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
. 2020 Nov 23;94(24):e01176-20.
doi: 10.1128/JVI.01176-20. Print 2020 Nov 23.

Cross-Protection against MERS-CoV by Prime-Boost Vaccination Using Viral Spike DNA and Protein

Affiliations

Cross-Protection against MERS-CoV by Prime-Boost Vaccination Using Viral Spike DNA and Protein

Jung-Ah Choi et al. J Virol. .

Abstract

Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe respiratory illness and has a high mortality of ∼34%. However, since its discovery in 2012, an effective vaccine has not been developed for it. To develop a vaccine against multiple strains of MERS-CoV, we targeted spike glycoprotein (S) using prime-boost vaccination with DNA and insect cell-expressed recombinant proteins for the receptor-binding domain (RBD), S1, S2, SΔTM, or SΔER. Our S subunits were generated using an S sequence derived from the MERS-CoV EMC/2012 strain. We examined humoral and cellular immune responses of various combinations with DNA plasmids and recombinant proteins in mice. Mouse sera immunized with SΔER DNA priming/SΔTM protein boosting showed cross-neutralization against 15 variants of S-pseudovirions and the wild-type KOR/KNIH/002 strain. In addition, these immunizations provided full protection against the KOR/KNIH/002 strain challenge in human DPP4 knock-in mice. These findings suggest that vaccination with the S subunits derived from one viral strain can provide cross-protection against variant MERS-CoV strains with mutations in S. DNA priming/protein boosting increased gamma interferon production, while protein-alone immunization did not. The RBD subunit alone was insufficient to induce neutralizing antibodies, suggesting the importance of structural conformation. In conclusion, heterologous DNA priming with protein boosting is an effective way to induce both neutralizing antibodies and cell-mediated immune responses for MERS-CoV vaccine development. This study suggests a strategy for selecting a suitable platform for developing vaccines against MERS-CoV or other emerging coronaviruses.IMPORTANCE Coronavirus is an RNA virus with a higher mutation rate than DNA viruses. Therefore, a mutation in S-protein, which mediates viral infection by binding to a human cellular receptor, is expected to cause difficulties in vaccine development. Given that DNA-protein vaccines promote stronger cell-mediated immune responses than protein-only vaccination, we immunized mice with various combinations of DNA priming and protein boosting using the S-subunit sequences of the MERS-CoV EMC/2012 strain. We demonstrated a cross-protective effect against wild-type KOR/KNIH/002, a strain with two mutations in the S amino acids, including one in its RBD. The vaccine also provided cross-neutralization against 15 different S-pseudotyped viruses. These suggested that a vaccine targeting one variant of S can provide cross-protection against multiple viral strains with mutations in S. The regimen of DNA priming/Protein boosting can be applied to the development of other coronavirus vaccines.

Keywords: MERS-CoV; vaccines.

PubMed Disclaimer

Figures

FIG 1
FIG 1
Comparison of immunogenicity induced by various MERS-CoV S subunit DNA constructs. (A) S subunit protein expression in 293T cells, includingsecretion into cell culture supernatants, was confirmed by Western blotting after transfection with each DNA construct. (Mock, Mock plasmid; pSΔER, SΔER plasmid; pSΔTM, SΔTM plasmid; pS1, S1 plasmid; pRBD, RBD (+signal peptide) plasmid; pS2, S2 (ΔTM + signal peptide) plasmid. (B and C) Immunogenicity of various MERS-CoV S subunit DNA constructs. BALB/c mice (n =5) were i.m. immunized with 50 μg of pSΔER, pSΔTM, pS1, pRBD, and pS2 DNA or 50 μl of PBS three times (at days 0, 14, and 28). Sera were collected 14 days after the third immunization (at day 42), and RBD protein-specific serum IgG level was measured by ELISA. Significant differences are indicated as follows: *, P < 0.05; **, P < 0.01; ***, P < 0.001; and NS, not significant. (B). Neutralizing activity of 1/4-, 1/16-, and 1/64-diluted sera against MERS-CoV EMC/2012 and KOR/KNIH/002 S-pseudovirions was analyzed by measuring luciferase activity (C). The results are expressed as means ± the standard deviations (SD). Significant differences are indicated as follows: *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001.
FIG 2
FIG 2
Humoral and cellular immune responses induced by immunization with pSΔER DNA/various recombinant MERS-CoV S subunit proteins. Enhancedimmunogenicity by priming with pSΔER DNA and boosting with various recombinant MERS-CoV S subunit proteins. (A) Antibody responses induced by DNA prime-protein boost. BALB/c mice (n =5) were i.m. immunized with pSΔER DNA (50 μg) two times, followed by various S-proteins (SΔTM, S1, S2, and RBD, 1 μg each), and pSΔER DNA (50 μg), SΔTM protein (1 μg), S1 protein (1 μg), RBD protein (1 μg), or PBS three times as controls. All of the proteins were adjuvanted with alum hydroxide. Mouse sera were collected 14 days after the last immunization (at day 42), and their titers were analyzed by ELISA. Serum IgG titers were measured against SΔTM, S1, and RBD proteins. IgG titer is expressed as the reciprocal log2 of serum dilution showing an absorbance of 0.2 at 450 nm. The results are expressed as means ± the SD. Significant differences are indicated as follows: *, P < 0.05; **, P < 0.01; ***, P < 0.001; and NS, not significant. Statistical significance was compared between the pSΔER DNA-immunized group, or SΔTM protein-immunized group against the pSΔER DNA prime-SΔTM protein boost immunized group. (B) Neutralizing activity of DNA prime-protein boost vaccine against with MERS-CoV vaccine. The neutralizing effect of serially diluted sera (1/10, 1/50, 1/250, 1/1,250, and 1/6,250) against EMC/2012 S-pseudovirions and KOR/KNIH/002 S-pseudovirions was assessed by measuring luciferase activity. The results are indicated as means ± the SD. Significant differences are indicated as follows: *, P < 0.05; **, P < 0.01; ***, P < 0.001; NS, not significant. (C) T-cell immune response induced by DNA prime–protein boost with MERS-CoV vaccine. Vaccinated C57BL/6 mice were sacrificed 14 days after the last immunization (at day 42), and splenocytes were stimulated with S1, RBD, and S2 pooled peptides. IFN-γ-producing T cells were enumerated by using an ELISpot assay. There were statistically significant differences between the PBS group versus all three DNA prime-protein boost groups and the SΔTM protein group (two-tail Mann-Whitney U test). The results are expressed as means ± the SD.
FIG 3
FIG 3
In vitro neutralizing effect of MERS-CoV vaccine against the wild-type MERS-CoV strain. Serially diluted mouse sera were incubated with the wild-type MERS-CoV KOR/KNIH/002 strain, and the mixture was added to VeroE6 cells. After 3 days, the plaque numbers were counted, and the last dilution factor showing a value greater than 50% is expressed as the PRNT50 titer. Statistical analyses of each group were performed with a two-tailed unpaired test. The results are expressed as means ± the SD. Significant differences are indicated as follows: *, P < 0.05; **, P < 0.01; and ***, P < 0.001. There was no statistical significance between the experimental groups except for the statistically marked experimental group.
FIG 4
FIG 4
Evaluation of cross-neutralizing activities against various MERS-CoV S-pseudovirions. The neutralizing activities of the DNA prime-protein boost vaccine, protein vaccine, and DNA vaccine were assessed. Diluted mouse sera (1/10, 1/50, 1/250, 1/1,250, and 1/6,250) were tested for their neutralizing activities against 15 pseudovirions by measuring the luciferase activities of target cells.
FIG 5
FIG 5
In vivo protective effect of MERS-CoV vaccine against wild-type MERS-CoV challenge. Immunization was performed using a total of 16 hDPP4 knock-in mice (7 to 9 weeks old). hDPP4 knock-in mice were i.m. immunized with pSΔER DNA two times, followed by SΔTM protein (n =4), pSΔER DNA (n =5), SΔTM protein (n =4), or PBS (n =3) as the negative control. All proteins were adjuvanted with alum hydroxide. Two weeks after the third immunization (at day 42), the mice were intranasally challenged with 2 × 105 PFU of the wild-type MERS-CoV KOR/KNIH/002 strain. The survival rates (A) and body weight changes (B) for all mice were then monitored daily for 14 days.

Similar articles

Cited by

References

    1. van Boheemen S, de Graaf M, Lauber C, Bestebroer TM, Raj VS, Zaki AM, Osterhaus AD, Haagmans BL, Gorbalenya AE, Snijder EJ, Fouchier RA. 2012. Genomic characterization of a newly discovered coronavirus associated with acute respiratory distress syndrome in humans. mBio 3:e00473-12. doi:10.1128/mBio.00473-12. - DOI - PMC - PubMed
    1. Hemida MG, Elmoslemany A, Al-Hizab F, Alnaeem A, Almathen F, Faye B, Chu DK, Perera RA, Peiris M. 2017. Dromedary camels and the transmission of Middle East respiratory syndrome coronavirus (MERS-CoV). Transbound Emerg Dis 64:344–353. doi:10.1111/tbed.12401. - DOI - PMC - PubMed
    1. Azhar EI, El-Kafrawy SA, Farraj SA, Hassan AM, Al-Saeed MS, Hashem AM, Madani TA. 2014. Evidence for camel-to-human transmission of MERS coronavirus. N Engl J Med 370:2499–2505. doi:10.1056/NEJMoa1401505. - DOI - PubMed
    1. Modjarrad K. 2016. MERS-CoV vaccine candidates in development: the current landscape. Vaccine 34:2982–2987. doi:10.1016/j.vaccine.2016.03.104. - DOI - PMC - PubMed
    1. Graham RL, Donaldson EF, Baric RS. 2013. A decade after SARS: strategies for controlling emerging coronaviruses. Nat Rev Microbiol 11:836–848. doi:10.1038/nrmicro3143. - DOI - PMC - PubMed

Publication types

MeSH terms

LinkOut - more resources

-