Cell lines
Vero, HEK293T and HEK293T-hACE2 cells were cultured in Dulbecco’s modified Eagle medium (DMEM, high glucose; BI) supplemented with 100 U/mL of penicillin, as well as 100 μg/mL streptomycin solution (VivaCell), and 10% fetal bovine serum (FBS, GIBCO) in a 5% CO2 environment at 37°C. Trypsin-EDTA (0.25%, BI) was used to detach cells for subculturing every 2-3 days. Bone marrow-derived cells (BMDCs) and peritoneal macrophages were derived from female BALB/c mice (6-8 weeks old) using standard techniques.
Animals
Female BALB/c mice (6-8-weeks old) and female K18-hACE2 mice were purchased from Beijing Vital River Laboratories and GemPharmatech respectively, and were randomly divided into various groups for subsequent experiments. This study was performed in strict accordance with the Regulations for the Care and Use of Laboratory Animals and Guideline for Ethical Review of Animal (China, GB/T 35892-2018). BALB/c mice were reviewed and approved by the Animal Ethics Committee of the Institute of Process Engineering (approval ID: IPEAECA 2020061). K18-hACE2 mice experiments were reviewed and approved by the Peking Union Medical College Animal Care and Use Committee (approval ID: DWSP202302 001).
Female Syrian Golden hamsters were purchased from Beijing Vital River Laboratories, and were randomly divided into various groups for subsequent experiments. All animal experiments were reviewed and approved by the Peking Union Medical College Animal Care and Use Committee (approval ID: DWSP202207 001, DWSP202302 001).
A total of 10 cynomolgus monkeys were purchased from the Animal Center of the Academy of Military Medical Sciences and were also housed in the Center. This study was performed in strict accordance with the Regulations for the Care and Use of Laboratory Animals and Guideline for Ethical Review of Animal (China, GB/T 35892-2018). All animal experiments were approved and conducted in accordance with the institutional guidelines of the Academy of Military Medical Sciences Institutional Animal Care and Use Committee (approval ID: IACUC-DWZX-2021-058)
Human sera
Sera from 5 convalescent patients (admission date from 2020/1/21 to 2020/1/26, discharge date from 2020/2/1 to 2020/2/13, follow-up after two weeks; severity grading: mild or moderate) were provided by Beijing You An Hospital, Capital Medical University. Written informed consent was obtained from each individual for serum collection. This study was reviewed and approved by the Clinical Ethics Committee of Beijing You An Hospital, Capital Medical University (approval ID: LL-2022-063-K).
METHOD DETAILS
Protein expression and purification
ST-CNP was expressed and purified from Escherichia coli. Briefly, DNA sequences of ST-CNP were cloned to pET28a vector in the following order: SpyTag, GSGTAGGGSGS linker, CTB, GGSG linker, trimer-forming peptide, and 6×His tag. The construct was transformed into E. coli BL21. A Single colony was amplified in 10 mL Luria-Bertani (LB) with kanamycin (50 μg/mL) for 12 h at 37℃ while shaking. The preculture was diluted 1:100 in 1L LB medium (50 µg/mL kanamycin) and cultured at 200 rpm at 37°C, until OD600~0.6. Protein expression was induced with isopropyl β-D-1-thiogalactopyranoside (IPTG) and grown at 30 °C, 200 rpm for 12 h. The culture was centrifuged and the pellet was resuspended in 20 mM Tris-HCl (pH8.0), 300 mM NaCl, 8 M Urea, and 20 mM Imidazole. The cell suspension was homogenized and centrifuged at 10,000 × g for 20 min at 4 °C. The precipitate was incubated with His-Tag purification resin column to enrich ST-CNP, followed by protein elution with wash buffer (20 mM Tris–HCl, pH 8.0, containing 300 mM NaCl, 8 M Urea, and 300 mM Imidazole). The purified protein was dialyzed and finally the buffer was replaced with conventional PBS buffer. The concentration of ST-CNP was determined by BCA assay.
SC-RBD was expressed and purified from HEK293T. Briefly, DNA sequences of SC-RBD were cloned to pcDNA3.4 vector in the following order: SpyCatcher, GGSG linker, RBD and 6×His tag. The construct was transformed into HEK293T cells. Five days later, supernatants were collected and centrifuged at 500 g for 10 min at 4 °C. The cleared supernatants were incubated through His-Tag purification resin column to enrich SC-RBD proteins, followed by elution with Imidazole-containing Tris buffer. The purified proteins were concentrated and buffer-replaced with PBS buffer. The concentrations of SC-RBD were determined by BCA assay.
For vaccine generation, SC-RBD was incubated with ST-CNP in PBS buffer and reacted overnight at 4℃. The ST/SC conjugated proteins were proceeded to size-exclusion chromatography (SEC). The elution of nanoparticles was concentrated, and the concentration was measured by BCA assay. Coomassie blue staining, western blotting against His tag and CTB, transmission electron microscopy, and dynamic light scattering, were executed to confirm the purity and homogeneity.
Surface Plasmon Resonance (SPR) assay
Interactions between the hACE2 protein and R-CNP were analyzed using the Biacore 8K system (Cytiva) at 25 °C. RBD or R-CNP were immobilized in pairs onto CM5 chips. Subsequently, gradient concentrations of hACE2 (from 7.813 nM to 500 nM for SARS-CoV-2 RBD and R-CNP) were then used to flow over the chip surface at 30 μL/min with a contact time of 120 s and a dissociation time of 300 s. After each cycle, the sensor surface was regenerated using 3 M magnesium chloride. Data were analyzed with the Biacore Insight Evaluation Software 3.0.12 by curve fitting using a 1:1 binding model.
Fabrication and characterization of R-CNP@M
Membrane emulsification
Briefly, given concentration (from 0.05% to 0.5%, w/v) NaCl solution was added into 12 mL of ethyl acetate containing 100 mg/ml compound (90 mg poly(DL-lactic-co-glycolic acid) and 10 mg poly(ethylene glycol)-b-poly-dl-lactide) to prepare the primary emulsion by ultrasonic equipment (120W, Digital Sonifier 450, Branson Ultrasonics Corp, USA). Then the emulsion was mixed with 90 ml 3% (w/v) PVA solution for membrane emulsification. Microporous tubular membranes with pore sizes of 5 μm, 8 μm, 12 μm, and 15 μm were used in the experiments. The membrane was wetted in the continuous phase under ultrasonic field before the installation. Membrane emulsifier (Senhui Microsphere Tech Co., Ltd., China) controls the emulsification pressure precisely and stably through the pressure sensor. The O/W emulsion was extruded through microporous membrane three times with a certain pressure under 250 rpm magnetic stirring, following pre-solidification by vertical suspension at 45 rpm for 30 min and solidification in 500 mL deionized water by magnetic stirring at 200 rpm for 10 min. The obtained microspheres were collected by centrifugation, following three washes with deionized water. The R-CNP were mixed with microspheres and incubated at 4 oC overnight, followed by 2 h of healing at approximately 40 oC using incubator (DGX-9143B-1, Shanghai Fuma Test Equipment Co., Ltd, China).
Characterization of microcapsules
The filled microcapsules came freeze-dried by freeze dryers (Alpha 2-4 LDplus, Martin Christ Gefriertrocknungsanlagen GmbH, Germany). The particle sizes of microsphere and microcapsule were analyzed by laser diffraction using Mastersizer (Mastersizer 2000, Malvern Instruments Ltd, UK). The aerodynamic particle diameter of lyophilized microcapsule was determined by the Aerodynamic Particle Sizer (TSI 3321, TSI Incorporated, USA). The shape and surface morphology of microsphere, microcapsule, and freeze-dried powder was observed using scanning electron microscopy (JSM-6700, JEOL, Japan). To obtain the distribution of nanoparticles in the cavities of the microcapsule, nanoparticles and microcapsules were labelled with FITC-SE and Nile red and observed by using confocal laser scanning microscopy (A1/SIM/STORM, Nikon, Japan). Protein concentrations of supernatant after encapsulation were determined by Micro BCA Protein Assay Kit (PI23235, Thermo Scientific) following the corresponding protocols for evaluation of loading capacity (mass of R-CNP in microcapsule/mass of microcapsules × 100%) and encapsulating capacity (mass of R-CNP in microcapsule/mass of total R-CNP × 100%).
In vivo antigen release
To evaluate the duration of nanoparticles with/without microcapsules in vivo, nanoparticle was labeled with near-IR water-soluble fluorescence probe Cy7-NHS ester (Cy7-SE), and microcapsule was labeled with hydrophobic Cy5 by mixing into the framework during primary emulsion. The inhaled vaccines containing Cy7-nanoparticles and Cy5-microcapsules were delivered into the lung using a customized dry powder inhaler for mice (DPI, Beijing Huironghe Technology). At different time intervals, the mice were captured using an In Vivo Imaging System (IVIS) optical imaging system (Caliper Life Sciences Inc., PerkinElmer, USA), and euthanized to extract organs following to capture again at the end of this experiment.
Immunofluorescence analysis
The distribution of R-CNP@M in lung
The animals were vaccinated with Nile Red-labeled R-CNP@M. The lungs were fixed with 4% paraformaldehyde and then dehydrated with 30% sucrose solution for subsequent sectioning on day 5 post-inhalation. Then 10 µm lung slides were prepared by freezing microtome (CM1950, Leica, Germany). Slides were mounted for imaging by the Vectra platform (v 3.0.5, PerkinElmer, USA) and counted by inform software.
GC production of monkey mLNs
The mLNs of the cynomolgus monkeys were harvested on day 14 after immunization, the mLNs were fixed with 4% paraformaldehyde and then embedded in paraffin blocks for serial sectioning. Then 5 µm mLN slides were prepared for immunofluorescence staining. Microwave treatment was conducted with ethylenediaminetetraacetic acid (EDTA) antigen retrieval buffer to retrieve antigen, followed by blocking with 5% (w/v) bovine serum albumin for 30 min. The sections were stained with anti-B220 (mouse), and anti-Ki67 (rat) at 4℃ overnight. The corresponding fluorescent secondary antibodies were incubated for 1 hour at room temperature. Counterstain was done using DAPI, and slides were mounted for imaging by the Vectra platform.
Light-sheet imaging
To verify whether microcapsules uniformly distribute over the whole lung, lung trachea and microcapsules were labeled with streptavidin-AF647 and Nile red, respectively. The whole lung was processed with Tissue-Clearing Reagent and captured by light-sheet microscopy (Z.1, Zeiss, Germany). Image reconstructions were generated using Imaris v9.0 software.
To get a visualization of germinal center in mLNs, B cells were labelled with B220-AF647. The mLNs were profile by CD31-FITC antibody. The mLNs were processed with Tissue-Clearing Reagent and captured in CUBIC-R+ reagent by light-sheet microscopy (Light Innovation Technology Limited.). Laser power and gain were adjusted consistently for each group. Image reconstructions were generated using LitScan v3.0 software.
Immunization
Dry powder inhaler
To simulate the process of human dry powder inhalation, we developed a dry powder inhaler (DPI, Supplementary Figure 2), which comprised the dry powder aerosol generator (DPAG) (Beijing Huironghe Technology Co., Ltd.), a cushion chamber and an endotracheal tube. To ensure that the dry powder aerosol is inhaled into the lungs through the mouth and trachea, the cushion chamber was designed with one end connected to an endotracheal tube and the other end connected to the DPAG. The volumes of the cushion chamber are distinguished for different animal species (about 35 mL, 80 mL, and 700 mL for mouse, hamster, and monkey, respectively). During immunization, the endotracheal tube of DPI is inserted into the animal's trachea, and a needle free syringe is used as the gas supply source to inject the dry powder vaccine in DPAG into the cushion chamber with the form of dry powder aerosol. The animal is separated from the DPI device after continuously inhaling the dry powder vaccine for 2 minutes. Note that mouse and hamster can be assisted with a small animal ventilator to assist when using the DPI device.
Immunization for mice
For long-term immunological evaluation, six female BALB/c mice (6-8-weeks old) were immunized twice with R-CNP, which contain 5 μg equivalent SARS-CoV-2 RBD protein intratracheally through a customized liquid aerosol delivery device for mice (Beijing Huironghe Technology); the second dose was the same as the first dose and given on day 14 post-prime vaccination. Six female BALB/c mice (6-8-weeks old) were immunized once with R-CNP@M, which contain 10 μg equivalent SARS-CoV-2 RBD protein intratracheally through DPI for mice. Six female BALB/c mice (6-8-weeks old) were immunized twice with CNP as control.
For the evaluation of mucosal response, another batch of mice were immunized with CNP, R-CNP and R-CNP@M as above described. Six female BALB/c mice (6-8-weeks old) were immunized with 5 μg SARS-CoV-2 RBD protein twice, combined with 100 μg aluminum adjuvant (RBD+Al) through intramuscular route as control, and the second dose was the same as the first dose and given on day 14 post-prime vaccination. On day 70 post-immunization, these mice were sacrificed for the harvest of BALF and serum. The obtained samples were subsequently inactivated at 56 ℃ for pseudovirus neutralization assay.
For the live SARS-CoV-2 challenge, another batch of mice were vaccinated with PBS, RBD+Al and R-CNP@M as above described.
Immunization for Syrian golden hamsters
Six-to-seven weeks old female Syrian Golden hamsters were immunized once with R-CNP@M, which contain 30 μg equivalent SARS-CoV-2 RBD protein intratracheally through DPI. Hamsters were intramuscularly immunized three times with licensed recombinant protein subunit vaccine Zifivax, which has been approved for use in China, Colombia, Indonesia, and Uzbekistan. The formulation contained 10 μg equivalent of SARS-CoV-2 dimer RBD protein. Blood samples were taken every 14 days, and BALF were collected on the 42nd day. The obtained samples were then subjected to ELISA and live virus neutralization tests. For the live SARS-CoV-2 challenge, another batch of hamsters were vaccinated with PBS, Zifivax and R-CNP@M as above described.
To evaluate the effectiveness of iterative vaccines, six-to-seven weeks old female Syrian Golden hamsters were immunized once with iterative vaccine (RWRO-CNP@M), which contain 30 μg equivalent SARS-CoV-2 RBD protein intratracheally through DPI. Hamsters were immunized twice with RWRO+Al, which contains 15 μg equivalent SARS-CoV-2 RBD protein intramuscularly. Blood samples and BALF were collected on the 42nd day. For the live SARS-CoV-2 challenge, another batch of hamsters were vaccinated with PBS, RWRO+Al and RWRO-CNP@M as above described.
Immunization for cynomolgus monkey
A total of 10 cynomolgus monkeys (female, 4-5-years old) were divided into PBS group and R-CNP@M group. All five animals were immunized once with R-CNP@M, which contain 50 μg equivalent SARS-CoV-2 RBD protein through DPI. Blood was collected every 2 weeks. The BALF was collected with assistance of bronchoscope at day 56.
Pulmonary single-cell RNA sequencing
The female BALB/c mice (6-8-weeks old) were immunized with CNP, R-CNP or R-CNP@M as described above, and mice were sacrificed on day 21 post-prime vaccination. The lungs were harvested and minced with scissors, following treated with collagenase I (1 mg/mL) and DNase I (0.05 mg/mL) and dissociated to a single-cell suspension. After being treated with red blood cell lysing reagent, the single-cell suspension was passed through a 40 μm strainers and resuspended in PBS. The single-cell suspension of four mice in each group was pooled. The CD45+ cells were isolated from single-cell suspension in each group by CD45 MicroBeads, and then these CD45+ single-cell suspensions containing about 10000 cells in each group were loaded on a 10X Genomics GemCode Single-cell instrument to generate single-cell Gel Bead-In-EMlusion (GEMs). Libraries were generated and sequenced from the cDNAs with Chromium Next GEM Single Cell 3’ Reagent Kits v3.1. The Gene Denovo Biotechnology Co. were entrusted to perform GEMs generation, cDNA amplification, library preparation, and sequencing in this study. Overall, 10156 cells from the PBS group, 6400 cells from the CNP group, 8400 cells from the R-CNP group, and 11753 cells from the R-CNP@M group passed the quality control threshold of more than 200 genes were identified in each cell.
BCR repertoires sequencing
RNA samples were obtained from pulmonary lymphocytes by cell sorting and subsequently analyzed by High-throughput sequencing of IGH using the ImmuHub® BCR profiling system at a deep level (ImmuQuad Biotech, Hangzhou China). Briefly, a 5’ RACE unbiased amplification protocol was used. This protocol uses unique molecular barcodes (UMBs) introduced in the course of cDNA synthesis to control bottlenecks and to eliminate PCR and sequencing errors. Sequencing was performed on an Illumina NovSeq® system with PE150 mode (Illumina). One common adaptor with UMB was added on the 5’ of cDNA during the first-strand cDNA synthesize and one reverse primer corresponding to the constant (C) regions of each of the IGH were designed to facilitate PCR amplification of cDNA sequences in a less biased manner. The UMB attached to each raw sequence reads were applied to correct PCR and sequencing errors correction and PCR duplicates removing. Map V, D, J and C segments with IMGT® and then extract CDR3 regions and assemble clonotype for all clones. The resulting nucleotide and amino acid sequences of CDR3 of IGH were determined and those with out-of-frame and stop codon sequences were removed from the identified IGH repertoire. We further defined amounts of each IGH clonotype by adding numbers of IGH clones sharing the same nucleotide sequence of CDR3.
Flow cytometry analysis
In vitro Activation of APC
To evaluate the activation of primary DCs and macrophages resulting from RBD, the mixture of RBD and CTB, and R-CNP, BMDCs and peritoneal macrophages were plated at 1.0 × 105 cells/well in a 24-well plate respectively, and then stimulated with RBD, CTB+RBD, and R-CNP for 24 h. Non-stimulated BMDCs and peritoneal macrophages were used as a negative control. Cells were collected and stained with anti-CD11c-FITC, anti-CD40-PE, anti-CD86-PE-Cy7, anti-MHC I-Percp-cy 5.5, and anti-MHC II-Alexa Fluor 700 at 4 °C for 30 min. After washing, the cells were resuspended in 300 μL of staining buffer and analyzed using a CytoFLEX LX flow cytometer (Beckman Coulter, USA). The cell culture supernatants of BMDCs and peritoneal macrophages were collected to assess the levels of IL-6, TNF-α, IL-12, IFN-γ, and MCP-1 by a CBA Mouse Inflammation Kit.
Analysis of antigen uptake by APC in lung
To investigate antigen uptake by APC in lung, flow cytometry was executed. After 10 days post-vaccination with microcapsule vaccine (Cy5-labeled R-CNP), single-cell suspensions from the lungs were prepared by mechanical disruption and isolated from red cells using red blood cell (RBC) lysis buffer, following centrifugation and resuspension with staining buffer. A part of the collected cells was stained with anti-CD11c-AF700, anti-CD64-BV421, anti-MERTK-PE and anti-CD11b-BV510 to define DCs/macrophages, and other cells were stained with anti-CD3-BV605 and anti-CD19-APC-Cy7 to define T cells/B cells. The proportion of APC that internalized antigen (Cy5+F4/80+ or Cy5+CD11c+) was measured using CytoFLEX LX flow cytometer (Beckman Coulter, USA), and analyzed using CytExpert software (version 2.3).
Analysis of activated APC in mLNs
To discover the activation of homed APC in mLNs, flow cytometry was executed. After 21 days post-vaccination, single-cell suspensions from the mLN were prepared by mechanical disruption and isolated from red cells using red blood cell (RBC) lysis buffer, following centrifugation and resuspension with staining buffer. The collected cells were stained with anti-CD11c-BV650 and anti-F4/80-FITC to define DCs/macrophages, and anti-CD86-PE-Cy7 as activation markers. The proportion of activated APC was measured using CytoFLEX LX flow cytometer (Beckman Coulter, USA), and analyzed using CytExpert software (version 2.3).
Analysis of T cells and B cells in mice
To evaluate T cell responses in lung, mLNs, NALT and nose of mice, flow cytometry was executed (Supplementary Figure 3). Mice were sacrificed and the tissues were harvested. Then tissues were mechanically ground to prepare single-cell suspensions and divided extra several parts. The following antibodies were used to detect GC B cells and CD4+ T cells in mLNs: anti-CD3-FITC, anti-CD4-PE-Cy7, anti-CD19-APC-Cy7, anti-GL7-AF647, anti-IgD-BV421. To evaluate memory T cell responses, tissue resident T cell responses and multifunctional T cell responses, cells were stained as follows. (1) The following antibodies were used to detect TRM CD8+ T cell in lung: anti-CD3-BV605, anti-CD4-PE-Cy7, anti-CD8-APC-Cy7, anti-CD44-PE, anti-CD69-APC, anti-CD103-AF700, anti-CD11a-FITC, and anti-IFN-γ-PE antibodies (2) The following antibodies were used to detect IFN-γ+ cells in NALT and nose: to anti-CD3-FITC, anti-CD4-PE-Cy7, anti-CD8-BV605, anti-IFN-γ-PE antibodies. (3) The following antibodies were used to detect multi-functionality T cell in spleen and lung: anti-CD3-FITC, anti-CD8-APC-Cy7, anti-IFN-γ-PE, anti-TNF-α-BV510, anti-IL2-PE-Cy5, (4) The following antibodies were used to detect TEM in spleen: anti-CD3-BV605, anti-CD4-PE-Cy7. anti-CD8-APC-Cy7, anti-CD44-FITC, anti-CD62L-APC antibodies. The stained cells were washed with PBS and measured using CytoFLEX LX flow cytometer, and analyzed using CytExpert software.
Analysis of T cells in monkey
To evaluate T cell responses in monkeys, flow cytometry was executed. First, about 1 mL of peripheral blood was taken from each monkey on day 21, and the lymphocytes were separated using Monkey blood lymphocyte separation Kit. Then the cells were stained with anti-CD8-Alexa Fluor 700 for 30 min at 4°C. Then the cells were washed twice and fixated with a fixative solution for 30 min, centrifugation to collect cells. Permeabilization reagent was then added to suspend the cells and incubated with anti-Granzyme B-Pacific Blue for 30 min at 4°C. The stained cells were washed with PBS and analyzed by flow cytometry (CytoFLEX, Beckman Coulter, USA).
IFNγ ELIspot assay
The assay was performed according to the manufacturer’s instructions (Mabtech, Sweden). Briefly, standard 96-well plates were incubated with anti-mouse IFN-γ antibody with a final concentration of 15 μg/mL overnight at 4 ℃. On day 21 post-vaccination, mice were sacrificed and the spleen was collected to prepare a single-cell suspension. After red-cell lysis and cell counts, these splenocytes were plated at 5 × 105 per well, in triplicate, with peptide at 5 μM. After overnight restimulation at 37°C in a cell incubator, plates were washed with cold ultrapure water and incubated with biotinylated anti-mouse IFN-γ antibody for 2 hours at room temperature. Then the plates were washed with PBST five times and incubated with the streptavidin-ALP conjugated antibody for 1 hour. After extensive washing, 100 μL of substrate (bromochloroindolyl phosphate-1-nitro blue tetrazolium step solution, Pierce, USA) was added into each well of plates for the coloration of IFN-γ spots. The plates were thoroughly washed with plenty of ultrapure water to stop the reaction. After complete air-dry of these plates, spots were counted using an automated ELISpot reader (AT-Spot 2100, Antai Yongxin Medical Technology, China).
Enzyme-linked immunosorbent assay (ELISA)
Binding properties of serum and BALF from BALB/c mice, hamsters and monkey to SARS-CoV-2 RBD protein were determined by ELISA. 96-well plates (Corning) were coated with 2 μg/mL of RBD protein (without his tag, 40592-VNAH, from Sino Biological, China) overnight at 4 ℃ in coating buffer, and blocked in 5% bovine serum albumin in PBS for 2 hours at 37℃. Serially diluted serum or BALF samples were added to the ELISA plates and incubated for 1 hour at 37℃. The plates were washed five times with 1× PBS containing 0.05% Tween-20 (PBST), followed by addition of 100 µL of HRP-conjugated goat anti-mouse IgM, goat anti-mouse IgG, goat anti-mouse IgG1, goat anti-mouse IgG2a, goat anti-mouse IgA, goat anti-hamster IgG, goat anti-monkey IgG secondary antibody with a 1:10000 dilution and goat anti-monkey IgA (4A Biotech) secondary antibody with a 1:5000 dilution. After being incubated at 37℃ for 0.5 hours, plates were again washed five times with PBST. Then the plates were developed with 3,3′,5,5′-tetramethytlbenzidine for 10 min at room temperature, and the reaction was stopped with 2 M H2SO4. The absorbance at 450 nm was measured by a microplate reader. The endpoint titer was defined as the highest reciprocal dilution of detected sample to give an absorbance greater than 2.1-fold of the OD450 value from the PBS group.
Pseudovirus neutralization assay
The SARS-CoV-2 pseudotyped viruses (for Figure 4), which were expressing a luciferase reporter gene, were gifted from Professor Youchun Wang of Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China. The SARS-CoV-2 pseudotyped viruses based neutralization assay was performed as described previously 48,49. The pseudotyped virus used in this study was developed based on vesicular stomatitis virus (VSV). All samples were heat-inactivated to eliminate any complement activity. In brief, 293T-hACE2 cells were seeded in 96-well plates (20,000 cells/well) and incubated with 200 TCID50 (50% tissue culture infective dose) SARS-CoV-2 pseudotyped viruses, which premixed with serial dilutions of the serum (starting at 16×, 3-fold serial dilutions) or BALF (starting at 3×, 3-fold serial dilutions) for 1 hour. After 24 hours of incubation, the luminescence of each well was detected. The sample pVNT50 was calculated using the Reed-Muench method.
The SARS-CoV-2 pseudotyped viruses (for Figure 6 and 7), which were expressing green fluorescent protein (GFP) gene, were gifted from Professor Xin Zhao of Institute of Microbiology, Chinese Academy of Sciences, Beijing, China. All samples were heat-inactivated to eliminate any complement activity. In brief, Vero cells were seeded in 96-well plates (20,000 cells/well) and incubated with SARS-CoV-2 pseudovirus-GFP for 1000 IU/well, which premixed with serial dilutions of the serum (starting at 16×, 3-fold serial dilutions) or BALF (starting at 3×, 3-fold serial dilutions) for 1 hour. After 24 hours of incubation, the counts of positive cells in each well were detected by high content analysis (Operetta CLS, Perkin Elmer). The sample pVNT50 was calculated using the Reed-Muench method.
Live virus neutralization assay
This study was performed as described previously in a biosafety level 3 (BSL3) laboratory following institutional biosafety guidelines by the Sinovac Life Sciences Co., Ltd. All samples were heat-inactivated to eliminate any complement activity. In brief, the serial dilutions of the serum (starting at 10×, 2-fold serial dilutions) or BALF (starting at 2×, 2-fold serial dilutions) were mixed with a SARS-CoV-2 virus suspension (SARS-CoV-2/human/CHN/CN1/2020, GenBank number MT407649.1 and SARS-CoV-2 Delta variants, GISAID number Delta EPI_ISL_1911197) of 100 CCID50 in 96-well plates, followed by 2h incubation at 36.5 °C in a 5% CO2 incubator. Then the Vero cell were added into the 96-well plates at 36.5°C in 5% CO2 for 5 days. Cytopathic effect (CPE) of each well was recorded under microscopes, and the neutralizing titer was the reciprocal of the highest serum dilution with 50% cytopathic effect. Geometric mean titer (GMT) was calculated based on these data.
In vivo cell depletion
Depletion of CD8+ T cells was achieved by intraperitoneal (i.p.) injection of 300 µg/mouse YST-169.4 (anti-CD8a) monoclonal antibodies (YTS 169.4, BE0117, BioXcell, West Lebanon, NH), at day 6 before vaccination with R-CNP@M and every 3 days until SARS-COV-2 challenging. For B cell depletion, 200 µg/mouse anti-CD19 (1D3, BE0150, BioXcell) and 200 µg/mouse anti-B220 (RA3.3A1/6.1, BE0067, BioXcell) were injected i.p. at day 6 before vaccination with R-CNP@M and every 3 days until SARS-COV-2 challenging. A mixture of isotype antibodies (2A3, BE0089; LTF-2, BE0090; BE0094.) were injected i.p. at day 6 before administration with PBS and every 3 days until SARS-COV-2 challenging.
Live SARS-CoV-2 virus challenge
The preimmunized K18-hACE2 mice were challenged with 1×103 TCID50 of wild-type strain (SARS-CoV-2-KMS1/2020 /GenBank accession number: MT226610.1) via the intranasal route at day 28. The body weight and number of deaths of mice was recorded every day post infection (dpi), and after being euthanized on 4 dpi, the lung and nose tissues were harvested for virus RNA levels (about 0.1-gram lung or nose tissues) determination and pathological examination (the left lung).
The Syrian Golden hamsters were challenged with 2×104 TCID50 of wild-type strain (SARS-CoV-2-KMS1/2020 /GenBank accession number: MT226610.1) via the intranasal route at day 42. Hamsters were euthanized and necropsied at day 3 post-challenge. The lung and nose tissues were harvested for virus RNA levels (about 0.2-gram lung) determination, viral titer assessment and pathological examination.
Airborne transmission protection was examined by housing hamsters in specially designed cages with a cage divider dividing the living space in half. The donor animal was inoculated with 1×105 TCID50 of the live Omicron strain (CCPM-B-V-049-2112-18) via the intranasal route and was introduced into one side of the cages at 24 h after infection. Three recipients, which were respectively vaccinated with PBS (mock group), mixed antigen via intramuscular injection and mosaic vaccine via inhalation, were placed on another side of the divider downflow from one infected hamster. Hamsters were sacrificed 1- and 3-days post-exposure, and lung tissues were obtained for viral load and pathological sections.
As to contact/airborne transmission protection, one donor and three recipients were placed in a cage without any barrier, sharing the diet, bedding, and airiness. Donor animals were inoculated with 1×105 TCID50 of the live Omicron strain (CCPM-B-V-049-2112-18) by intranasal route, and 24 h after infection, they were raised in the same cage with three recipients for 24 h, and then sacrificed on the 1st and 3rd day to collect lung tissues.
In airborne transmission blocking model, the cages used were the same as those used in the airborne transmission protection model. One of the donor hamsters (given PBS, inhaled vaccine, or intramuscular vaccine) was inoculated with 2×104 TCID50 of live Omicron strain (CCPM-B-V-049-2112-18) via the intranasal route, and was introduced into one side of the cage divider at 24 h after infection. Two recipients (naïve hamsters) were placed on the other side of the divider (downflow from the infected hamster). After 24 h of exposure to the donor, the recipient hamsters were taken out and raised in a clean individually ventilated cage (IVC). The recipient hamsters were sacrificed at 72 h post exposure (not including the initial 24 hours of exposure), and the viral loads of their nose homogenates were used for further analysis. Note that all of hamsters were survived until were sacrificed post challenged with SARS-CoV-2.
RT-qPCR
SARS-CoV-2 viral RNA in lung and nose tissues from challenged mice and hamsters were detected by quantitative reverse transcription PCR (RT-qPCR). Briefly, the lung and nose tissues were weighed and homogenized with Trizol, and virus RNA could be isolated according to the manufacturer’s protocol. SARS-CoV-2 viral RNA levels were measured by One Step PrimeScript RT-PCR Kit (RR064B, Takara) on a CFX96 real-time PCR detection system (Bio-Rad), and one sets of primers and probes were used to detect a region of the ORF1a/b of the viral genome of SARS-CoV-2, with sequences as follows: ORF1a/b-F, CCCTGTGGGTTTTACACTTAA; ORF1a/b-R, ACGATTGTGCATCAGCTGA; ORF1a/b-probe, FAM-CCGTCTGCGGTATGTGGAAAGGTTATGG-BHQ1. Viral RNA levels in lung and nose tissues were expressed as ORF1a/b gene copy numbers per milligram after comparison with a standard curve produced using serial ten-fold dilutions of SARS-CoV-2 RNA.
Viral titration
Lung tissues from hamsters were weighed and homogenized with 1mL of DMEM medium (supplemented with 100 U/mL of penicillin and 100 μg/mL streptomycin solution). Virus titrations were performed by end point titration in Vero cells, which were inoculated with 10-fold serial dilutions of tissue homogenates in 96-well plates (100μL/well). The titrating tissue homogenate were added to 100 μL of Vero cell suspension (DMEM with 4% FBS, 200 U/mL of penicillin and 200 μg/mL streptomycin solution, 2×105 cells/mL) in each well, and then they were incubated at 37℃ with 5% CO2. Cytopathic effect was read 6 days later.
Immunohistochemical/histopathology analysis
The lung tissues were fixed for more than 48 hours in 4% paraformaldehyde, dehydrated, and embedded in paraffin. 5-μm-thick tissue sections were sectioned and stained with hematoxylin and eosin (H&E). Double-blind evaluations were made for two randomly selected sections of each hamster on the basis of the histopathological changes, including inflammation, structure change and hemorrhage, which were graded according to the pre-defined scoring system.
Briefly, the clear structure of alveolar without inflammatory infiltration was recorded as score 0. The mild inflammation, slightly widened alveolar septum and sparse mononuclear cells (infiltration). The severe inflammation, thickening of alveolar wall and increased infiltration of interstitial monocytes was recorded as score 2. The significantly widened alveolar septum and increased infiltration of inflammatory cells was recorded as score 3-4. The extensive exudation and widened septum, smaller alveolar cavity, septal bleeding, and alveolar cells infiltration was recorded as score 5.
The immunohistochemical reaction in this study was performed on paraffin-embedded lung sections using a rabbit anti-SARS-CoV-2-Nucleocapsid Protein monoclonal antibody followed by an HRP-conjugated goat anti-rabbit secondary antibody. Then the DAB Substrate Kit was used for the chromogenic reaction. After nuclear counterstaining, dehydration, and mounting, the tissue staining was visualized under a microscope to detect virus infection.
Quantification and statistical analysis
Cell type annotation, relative proportion analysis of each cell subsets, differentially expressed genes analysis, Kyoto Encyclopedia of Genes and Genomes pathway and Gene Ontology enrichment analysis were performed on Omicsmart (https://www.omicsmart.com/home.html#/) where the datasets were analyzed and processed based on the R 3.6.1 Seurat package.
To show the multivariate antibody profiles across vaccination groups (RW-CNP@M, RO-CNP@M, or RWRO-CNP@M), principal component analysis (PCA) were performed and the heatmap were plotted based on corresponding Z score of ten immunological indices (the letters represent different biological meanings as follows: W, RBDWild-specific/wild-type strain; O, RBDOmicron-specific/wild-type stain; B, BALF; S, Serum; P, Pseudovirus neutralization assay; A, IgA titer; G: IgG titer).
For other data, GraphPad Prism 9.0.0 and Origin 2023 were used for plotting and statistical analysis; the values were expressed as means ± SEM. Significance were calculated using unpaired t test and one-way ANOVA with Dunnett’s multiple comparison test or Tukey's test when the data conformed to a normal distribution and exhibited homogeneity of variance. Otherwise, the Mann-Whitney tests and Kruskal‑Wallis test was performed. The p values and R2 values in correlation analysis reflect Spearman rank-correlation tests.
Materials & Correspondence
Further information and requests for resources and reagents should be directed to and will be fulfilled by Prof. Wei Wei ([email protected]). All unique reagents generated in this study are available from Prof. W. W. with a completed Materials Transfer Agreement. The published article contains all datasets analyzed during the study can be requested from Wei Wei upon reasonable request. The sequences data reported in this study were archived in the Sequence Read Archive (accession number: PRJNA813749, PRJNA1002859).
REFERENCES
48 Li, Q. et al. The Impact of Mutations in SARS-CoV-2 Spike on Viral Infectivity and Antigenicity. Cell 182, 1284-1294 e1289, doi:10.1016/j.cell.2020.07.012 (2020).
49 Nie, J. et al. Quantification of SARS-CoV-2 neutralizing antibody by a pseudotyped virus-based assay. Nat Protoc 15, 3699-3715, doi:10.1038/s41596-020-0394-5 (2020).