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
. 2015 Apr;89(7):3870-87.
doi: 10.1128/JVI.03566-14. Epub 2015 Jan 21.

Severe acute respiratory syndrome coronaviruses with mutations in the E protein are attenuated and promising vaccine candidates

Affiliations

Severe acute respiratory syndrome coronaviruses with mutations in the E protein are attenuated and promising vaccine candidates

Jose A Regla-Nava et al. J Virol. 2015 Apr.

Abstract

Severe acute respiratory syndrome coronavirus (SARS-CoV) causes a respiratory disease with a mortality rate of 10%. A mouse-adapted SARS-CoV (SARS-CoV-MA15) lacking the envelope (E) protein (rSARS-CoV-MA15-ΔE) is attenuated in vivo. To identify E protein regions and host responses that contribute to rSARS-CoV-MA15-ΔE attenuation, several mutants (rSARS-CoV-MA15-E*) containing point mutations or deletions in the amino-terminal or the carboxy-terminal regions of the E protein were generated. Amino acid substitutions in the amino terminus, or deletion of regions in the internal carboxy-terminal region of E protein, led to virus attenuation. Attenuated viruses induced minimal lung injury, diminished limited neutrophil influx, and increased CD4(+) and CD8(+) T cell counts in the lungs of BALB/c mice, compared to mice infected with the wild-type virus. To analyze the host responses leading to rSARS-CoV-MA15-E* attenuation, differences in gene expression elicited by the native and mutant viruses in the lungs of infected mice were determined. Expression levels of a large number of proinflammatory cytokines associated with lung injury were reduced in the lungs of rSARS-CoV-MA15-E*-infected mice, whereas the levels of anti-inflammatory cytokines were increased, both at the mRNA and protein levels. These results suggested that the reduction in lung inflammation together with a more robust antiviral T cell response contributed to rSARS-CoV-MA15-E* attenuation. The attenuated viruses completely protected mice against challenge with the lethal parental virus, indicating that these viruses are promising vaccine candidates.

Importance: Human coronaviruses are important zoonotic pathogens. SARS-CoV caused a worldwide epidemic infecting more than 8,000 people with a mortality of around 10%. Therefore, understanding the virulence mechanisms of this pathogen and developing efficacious vaccines are of high importance to prevent epidemics from this and other human coronaviruses. Previously, we demonstrated that a SARS-CoV lacking the E protein was attenuated in vivo. Here, we show that small deletions and modifications within the E protein led to virus attenuation, manifested by minimal lung injury, limited neutrophil influx to the lungs, reduced expression of proinflammatory cytokines, increased anti-inflammatory cytokine levels, and enhanced CD4(+) and CD8(+) T cell counts in vivo, suggesting that these phenomena contribute to virus attenuation. The attenuated mutants fully protected mice from challenge with virulent virus. These studies show that mutations in the E protein are not well tolerated and indicate that this protein is an excellent target for vaccine development.

PubMed Disclaimer

Figures

FIG 1
FIG 1
Schematic of mutations and deletions introduced within SARS-CoV E protein and growth kinetics of the mutant viruses (rSARS-CoV-MA15-E*). (A) The SARS-CoV genome is shown in the top, and the expanded region shows the E protein sequence and its different regions. White boxes represent the amino acids deleted within the E protein in each virus. Gray letters indicate the amino acids mutated to change the amino-terminal region of the protein. (B) Mutant virus growth kinetics. Subconfluent monolayers of Vero E6 and Huh7.5.1 cells were infected with wt, ΔE, and rSARS-CoV-MA15-E* viruses at an MOI of 0.001 on Vero E6 cells. Error bars represent standard deviations of the mean using data from three independent experiments. Hexagon, WT; diamond, ΔE; pentagon, Mut 1; triangle, Δ2; star, Δ3; inverted triangle, Δ4; square, Δ5; and circle, Δ6.
FIG 2
FIG 2
Subcellular localization of mutant E proteins. (A) Vero E6 cells were infected with either rSARS-CoV-MA15-ΔE, -Δ3, or -Δ5 or wt recombinant viruses, at an MOI of 0.3, and fixed at 24 hpi. E protein (green) and ERGIC (red) were labeled with specific antibodies. Nuclei were stained with DAPI (blue). Merge indicates superposition of both labels. Original magnification was ×126. (B) The panel represents the percentage of overlap coefficient between E protein and ERGIC, calculated with Leica LAS AF v2.6.0 software.
FIG 3
FIG 3
Stability of rSARS-CoV-MA15-E* after serial infections. The stability of rSARS-CoV-MA15-E* virus deletion mutants was examined after 8 passages in Vero E6 cells by sequence analysis. Asterisks denote the presence of nucleotide substitutions.
FIG 4
FIG 4
Weight loss and survival rate of mice inoculated with rSARS-CoV-MA15-E* mutants. BALB/c mice were intranasally infected with 1 × 105 PFU of each virus (n = 5 mice). Animals were monitored daily for weight loss (A) and survival (B). Animals that lost more than 30% of their initial body weight were euthanized. Differences in weight loss between attenuated and virulent viruses were statistically significant (*, P < 0.05).
FIG 5
FIG 5
rSARS-CoV-MA15-E* growth in the lungs of infected mice. BALB/c mice were intranasally inoculated with 1 × 105 PFU of the indicated viruses. At 2 and 4 days p.i., lung tissue was harvested and viral titers were analyzed in Vero E6 cell monolayers. Means and standard deviations are shown (n = 3 mice).
FIG 6
FIG 6
Lung pathology caused by infection with rSARS-CoV-MA15-E* mutants. BALB/c mice were intranasally inoculated with 1 × 105 PFU of the indicated SARS-CoV deletion mutants and sacrificed at days 2 and 4 p.i. (A) Lungs were removed and sections were prepared and stained with hematoxylin and eosin. Asterisks indicate edema accumulation in both bronchiolar and alveolar airways. Original magnification was ×20. Representative images are shown. (B) Macroscopic lung pathology in rSARS-CoV-MA15-E*-infected mice. Representative images of lung gross pathology. (C) Lung weight. Three lungs were evaluated in each case (n = 3 mice). Statistically significant data compared to mice infected with the wt virus are indicated (**, P < 0.01).
FIG 7
FIG 7
Specific infectivity of rSARS-CoV-MA15-E* mutants. Vero E6 cells were independently infected with each of the rSARS-CoV-MA15-ΔE, -Δ3, and -Δ5 and wt viruses, at an MOI of 0.3. At 8 hpi, the culture medium was harvested and replaced with fresh medium supplemented with 2% FBS. Cell supernatants were collected at 11 hpi (“nascent virus”), and the levels of genomic RNA and infectious virus titer were analyzed by RT-qPCR and plaque assay, respectively. The ratio of infectious virus titer (PFU/ml) to genomic RNA is depicted in the graph as percentage of specific infectivity. Means and standard deviations are shown (n = 3 mice).
FIG 8
FIG 8
E protein stability of rSARS-CoV-MA15* mutants. Stability of E protein deletion mutants was evaluated in relation to that of the full-length E protein after infection of Vero E6 cells with rSARS-CoV-MA15-E* mutants. At 12 hpi, the cells were treated with cycloheximide. Cells were harvested at the indicated times, and the amount of E protein mutants was determined by Western blotting. (A) Membranes were probed with anti-E protein and with β-actin-specific antibodies as a loading control. (B) The graph represents the values obtained after densitometry analysis. The percentage of protein remaining after cycloheximide addition is represented. Bars represent standard deviations of the mean (n = 3 mice).
FIG 9
FIG 9
Interaction of SARS-CoV E protein deletion mutants with M protein. Vero E6 cells were cotransfected with a plasmid pcDNA3 encoding the N-terminal HA-tagged M protein, combined with plasmids expressing the E-wt protein or the E protein with a small deletion, E-Δ3. Cotransfections of a plasmid encoding HA-M protein and of a plasmid without E protein were used as controls. Cells were lysed and analyzed by Western blotting with specific antibodies for the E protein and HA (left) or subjected to immunoprecipitation with the monoclonal HA-specific antibody. The presence of E and M proteins was analyzed in the precipitated fractions using E- or HA-specific antibodies (right).
FIG 10
FIG 10
Leukocyte infiltrates present in the lungs of rSARS-CoV-MA15-E*-infected mice. BALB/c mice were intranasally infected with 1 × 105 PFU of the indicated SARS-CoV deletion mutants and sacrificed at 4 days p.i. The total number of leukocytes (A) and total numbers and percentages of macrophages (B), neutrophils (C), CD4+ T cells (D), and CD8+ T cells (E) were determined. Error bars represent the standard deviations of the means. Data are representative of three independent experiments (n = 3 mice). Statistically significant differences compared to infection with the wt virus are indicated (*, P < 0.05).
FIG 11
FIG 11
Differential gene expression in rSARS-CoV-MA15-E*-infected mice. Total lung RNA was extracted from mice infected with the indicated SARS-CoV deletion mutants at 2 days p.i. (n = 3 mice). (A) Differential gene expression was measured using microarrays. Only genes with an FDR less than 0.05 were considered candidate genes. Points with a differential expression higher or lower than 2-fold are represented as darker or lighter dots, respectively. (B) Candidate genes that were up- and downregulated in ΔE, Δ3, and Δ5 viruses were grouped according to Gene Ontology terms. Numbers on the x axis indicate DAVID FDR values. (C) Genes differentially expressed in ΔE-, Δ3-, and Δ5-infected mice compared to in wt-infected mice were classified according to their main biological functions. Black and gray lettering is used to indicate up- and downregulated genes, respectively. Asterisks indicates genes whose expression was confirmed by RT-qPCR. The numbers indicate the fold change for each gene in ΔE-, Δ3-, and Δ5-infected mice compared to in wt-infected mice. For those genes detected with more than one probe, the value corresponding to the highest upregulation or downregulation is represented.
FIG 12
FIG 12
Effect of rSARS-CoV-MA15-E* deletion mutants on the expression of cytokine mRNAs in BALB/c mouse lung. Mice were infected with 1 × 105 PFU of rSARS-CoV-MA15-Δ3, -Δ5, and -ΔE and wt viruses, and total RNA was extracted at 2 and 4 days p.i. The expression of mRNAs encoding inflammatory genes (A) and immune response and IFN response genes encoding TGFβ and 18S rRNA (B) was measured by qRT-PCR. In each case, levels of expression in infected lungs were compared to those in mock-infected ones. Bars represent standard deviations of the mean (n = 3 mice). Statistically significant data compared to mice infected with the wt virus are indicated (*, P < 0.05; **, P < 0.01).
FIG 13
FIG 13
Expression of cytokines in rSARS-CoV-MA15-E*-infected mice. Mice were infected with 1 × 105 PFU of rSARS-CoV-MA15-wt, -Δ3, -Δ5, or -ΔE virus or were mock infected (mock). Lung proteins were extracted at 2 days p.i., and the accumulation of several cytokine proteins was measured. Protein concentration is expressed as picograms per milliliter of lung tissue extract. Means and standard deviations are shown (n = 3 mice). Statistically significant differences compared to mice infected with wt virus are indicated (*, P < 0.05).
FIG 14
FIG 14
Protection conferred by immunization with the rSARS-CoV-MA15-E* mutants. Six-week-old BALB/c mice were mock immunized or immunized with 6,000 PFU of the SARS-CoV-MA15-E* mutants and challenged at day 21 postimmunization with 1 × 105 PFU of MA15 virus (n = 5 mice). Weight loss (A) and survival (B) were recorded daily.

Similar articles

Cited by

References

    1. Perlman S, Netland J. 2009. Coronaviruses post-SARS: update on replication and pathogenesis. Nat Rev Microbiol 7:439–450. doi:10.1038/nrmicro2147. - DOI - PMC - PubMed
    1. Drosten C, Gunther S, Preiser W, van der Werf S, Brodt HR, Becker S, Rabenau H, Panning M, Kolesnikova L, Fouchier RA, Berger A, Burguiere AM, Cinatl J, Eickmann M, Escriou N, Grywna K, Kramme S, Manuguerra JC, Muller S, Rickerts V, Sturmer M, Vieth S, Klenk HD, Osterhaus AD, Schmitz H, Doerr HW. 2003. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med 348:1967–1976. doi:10.1056/NEJMoa030747. - DOI - PubMed
    1. Rota PA, Oberste MS, Monroe SS, Nix WA, Campganoli R, Icenogle JP, Peñaranda S, Bankamp B, Maher K, Chen M-H, Tong S, Tamin A, Lowe L, Frace M, DeRisi JL, Chen Q, Wang D, Erdman DD, Peret TC, Burns C, Ksiazek TG, Rollin PE, Sanchez A, Liffick S, Holloway B, Limor J, McCaustland K, Olsen-Rassmussen M, Fouchier R, Gunther S, Osterhaus AD, Drosten C, Pallansch MA, Anderson LJ, Bellini WJ. 2003. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science 300:1394–1399. doi:10.1126/science.1085952. - DOI - PubMed
    1. Kuiken T, Fouchier RAM, Schutten M, Rimmelzwaan GF, van Amerongen G, van Riel D, Laman JD, de Jong T, van Doornum G, Lim W, Ling AE, Chan PKS, Tam JS, Zambon MC, Gopal R, Drosten C, van der Werf S, Escriou N, Manuguerra J-C, Stohr K, Peiris JSM. 2003. Newly discovered coronavirus as the primary cause of severe acute respiratory syndrome. Lancet 362:263–270. doi:10.1016/S0140-6736(03)13967-0. - DOI - PMC - PubMed
    1. Marra MA, Jones SJM, Astell CR, Holt RA, Brooks-Wilson A, Butterfield YSN, Khattra J, Asano JK, Barber SA, Chan SY, Cloutier A, Coughlin SM, Freeman D, Girn N, Griffith OL, Leach SR, Mayo M, McDonald H, Montgomery SB, Pandoh PK, Petrescu AS, Robertson AG, Schein JE, Siddiqui A, Smailus DE, Stott JM, Yang GS, Plummer F, Andonov A, Artsob H, Bastien N, Bernard K, Booth TF, Bowness D, Czub M, Drebot M, Fernando L, Flick R, Garbutt M, Gray M, Grolla A, Jones S, Feldmann H, Meyers A, Kabani A, Li Y, Normand S, Stroher U, Tipples GA, Tyler S, et al. . 2003. The genome sequence of the SARS-associated coronavirus. Science 300:1399–1404. doi:10.1126/science.1085953. - DOI - PubMed

Publication types

MeSH terms