Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Nov 9:13:1023255.
doi: 10.3389/fimmu.2022.1023255. eCollection 2022.

Non-replicative antibiotic resistance-free DNA vaccine encoding S and N proteins induces full protection in mice against SARS-CoV-2

Affiliations

Non-replicative antibiotic resistance-free DNA vaccine encoding S and N proteins induces full protection in mice against SARS-CoV-2

Pedro J Alcolea et al. Front Immunol. .

Abstract

SARS-CoV-2 vaccines currently in use have contributed to controlling the COVID-19 pandemic. Notwithstanding, the high mutation rate, fundamentally in the spike glycoprotein (S), is causing the emergence of new variants. Solely utilizing this antigen is a drawback that may reduce the efficacy of these vaccines. Herein we present a DNA vaccine candidate that contains the genes encoding the S and the nucleocapsid (N) proteins implemented into the non-replicative mammalian expression plasmid vector, pPAL. This plasmid lacks antibiotic resistance genes and contains an alternative selectable marker for production. The S gene sequence was modified to avoid furin cleavage (Sfs). Potent humoral and cellular immune responses were observed in C57BL/6J mice vaccinated with pPAL-Sfs + pPAL-N following a prime/boost regimen by the intramuscular route applying in vivo electroporation. The immunogen fully protected K18-hACE2 mice against a lethal dose (105 PFU) of SARS-CoV-2. Viral replication was completely controlled in the lungs, brain, and heart of vaccinated mice. Therefore, pPAL-Sfs + pPAL-N is a promising DNA vaccine candidate for protection from COVID-19.

Keywords: DNA vaccine; N protein; S protein; SARS-CoV-2; furin; mouse model; pPAL.

PubMed Disclaimer

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Figures

Figure 1
Figure 1
The pPAL-Sfs + pPAL-N vaccine candidate. (A) pPAL-Sfs map. Sfs is a codon-optimized modified version of the SARS-CoV-2 Wuhan reference sequence that contains a modification to avoid furin cleavage of the protein product. Large-scale laboratory preparation of the pPAL-Sfs plasmid containing supercoiled (CCC), open-circular (OC), CCC-dimer, and CCC-trimer conformers. Western blot of whole protein extracts from HEK293 cells transfected with pPAL, pPAL-S, and pPAL-Sfs. The primary antibody was 1:800-diluted goat anti-S polyclonal antibody (Abcam #ab272504, Cambridge, United Kingdom), which only recognizes the S2 subunit. The secondary antibody was 1:2,000-diluted HRP-conjugated goat polyclonal anti-rabbit Ig. Furin cleaves the S protein obtained from pPAL-S but not Sfs from pPAL-Sfs. (B) Large-scale laboratory preparation of the pPAL-N plasmid containing CCC, OC, CCC-dimer, and CCC-trimer conformers. Western blot of whole protein extracts from HEK293 cells transfected with pPAL and pPAL-N. The primary antibody was 1:500-diluted rabbit anti-N polyclonal antibody. The secondary antibody was 1:2,000-diluted HRP-conjugated goat polyclonal anti-rabbit Ig. (C, D) immunofluorescence of cultured HEK293 cells transfected with pPAL, pPAL-Sfs, and pPAL-N plasmids. The anti-S and anti-N antibody dilutions were 1:50. The pPAL control was incubated with both antibodies. The secondary antibody was Alexa Fluor® 488-conjugated goat anti-rabbit IgG diluted to 1:200. Sfs and N expression is observed.
Figure 2
Figure 2
The pPAL-Sfs + pPAL-N vaccine candidates activate SARS-CoV-2 S- and RBD-specific humoral immunity in C57BL/6J mice. The serum samples were collected 15 days after the booster dose. (A) ELISA determinations of individual and average titers of total circulating anti-S and anti-RBD IgG determined by ELISA using HRP-conjugated protein (A) ELISA determinations of individual and average titers of circulating anti-RBD IgG, IgG1, and IgG2c using HRP-conjugated goat anti-mouse IgG1, anti-IgG2, and anti-IgG2c. The average anti-S and anti-RBD IgG titers are ≥2,000. The IgG2c/IgG1 ratio is ~750 in vaccinated animals with respect to the pPAL control group. (B) The neutralizing antibody titer average is >100. Statistical inference was performed using the Student’s t-test applying p-value adjustment by the Holm-Sidak method (α = 0.05; *p<0.05; **p<0.01; ***p<0.001; ns, non-significant).
Figure 3
Figure 3
S- and N-specific activation of IFN-γ-producing splenocytes induced by the pPAL-Sfs + pPAL-N vaccine candidate in C57BL/6J mice is dose-dependent. Splenocytes were obtained 7 days post-boost after prime/boost inoculation of pPAL and pPAL-Sfs + pPAL-N. Splenocytes (2x105 cells/well) were stimulated at 37 °C in IFN-γ ELISpot plates for 16 h with 11-mer 8 amino acid-overlap peptide pools (25 µg/well) representing the whole S and N amino acid sequences in equimolar amounts. Negative stimulation controls were subtracted from the SFC per million cell values. Statistical inference was performed using the Student’s t-test applying p-value adjustment by the Holm-Sidak method (α = 0.05; **p<0.01; ***p<0.001). (A) Robust S- and N-specific activation of IFN-γ-producing splenocytes is observed with a prime/boost immunization regime with pPAL-Sfs + pPAL-N. (B) S-specific activation of IFN-γ-producing splenocytes with pPAL-Sfs + pPAL-N is dose dependent.
Figure 4
Figure 4
Specific CD3+CD4+ and CD3+CD8+ T-cell response induced by pPAL-Sfs + pPAL-N in C57BL/6J mice. Splenocytes were obtained from mice after prime/boost inoculation with pPAL and pPAL-Sfs + pPAL-N. Splenocytes were stimulated for 4 h with 11-mer 8 amino acid-overlap peptide pools representing the whole S and N amino acid sequences in equimolar amounts. (A) Percentage of CD3+CD4+ and CD3+CD8+ T cells producing CD107a, IFN-γ, TNF-α, and IL-2 in response to stimulation with peptide S or N pools. (B) Percentage of CD4+ and CD8+ T cells responding to S and N peptide pools above background, calculated as the sum of T cells positive for CD107a and/or IFN-γ and/or TNF-α and/or IL-2. Statistical inference using Two-way ANOVA with Fisher’s LD post-hoc test (*p<0.05; **p<0.01; ***p<0.001).
Figure 5
Figure 5
Polyfunctionality analysis of specific CD3+CD4+ and CD3+CD8+ T cell populations stimulated by pPAL-Sfs + pPAL-N immunization in C57BL/6J mice. Analyses of concomitant expression of CD107a, IFN-γ, TNF-α, and IL-2 in CD3+CD4+ T cells and CD3+CD8+ T cells in response to N peptide pool or S peptide pool stimulation. Pie charts split the percentage of T cells expressing either 1, 2, 3, or 4 markers upon stimulation. Statistical inference using Two-way ANOVA with Fisher’s LD post-hoc test (*p<0.05; **p<0.01; ***p<0.001).
Figure 6
Figure 6
The pPAL-Sfs + pPAL-N vaccine confers full protection against challenge with 105 PFU of the MAD6 SARS-CoV-2 isolate in the K18-hACE2 murine model. (A) Titration of circulating anti-RBD IgG, IgG1, IgG2c, and neutralizing antibodies 15 days after prime/boost immunization with pPAL-Sfs + pPAL-N. (B) Body weight evolution after 105 PFU SARS-CoV-2 challenge. Vaccinated animals maintained their weight throughout the experiment. Weight decreased in the control group until the endpoint criteria had to be applied (20% of weight loss). (C) Clinical sign follow-up after challenge. (D) Reduction of the viral load in target organs. ΔCt accounting for SARS-CoV-2 mRNA levels. β-actin was the reference gene. (E) Reduction of viral replication (PFU/g tissue) in VERO cells. Statistical inference was performed using the Student’s t-test applying p-value adjustment by the Holm-Sidak method (α = 0.05; *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001).
Figure 7
Figure 7
Full protection of K18-hACE2 mice after three months of vaccination with pPAL-Sfs+ pPAL-N against challenge with 105 PFU of the MAD6 SARS-CoV-2 isolate in the K18-hACE2 murine model. (A) Titration of circulating anti-RBD IgG, IgG1, and IgG2c three months after the pPAL-Sfs + pPAL-N booster dose administration. (B) Body weight evolution after 105 PFU SARS-CoV-2 challenge. Vaccinated animals maintained their weight throughout the experiment, whereas weight decreased in the control group. (C) Clinical sign follow-up after challenge with 105 PFU SARS-CoV-2. Statistical inference was performed using the Student’s t-test applying p-value adjustment by the Holm-Sidak method (α = 0.05; *p<0.05; **p<0.01).
Figure 8
Figure 8
The pPAL-Sfs + pPAL-N vaccine confers full protection against challenge with 105 PFU of the SARS-CoV-2 Delta variant in the K18-hACE2 murine model. (A) Titration of circulating anti-RBD IgG, IgG2c, and neutralizing antibodies 15 days after prime/boost immunization with pPAL-Sfs + pPAL-N. (B) Body weight evolution after 105 PFU SARS-CoV-2 challenge. Vaccinated animals maintained their weight throughout the experiment, whereas weight decreased in the control group. (C) Clinical sign follow-up after challenge with 105 PFU SARS-CoV-2. Statistical inference was performed using the Student’s t-test applying p-value adjustment by the Holm-Sidak method (α = 0.05; *p<0.05; ****p<0.0001).

References

    1. Richner JM, Himansu S, Dowd KA, Butler SL, Salazar V, Fox JM, et al. . Modified mRNA vaccines protect against zika virus infection. Cell (2017) 169(1):176. doi: 10.1016/j.cell.2017.02.017 - DOI - PubMed
    1. Pardi N, Hogan MJ, Pelc RS, Muramatsu H, Andersen H, DeMaso CR, et al. . Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature (2017) 543(7644):248–51. doi: 10.1038/nature21428 - DOI - PMC - PubMed
    1. Reynolds CJ, Gibbons JM, Pade C, Lin KM, Sandoval DM, Pieper F, et al. . Heterologous infection and vaccination shapes immunity against SARS-CoV-2 variants. Science (2022) 375(6577):183–92. doi: 10.1126/science.abm0811 - DOI - PMC - PubMed
    1. Muik A, Lui BG, Wallisch AK, Bacher M, Muhl J, Reinholz J, et al. . Neutralization of SARS-CoV-2 omicron by BNT162b2 mRNA vaccine-elicited human sera. Science (2022) 375(6581):678–80. doi: 10.1126/science.abn7591 - DOI - PMC - PubMed
    1. Mannar D, Saville JW, Zhu X, Srivastava SS, Berezuk AM, Tuttle KS, et al. . SARS-CoV-2 omicron variant: Antibody evasion and cryo-EM structure of spike protein-ACE2 complex. Science (2022) 375(6582):760–4. doi: 10.1126/science.abn7760 - DOI - PMC - PubMed

Publication types