Ethics statement. This study was carried out in strict accordance with the recommendations described in the Guide for the Care and Use of Laboratory Animals of the Institute of Microbiology, Chinese Academy of Sciences (IMCAS) Ethic Committee. All animal experiments were reviewed and approved by the Committee on the Ethics of Animal Experiments of IMCAS.
Cells, viruses and animals. HEK293T cells (ATCC CRL-3216), Huh7 cells (3111C0001CCC000679) and Vero E6 cells (ATCC CRL-1586) were cultured at 37℃ in Dulbecco’s Modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS). BALB/c and C57BL/6 mice were purchased from Beijing Vital River Animal Technology Co., Ltd. (licensed by Charles River), and housed in specific-pathogen-free (SPF) mouse facilities in IMCAS. hACE2 transgenic mice were kindly provided by Professor Zhengli Shi from Wuhan Institute of Virology, CAS. SARS-CoV-2 virus strain HB-01 used in this study was provided by Professor Wenjie Tan from Chinese center for disease control and prevention. Vero E6 cells were applied to the amplification and titer titration of the virus stocks.
mRNA production. mRNA was produced using T7 RNA polymerase on linearized plasmids (synthesized by Genescript) encoding codon-optimized SARS-CoV-2 RBD glycoproteins (residues 319-541, GISAID: EPI_ISL_402119). mRNA was transcribed to contain 104 nucleotide-long poly(A) tail. 1-methylpseudourine-5’-triphosphate was used instead of UTP to generate modified nucleoside-containing mRNA. After transcription and purification, mRNA was analyzed by agrose gel electrophoresis and stored frozen at -20℃.
mRNA transfection. Transfection of HEK293T cells was performed with TransIT-mRNA (Mirus Bio) according to the manufacturer’s instructions. In briefly, mRNA (0.5 μg) was combined with TransIT-mRNA regent (1 μl) and boost regent (1 μl) in 50 μl of serum-free medium, and the complex was added to 2.5 × 105 cells in 500 μl complete medium. Supernatant was collected and concentrated, and cells were lysed on ice in RIPA buffer (Beyotime) at 48 h after transfection.
Western blot. Whole-cell lysates and supernatants from cells transfected with mRNA-RBD were assayed for SARS-CoV-2 RBD expression by western blot. Samples were combined with loading buffer with dithiothreitol (DTT) and separated by 12% SDS-PAGE. Transfer to PVDF membrane was performed using a semi-dry apparatus (Ellard Instrumentation). The membrane was blocked with non-fat milk in TBS buffer containing 0.5% Tween-20. RBD protein was detected using mouse serum immunized with SARS-CoV-2 S1 proteins (Sino Biological) for 1 h, followed by secondary goat anti-rabbit IgG HRP (Santa Cruz) for 1 h. The membrane was developed by SuperSignal West Pic chemiluminescent substrate (Thermo Fisher Scientific).
Lipid-nanoparticle encapsulation of mRNA. SARS-CoV-2 RBD-encoded mRNA (mRNA-RBD) was encapsulated in LNPs using a self-assembly process in which an aqueous solution of mRNA at pH=4.0 was rapidly mixed with a solution of lipids dissolved in ethanol. LNPs used in this study contained an ionizable cationic lipid, phosphatidylcholine, cholesterol and PEG-lipid with a ratio of 50:10:38.5:1.5 mol/mol and were encapsulated at an mRNA to lipid ratio of around 0.05 (wt/wt). mRNA-RBD LNPs were stored at 4℃ at a concentration of RNA of about 1 mg/ml.
Particle size, zeta potential and encapsulation efficiency. Zetasizer was used to determine the particle size and zeta potential. Zeta potential was measured on particles after suspending LNPs in deionized water at pH 4.0 and 7.4. The free and total mRNA concentrations in LNPs were determined using the Quant-iT Ribogreen RNA assay kit according to the manufacturer’s instructions. The encapsulation efficiency (EE, %) was calculated as follows: EE (%) = (1- free mRNA concentration /total mRNA concentration) ×100.
Cryo-electron microscopy of LNPs. LNPs were transferred onto a glow-discharged ultrathin carbon-coated copper grid (Zhongjingkeyi Company) followed by 60 s of waiting and blotted for 2 s with filter paper before plugging into liquid ethane using the Vitrobot Mark IV. The frozen grids were transferred at liquid nitrogen temperature and loaded into a Talos transmission electron microscope (Thermo Fisher Scientific) equipped with a field emission gun operated at 200 kV. The images were recorded on a direct electron detector (ED20) at a total electron dose of ~50 e−/Å2.
Animal experiments. LNPs-encapsulated mRNA-RBD was diluted with PBS. Female BALB/c or C57BL/6 mice aged 6-8 weeks were inoculated intramuscularly (i.m.) with a high (15 μg) or low (2μg) dose of mRNA-RBD LNP formulations or poly(C) LNP formulations as placebo control. Serum samples were collected at indicated time after vaccination, inactivated at 56°C for 30 min and stored at -20℃.
For SARS-CoV-2 challenge experiments, hACE2 transgenic mice (n=6) were immunized with one (prime group) or two (boost group) doses of mRNA-RBD via i.m. route. Four weeks post initial immunization, mice were infected with 1 × 105 FFU of SARS-CoV-2 (strain HB- 01) in a total volume of 50 μl of DMEM medium via intranasal (i.n.) route. Animals were monitored daily for survival and weight loss. The mice were enthanized 5 days following challenge. Lung tissues were harvested for virus load detection (four mice per group), and pathological and immunohistochemical examination (two mice per group).
For passive immunization and SARS-CoV-2 challenge experiments, groups of BALB/c mice (n=10) received 15 μg of mRNA-RBD or placebo. Half mice per group (n=5) were euthanized at 8 weeks (short term) post vaccination. The other mice per group (n=5) were euthanized at 26 weeks (long term) post vaccination. hACE2 transgenic mice (n=5) were administered 350 μl per mouse of pooled immune sera collected from placebo or mRNA-RBD vaccinated mice via i.p. route. One day following serum transfer, mice were challenged with 1× 105 FFU of SARS-CoV-2 via i.n. route. Animals were monitored daily for survival and weight loss. Lung tissues were harvested for virus load detection at 5 days following challenge. All animal experiments with SARS-CoV-2 challenge were conducted under animal biosafety level 3 (ABSL3) facilities in IMCAS.
Enzyme-linked immunosorbent assay (ELISA). ELISA plates (Corning) were coated overnight with 2 μg/ml of SARS-CoV-2 RBD, SARS-CoV RBD or MERS-CoV RBD recombinant protein in 0.05 M carbonate-bicarbonate buffer, pH 9.6, and blocked in 5% skim milk in PBS. Serum samples were 2-fold serially diluted and added to each well. Plates were incubated with goat anti-mouse IgG-HRP, IgG1-HRP or IgG2a-HRP antibodies and developed with 3, 3’, 5, 5’-tetramethylbenzidine (TMB) substrate. Reactions were stopped with 2 M hydrochloric acid, and the absorbance was measured at 450 nm using a microplate reader (PerkinElmer, USA). The endpoint titers were defined as the highest reciprocal dilution of serum to give an absorbance greater than 2.1-fold of the background values. Antibody titer below the limit of detection was determined as half the limit of detection.
Pseudovirus neutralization assay. SARS-CoV-2, SARS-CoV and MERS-CoV pseudovirus preparation and titration determination were performed as described previously 49. Briefly, the plasmids of 14 μg pCAGGS-SARS-CoV-2-S, pCAGGS-SARS-CoV-S or pCAGGS-MERS-CoV-S and 7 μg pNL4-3.luc.RE were cotransfected into HEK293T cells. After 48 h, the supernatant containing pseudovirus was harvested, centrifuged and filtered through a 0.45μm sterilized membrane. Single use aliquots were stored at -80℃. The TCID50 was determined by infection of Huh7 cells. For the neutralization assay, 100 TCID50/well was incubated with 2-fold serially diluted mouse sera for 30 min at 37℃. The mixtures were then used to infect Huh7 cells seeded in 96-well plates. After 5 h incubation, the medium was replaced with DMEM containing 10% FBS, and the samples were incubated for an additional 24 h at 37℃. Luciferase activity was measured using a GloMax 96 Microplate luminometer (Promega). The neutralization endpoint was defined as the fold-dilution of serum necessary for 90% inhibition of luciferase activity in comparison with virus control samples.
Live SARS-CoV-2 neutralization assay. The neutralizing activity of mice sera was assessed using a SARS-CoV-2 microneutralization assay. Briefly, heat-inactivated serum was 2-fold serially diluted and incubated with SARS-CoV-2 strain 01 (100TCID50) for 1 h at 37℃. The mixture was added to pre-seeded Vero E6 cell monolayers in 96-well plates. After incubation for 48 h at 37℃, the supernatant was collected for virus detection. The neutralization titers were defined as serum dilution required for 50% neutralization of viral infection.
Enzyme-linked immunosorbent spot (ELISPOT) assay. To detect antigen-specifific T lymphocyte responses, an IFN-γ-based ELISPOT assay was performed. Briefly, spleens were removed from vaccinated C57BL/6 mice at 4 weeks post immunization and splenocytes were isolated. Flat-bottom, 96-well plates were pre-coated with 10 g/ml anti-mouse IFNγ Ab (BD Biosciences, USA) overnight at 4°C and then blocked for 2 h at RT. Mouse splenocytes were added to the plate (1 × 105/well). Then, a peptide pool (2 μg/ml of individual peptide) consisting of 18-mers (overlapping by 10 amino acids) spanning the entire SARS-CoV-2 RBD proteins was added to the wells. Phytohemagglutinin (PHA) was added as a positive control. Cells incubated without stimulation were employed as a negative control. After 18 h of incubation, the cells were removed, and the plates were processed in turn with biotinylated IFNγ-detection antibody, streptavidin-HRP conjugate, and substrate. When the colored spots were intense enough to be visually observed, the development was stopped by thoroughly rinsing samples with deionized water. The numbers of the spots were determined using an automatic ELISPOT reader and image analysis software (Cellular Technology Ltd.).
Intracellular cytokine staining (ICS) assay. An ICS assay was performed to characterize antigen-specific CD4+ and CD8+ immune response. Briefly, mouse splenocytes were added to the plate (1 × 106 cells/well) and then stimulated with the peptide pool (2 μg/ml of individual peptide) for 4h. The cells were incubated with Golgiplug (BD Biosciences) for additional 14 h at 37℃. Then the cells were harvested and stained with anti-CD3, anti-CD4 and anti-CD8α surface makers (Biolegend). The cells were subsequently fixed and permeabilized in permeabilizing buffer (BD Biosciences) and stained with anti-IFN-γ (Biolegend). All fluorescent lymphocytes were gated on a FACSCanton flow cytometer (BD Biosciences).
Viral RNA extraction and RT-PCR. Viral RNA was extracted from 100 μl of samples using the automated nucleic acid extraction system (TIANLONG, China) and under the manufacturer’s instructions. Detection of the SARS-CoV-2 virus was performed using the One Step Prime Script RT-PCR kit (TaKaRa, Japan) on the Light Cycler 480 Real-Time PCR system (Roche, Rotkreuz, Switzerland) with primers. The following sequences were used:
forward primer: 5ʹ-AGAAGATTGGTTAGATGATGATAGT-3ʹ;
reverse primer:5ʹ-TTCCATCTCTAATTGAGGTTGAACC-3ʹ;
and probe:5ʹ-FAM-TCCTCACTGCCGTCTTGTTG ACCA-BHQ1-3ʹ.
Real-time RT-PCR was performed using the following conditions: 50°C for 15 min and 95°C for 3 min, 50 cycles of amplification at 95°C for 10 s and 60°C for 45 s. All experiments were conducted in triplicates.
Histopathology and immunohistochemistry. The lungs were fixed in 4% (v/v) paraformaldehyde solution for 72 hours, and the paraffin sections (3-4 μm) were prepared routinely. Hematoxylin and Eosin (H&E) stain was used to identify histopathological changes in the lungs. The histopathology of the lung tissue was observed by light microscopy. For immunohistochemistry, SARS-CoV-2 N protein was detected using monoclonal antibody clone 019 (Sino Biological, China). Images were captured using LEICA Versa 200 and were processed using software HALO v3.1.1076.379.
Statistical analysis. All data are expressed as the means ± standard errors of the means (SEM). For all analyses, P values were analyzed with one-way analysis of variance or Student’s t test. All graphs were generated with GraphPad Prism, version 7.0, software.