1 h at 37°C. An equal volume of methylcellulose solution was added to each well. The cells were incubated for 16 h at 37°C and then fixed with formalin. After the formalin was removed, the cells were immunostained with a mouse monoclonal antibody against SARS-CoV-1/2 nucleoprotein (clone 1C7C7, Sigma-Aldrich, catalog #MA5-29982, 1:10,000 dilution), followed by a horseradish peroxidase-labeled goat anti-mouse immunoglobulin (ThermoFisher, catalog #31430, 1:2000 dilution). The infected cells were stained with TrueBlue Substrate (SeraCare Life Sciences) and then washed with distilled water. After cell drying, the focus numbers were quantified by using an ImmunoSpot S6 Analyzer, ImmunoCapture software, and BioSpot software (Cellular Technology). The results are expressed as the 50% focus reduction neutralization titer (FRNT50).
Antibody-dependent Cellular Cytotoxicity (ADCC) Assay
ADCC reporter assays were performed using ADCC Reporter Bioassay kits (Promega) according to the manufacturer’s instructions. The activation of ADCC signaling in effector cells were measured using CHO cells expressing SARS-CoV-2 spike. Briefly, CHO cells were plated at a density of 10,000 cells/well in a flat-bottom white 96-well plate (Corning). The cells were used as target cells 24 h later. The medium of the target cells was replaced with test mAb five-fold-diluted (10–0.08 µg/mL) in the assay buffer (Promega). The mAb clone 241-15-068A 1E04 (anti-HA mAb) was used as a negative control.45 ADCC bioassay effector cells (a Jurkat cell line stably expressing human FcγRIIIa, human CD3γ and an NFAT-response element driving expression of a firefly luciferase) were added to the antibody-treated target cells and incubated for 6 h at 37°C. The firefly luciferase activity was then measured using luciferase assay regents (Promega).
Statistics and Reproducibility
SDS-PAGE gel of S2 and S2-VLP samples (Fig. 2b) was run twice using different preparations of each sample with similar results. At least 80 images were collected for TEM imaging (Fig. 2d) and analyzed from one S2-VLP preparation per sample with similar results. The binding characterization of S2P6 by ELISA (Fig. 2e) was conducted once in triplicate for each condition, presented as mean ± SD. Endpoint titers (Fig. 3b) were determined by conducting a single assay using sera from each mouse (n = 4). The data is presented as geometric mean ± geometric SD, significance was determined by Brown-Forsythe ANOVA and Games-Howell's multiple comparisons between groups for SARS-CoV-2, Pangolin-GD, SARS-CoV-1, and RsSHC014 S proteins, and by ordinary one-way ANOVA and Tukey post hoc multiple comparisons between groups for Rc-o319 and WIV1 S proteins. Virus titers in the lung of VLP-S2 immunized mice challenged with MA10, XBB, Pangolin-GD, BANAL-236, WIV1, and RsSHC014 (Fig. 3a and 3c) were presented as mean ± SD (n = 4 for all groups). The significance was determined by an ordinary one-way analysis of variance (ANOVA) and Tukey's multiple comparisons between groups (α = 0.05) for MA10, XBB, Pangolin-GD, and RsSHC014 lung titers and by Brown-Forsythe (ANOVA) and Games-Howell’s multiple comparisons between groups (α = 0.05) for BANAL-236 and WIV1 lung titers. Lung titers for Pfizer-BioNTech bivalent vaccine immunized mice challenged with WIV1 (Fig. 3d) were presented as mean ± SD (n = 4 for all groups). Significance was determined by two-tailed Welch’s t-test. For the T-cell depletion study (Fig. 4a), virus titers in the lung of immunized mice were presented as mean ± SD (mean ± SD, n = 5 for VLP control and S2 CD8(+) groups, n = 4 for S2 CD8(-) group). The significance was determined by an ordinary one-way ANOVA and Tukey’s multiple comparisons between groups (α = 0.05). For the FcgR KO mice study (Fig. 4b), virus titers in the lung of immunized mice were presented as mean ± SD (n = 5 for wild-type BALB/c mice and n = 4 for FcR KO mice). Significance for somatic hypermutations and complementarity determining region 3 (CDR3) amino acid length in the IGHV (Fig. 5d and 5e) was determined by ordinary one-way ANOVA and Dunnett's test. Significance was determined by two-tailed Welch’s t-test. Virus titers in the lung of mice vaccinated with two doses of the VLP-S2s and challenged with XBB, Pangolin-GD, BANAL-236, and WIV1 (Fig. 6) were presented as mean ± SD (n = 5 for all groups). The significance was determined by an ordinary one-way analysis of variance (ANOVA) and Tukey's multiple comparisons between groups (α = 0.05) for XBB and WIV1 lung titers and by Brown-Forsythe (ANOVA) and Games-Howell's multiple comparisons between groups (α = 0.05) for BANAL-236 and Pangolin-GD lung titers. For tests of significance, assumptions of the normality of residuals and homogeneity of variance were validated by the D’Agostino-Pearson test and Brown–Forsythe test, respectively. All statistical analysis was carried out using Prism 9 (GraphPad).