Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Type of study
Publication year range
1.
Arch Virol ; 161(7): 1821-30, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27068165

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) causes significant economic losses for the swine industry worldwide. The PRRSV E protein, encoded by ORF 2b, is one of the non-glycosylated minor structural proteins. In this study, we present evidence for the interaction of the E protein with mitochondrial proteins ATP5A (part of ATP synthase complex), prohibitin, and ADP/ATP translocase. We additionally demonstrate partial mitochondrial localization of the E protein in transfected cells. To functionally investigate these interactions, we infected MARC-145 cells with PRRSV or alphavirus replicon particles (VRPs) expressing PRRSV E protein. In infected cells, production of ATP was significantly reduced. The E protein also induced apoptosis by activating caspase-3, which results in PARP cleavage. Taken together, these data suggest that the PRRSV E protein interacts with mitochondrial proteins and induces apoptosis by inhibiting ATP production.


Subject(s)
Apoptosis , Mitochondrial Proton-Translocating ATPases/metabolism , Porcine Reproductive and Respiratory Syndrome/enzymology , Porcine Reproductive and Respiratory Syndrome/physiopathology , Porcine respiratory and reproductive syndrome virus/metabolism , Viral Proteins/metabolism , Animals , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Protein Binding , Swine , Viral Proteins/genetics
3.
Open Virol J ; 9: 1-6, 2015.
Article in English | MEDLINE | ID: mdl-26069516

ABSTRACT

Adenoviral vectors are now being explored as vaccine carriers to prevent infectious diseases in humans and animals. There are two strategies aimed at the expression of a vaccine antigen by adenoviral vectors. The first includes an insertion of the foreign gene expression cassette into the E1 region. The second strategy is antigen incorporation into the viral capsid proteins. To extend this methodology, we have searched for new sites at the human adenovirus serotype 5 major capsid protein hexon for a vaccine antigen insertion. To this end, we utilized sites in the hexon hypervariable region (HVR) 7, 8 and 9 to display a 15-mer peptide containing the main neutralizing epitope of porcine reproductive and respiratory syndrome virus. However, we could not rescue the viruses with the insertions of the peptide into HVR 8 and 9, consistent with the viruses being unable to tolerate insertions at these sites. In contrast, the virus with the insertion of the peptide in HVR 7 was viable - growing well in cell culture and the inserted peptide was exposed on the virion surface.

4.
Virus Res ; 194: 138-44, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25304692

ABSTRACT

Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) is a positive sense, single-stranded RNA genome virus that has become a major infection in swine, exerting huge economic losses to the industry worldwide. Detailed knowledge concerning the molecular mechanisms by which the virus manipulates the host cell signals transduction machinery is not only critical to further our understanding of viral replication and pathogenesis, but also guides our efforts to design new and improved therapeutic strategies. The phosphatidylinositol-3-kinase (PI3K)-dependent Akt and the mammalian target of rapamycin (mTOR) (PI3K/Akt/mTOR) are major host cell signalling pathways that regulate protein synthesis, cell growth, proliferation, migration and survival. It is also established that many viruses exploit these signalling cascades for their own benefit, driving viral protein expression, replication, as well as the suppression of the host's antiviral activities. In this article, we will review the role of these signalling pathways during PRRSV replication, and discuss some of our recent findings implicating mTOR.


Subject(s)
Host-Pathogen Interactions , Phosphatidylinositol 3-Kinase/metabolism , Porcine respiratory and reproductive syndrome virus/physiology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Virus Replication , Animals , Swine
5.
Biomed Res Int ; 2014: 160505, 2014.
Article in English | MEDLINE | ID: mdl-24511529

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is a serious threat to the pork industry, and its pathogenesis needs further investigations. To study the role of two structural proteins of PRRSV in virus-host cells interactions, two stable cell lines (MARC-2a and MARC-N) expressing GP2 and N proteins, respectively, were established. We induced apoptosis in these cells by treating them with staurosporine and found a significant reduction in the number of apoptotic cells in MARC-2a as compared to MARC-N and MARC-145 cells. In addition, we found significantly higher activities of transcriptional factors (NF- κ B and AP-1) in both cell lines as compared to MARC-145 (parent cells). Overall, our data suggest that, although both stable cell lines activate NF- κ B and AP-1, GP2 triggers the antiapoptotic process through an intermediate step that needs to be further investigated.


Subject(s)
Apoptosis/genetics , Porcine respiratory and reproductive syndrome virus/genetics , Viral Structural Proteins/genetics , Animals , Cell Line/virology , Meat , NF-kappa B/genetics , Porcine respiratory and reproductive syndrome virus/metabolism , Staurosporine/administration & dosage , Swine
6.
Virus Res ; 173(2): 321-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23353778

ABSTRACT

Though a modified live attenuated vaccine (MLV) is available against porcine reproductive and respiratory syndrome virus (PRRSV), its limitations in protective efficacy, safety and few others warrant the development of newer vaccines. In this study, we have constructed a propagation-defective DNA-launched PRRSV replicon as a vaccine candidate and evaluated its immunogenicity and protective efficacy in a group of pigs along with MLV vaccinated group. Our data showed that prior to the intranasal challenge with a homologous strain of PRRSV, only MLV vaccinated pigs developed antibody response measured by ELISA and none of the pigs in any group developed PRRSV neutralizing antibodies in serum. The MLV vaccinated group also showed high PRRSV-specific INF-γ response, whereas the replicon-vaccinated pigs showed low but detectable INF-γ response. After 14 days post challenge, all groups showed similar PRRSV-specific serum neutralizing titers and were positive for PRRSV-specific ELISA antibody. In addition, the replicon-vaccinated group showed a significant reduction in viremia in comparison to the control group. In conclusion, vaccination with the PRRSV DNA-launched replicon decreased the viremia and viral load in bronchoalveolar lavage fluids of the PRRSV-challenged pigs and increased numbers of IFN-γ producing cells. Thus, the vaccine is partially protective and is a potential vaccine candidate for future with further improvement. The possible means of improvement is the expression of immunostimulatory genes by the replicon. We demonstrated the feasibility of this approach by expression of a foreign gene encoding firefly luciferase after transfection of cultured cells with the replicon plasmid DNA.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Bronchoalveolar Lavage Fluid/virology , Enzyme-Linked Immunosorbent Assay , Gammaretrovirus , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/genetics , Swine , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Load , Viremia/prevention & control
7.
Vaccine ; 30(20): 3061-7, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22406109

ABSTRACT

Despite the availability of live attenuated measles virus (MV) vaccines, a large number of measles-associated deaths occur among infants in developing countries. The development of a measles subunit vaccine may circumvent the limitations associated with the current live attenuated vaccines and eventually contribute to global measles eradication. Therefore, the goal of this study was to test the feasibility of producing the recombinant globular head domain of the MV hemagglutinin (H) protein by stably transfected human cells and to examine the ability of this recombinant protein to elicit MV-specific immune responses. The recombinant protein was purified from the culture supernatant of stably transfected HEK293T cells secreting a tagged version of the protein. Two subcutaneous immunizations with the purified recombinant protein alone resulted in the production of MV-specific serum IgG and neutralizing antibodies in mice. Formulation of the protein with adjuvants (polyphosphazene or alum) further enhanced the humoral immune response and in addition resulted in the induction of cell-mediated immunity as measured by the production of MV H-specific interferon gamma (IFN-γ) and interleukin 5 (IL-5) by in vitro re-stimulated splenocytes. Furthermore, the inclusion of polyphosphazene into the vaccine formulation induced a mixed Th1/Th2-type immune response. In addition, the purified recombinant protein retained its immunogenicity even after storage at 37°C for 2 weeks.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Measles/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cell Line , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/isolation & purification , Humans , Interferon-gamma/metabolism , Interleukin-5/metabolism , Leukocytes, Mononuclear/immunology , Measles Vaccine/administration & dosage , Mice , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Th1 Cells/immunology , Th2 Cells/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
8.
Vaccine ; 28(48): 7613-9, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-20887832

ABSTRACT

Several problems associated with the available anti-measles vaccine emphasize the need for a single shot anti-measles vaccine which is efficacious by mucosal route of administration and functional in the presence of anti-measles neutralizing antibodies. To achieve these goals, we constructed two recombinant human adenoviruses (collectively designated Ad-F/H) carrying genes for measles virus (MV) fusion (F) and haemagglutinin (H) proteins. Single intranasal or intramuscular vaccination of mice and cotton rats with Ad-F/H elicited high MV-specific serum neutralizing-antibody titers. Furthermore, bronchoalveolar lavage samples from mice vaccinated intranasally with Ad-F/H showed a 100-fold increase in MV-specific IgA titers compared with intramuscularly vaccinated mice. Moreover, Ad-F/H vaccine administered intranasally, but not intramuscularly, completely protected challenged cotton rats from MV replication in the lungs.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Measles Vaccine/immunology , Measles/prevention & control , Administration, Intranasal , Administration, Mucosal , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chlorocebus aethiops , Female , HEK293 Cells , Hemagglutinins, Viral/immunology , Humans , Immunity, Mucosal , Immunoglobulin A/immunology , Injections, Intramuscular , Lung/immunology , Lung/microbiology , Measles/immunology , Measles Vaccine/genetics , Measles virus/immunology , Mice , Mice, Inbred C57BL , Neutralization Tests , Rats , Vero Cells , Viral Fusion Proteins/immunology
9.
DNA Cell Biol ; 26(10): 721-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17665998

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV) first appeared in Southern China in November 2002, and then quickly spread to 33 countries on five continents along international air travel routes. Although the SARS epidemic has been contained, there is a clear need for a safe and effective vaccine should an outbreak of a SARS-CoV infection reappear in human population. In this study, we tested four DNA-vaccine constructs: (1) pLL70, containing cDNA for the SARS-CoV spike (S) gene; (2) pcDNA-SS, containing codon-optimized S gene for SARS-CoV S protein (residues 12-1255) fused with a leader sequence derived from the human CD5 gene; (3) pcDNA-St, containing the gene encoding the N-portion of the codon-optimized S gene (residues 12-532) with the CD5 leader sequence; (4) pcDNA-St-VP22C, containing the gene encoding the N-portion of the codon-optimized S protein with the CD5 leader sequence fused with the C-terminal 138 amino acids of the bovine herpesvirus-1 (BHV-1) major tegument protein VP22. Each of these plasmids was intradermally administered to C57BL/6 mice in three separate immunizations. Analysis of humoral and cellular immune responses in immunized mice demonstrated that pcDNA-SS and pcDNA-St-VP22C are the most immunogenic SARS vaccine candidates.


Subject(s)
Membrane Glycoproteins/immunology , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology , Viral Structural Proteins/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Antibody Formation , CD5 Antigens/genetics , Female , Humans , Immunity, Cellular , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Plasmids , Protein Sorting Signals , Severe Acute Respiratory Syndrome/prevention & control , Spike Glycoprotein, Coronavirus , Vaccines, DNA/genetics , Viral Envelope Proteins/genetics , Viral Structural Proteins/genetics , Viral Vaccines/genetics
10.
Virus Res ; 127(1): 116-21, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17448558

ABSTRACT

Open reading frame 9b (ORF 9b) encodes a 98 amino acid group-specific protein of severe acute respiratory syndrome (SARS) coronavirus (CoV). It has no homology with known proteins and its function in SARS CoV replication has not been determined. The N-terminal part of the 9b protein was used to raise polyclonal antibodies in rabbits, and these antibodies could detect 9b protein in infected cells. We analyzed the sub-cellular localization of recombinant 9b protein using fluorescence microscopy of live transfected cells and indirect immunofluorescence of transfected fixed cells. Our findings indicate that the 9b protein is exported outside of a cell nucleus and localizes to the endoplasmic reticulum. Our data also suggest that the 46-LRLGSQLSL-54 amino acid sequence of 9b functions as a nuclear export signal (NES).


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/virology , Severe acute respiratory syndrome-related coronavirus/chemistry , Severe acute respiratory syndrome-related coronavirus/metabolism , Animals , Antibodies, Viral/metabolism , Biological Transport , Capsid Proteins/genetics , Cell Line , Chlorocebus aethiops , Fluorescent Antibody Technique, Direct , Nuclear Localization Signals/metabolism , Rabbits , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/immunology , Vero Cells
11.
Methods Mol Med ; 130: 91-106, 2007.
Article in English | MEDLINE | ID: mdl-17401166

ABSTRACT

Adenoviruses have become a popular vehicle for gene transfer into animal and human cells. However, wide prevalence of preexisting immunity to human adenovirus (HAdV) and the promiscuous nature of the virus have made the use of nonhuman adenoviruses an attractive alternative. Moreover, readministration of viral vectors is often required to maintain therapeutic levels of transgene expression, resulting in vector-specific immune responses. Although a number of features of bovine adenovirus (BAdV)-3 make it attractive for use as a vector in human vaccination, BAdV-3 transduces nonbovine cells, including human cells, poorly. However, genetic modification of capsid proteins (e.g., fiber, pIX) has helped in increasing the utility of BAdV-3 as a vector for transducing nonbovine cells. Here, we will describe the methods used to construct recombinant BAdV-3 expressing chimeric fiber or chimeric pIX proteins.


Subject(s)
Atadenovirus/genetics , Capsid Proteins/genetics , Recombination, Genetic , Animals , Atadenovirus/immunology , Atadenovirus/isolation & purification , Cattle , Gene Transfer Techniques , Genetic Vectors , Humans , Plasmids , Restriction Mapping , Viral Vaccines
12.
Vaccine ; 25(1): 136-43, 2007 Jan 02.
Article in English | MEDLINE | ID: mdl-16919855

ABSTRACT

We studied the immunogenicity of an anti-SARS subunit vaccine comprised of the fragment of the SARS coronavirus (SARS-CoV) spike protein amino acids 318-510 (S318-510) containing the receptor-binding domain. The S protein fragment was purified from the culture supernatant of stably transformed HEK293T cells secreting a tagged version of the protein. The vaccine was given subcutaneously to 129S6/SvEv mice in saline, with alum adjuvant or with alum plus CpG oligodeoxynucleotides (ODN). Mice immunized with the adjuvanted antigen elicited strong antibody and cellular immune responses; furthermore, adding the CpG ODN to the alum resulted in increased IgG2a antibody titers and a higher number of INF-gamma-secreting murine splenocytes. Mice vaccinated with S318-510 deglycosylated by PNGase F (dgS318-510) showed a lower neutralizing antibody response but had similar numbers of INF-gamma-producing cells in the spleen. This finding suggests that carbohydrate is important for the immunogenicity of the S318-510 protein fragment and provide useful information for designing an effective and safe SARS subunit vaccine.


Subject(s)
Membrane Glycoproteins , Receptors, Virus/metabolism , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins , Viral Vaccines/immunology , Alum Compounds , Animals , Antibodies, Viral/blood , Cell Line , Female , Humans , Interferon-gamma/biosynthesis , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Oligodeoxyribonucleotides , Severe Acute Respiratory Syndrome/immunology , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Vaccines/administration & dosage
13.
J Gen Virol ; 87(Pt 3): 641-650, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16476986

ABSTRACT

Two different severe acute respiratory syndrome (SARS) vaccine strategies were evaluated for their ability to protect against live SARS coronavirus (CoV) challenge in a murine model of infection. A whole killed (inactivated by beta-propiolactone) SARS-CoV vaccine and a combination of two adenovirus-based vectors, one expressing the nucleocapsid (N) and the other expressing the spike (S) protein (collectively designated Ad S/N), were evaluated for the induction of serum neutralizing antibodies and cellular immune responses and their ability to protect against pulmonary SARS-CoV replication. The whole killed virus (WKV) vaccine given subcutaneously to 129S6/SvEv mice was more effective than the Ad S/N vaccine administered either intranasally or intramuscularly in inhibiting SARS-CoV replication in the murine respiratory tract. This protective ability of the WKV vaccine correlated with the induction of high serum neutralizing-antibody titres, but not with cellular immune responses as measured by gamma interferon secretion by mouse splenocytes. Titres of serum neutralizing antibodies induced by the Ad S/N vaccine administered intranasally or intramuscularly were significantly lower than those induced by the WKV vaccine. However, Ad S/N administered intranasally, but not intramuscularly, significantly limited SARS-CoV replication in the lungs. Among the vaccine groups, SARS-CoV-specific IgA was found only in the sera of mice immunized intranasally with Ad S/N, suggesting that mucosal immunity may play a role in protection for the intranasal Ad S/N delivery system. Finally, the sera of vaccinated mice contained antibodies to S, further suggesting a role for this protein in conferring protective immunity against SARS-CoV infection.


Subject(s)
Antibodies, Viral/blood , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/immunology , Vaccination , Viral Vaccines/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Antibody Specificity , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Immunoglobulin A/blood , Immunoglobulin A/immunology , Injections, Intramuscular , Injections, Subcutaneous , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Neutralization Tests , Nucleocapsid Proteins/genetics , Severe acute respiratory syndrome-related coronavirus/chemistry , Spike Glycoprotein, Coronavirus , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
14.
Vaccine ; 23(35): 4385-91, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16005746

ABSTRACT

We studied the immunogenicity of a DNA SARS-vaccine, a whole killed virus, or a whole killed and DNA vaccine combination. The DNA vaccine contained a plasmid encoding the SARS coronavirus (SARS-CoV) S protein under the control of the human CMV promoter and intron A. The whole killed virus vaccine was comprised of SARS-CoV, propagated in Vero-E6 cells, with subsequent beta-propilactone inactivation and formulated with aluminum hydroxide adjuvant. Mice immunized twice with the DNA vaccine and once with the whole killed virus elicited higher antibody responses than mice immunized three times with the DNA vaccine or once with the whole killed virus vaccine. Mice immunized twice with the whole killed virus vaccine elicited higher antibody responses than mice immunized three times with the DNA vaccine or once with the whole killed virus vaccine. However, a combination of the vaccines induced T-helper type 1 (Th1) immune responses while the whole killed virus vaccine induced T helper type 2 (Th2) immune response. These results demonstrate that combination of the DNA vaccine and the whole killed virus vaccine can be used to enhance the magnitude and change the bias of the immune responses to SARS-CoV.


Subject(s)
Severe Acute Respiratory Syndrome/prevention & control , Vaccines, DNA/immunology , Vaccines, Inactivated/administration & dosage , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Animals , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Mice , Vaccines, Inactivated/immunology , Vero Cells
15.
J Gen Virol ; 86(Pt 6): 1681-1686, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15914845

ABSTRACT

Simian adenovirus 1 (SAdV-1) is one of many adenovirus strains that were isolated from Old World monkey cells during poliomyelitis vaccine production several decades ago. Despite the availability of these viruses, knowledge of their genetic content and phylogeny is rudimentary. In the present study, the genome sequence of SAdV-1 (34,450 bp) was determined and analysed. In regions where genetic content varies between primate adenoviruses, SAdV-1 has a single virus-associated RNA gene, six genes in each of the E3 and E4 regions and two fiber genes. SAdV-1 clusters phylogenetically with HAdV-40, a member of human adenovirus species HAdV-F, which also has two fiber genes. However, based on phylogenetic distances and other taxonomic criteria, SAdV-1 is proposed to represent a novel adenovirus species.


Subject(s)
Adenoviruses, Simian/genetics , Antigens, Viral/genetics , Capsid Proteins/genetics , Cercopithecidae/virology , Genome, Viral , Amino Acid Sequence , Animals , Molecular Sequence Data , Phylogeny , Sequence Alignment , Species Specificity
16.
J Gen Virol ; 86(Pt 1): 211-215, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15604448

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV) has been identified as the aetiological agent of SARS. Thus, vaccination against SARS-CoV may represent an effective approach towards controlling SARS. The nucleocapsid (N) protein is thought to play a role in induction of cell-mediated immunity to SARS-CoV and thus it is important to characterize this protein. In the present study, an E1/partially E3-deleted, replication-defective human adenovirus 5 (Ad5) vector (Ad5-N-V) expressing the SARS-CoV N protein was constructed. The N protein, expressed in vitro by Ad5-N-V, was of the expected molecular mass of 50 kDa and was phosphorylated. Vaccination of C57BL/6 mice with Ad5-N-V generated potent SARS-CoV-specific humoral and T cell-mediated immune responses. These results show that Ad5-N-V may potentially be used as a SARS-CoV vaccine.


Subject(s)
Nucleocapsid Proteins/biosynthesis , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/genetics , Vaccination/methods , Viral Vaccines/biosynthesis , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antibodies, Viral/biosynthesis , Drug Evaluation, Preclinical , Female , Genetic Vectors , Mice , Mice, Inbred C57BL , Molecular Weight , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Phosphorylation , Recombinant Proteins/biosynthesis , Severe acute respiratory syndrome-related coronavirus/immunology , Severe Acute Respiratory Syndrome/prevention & control , T-Lymphocytes/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...