Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Viruses ; 13(9)2021 08 27.
Article in English | MEDLINE | ID: mdl-34578289

ABSTRACT

Influenza hemagglutinin (HA) is considered a major protective antigen of seasonal influenza vaccine but antigenic drift of HA necessitates annual immunizations using new circulating HA versions. Low variation found within conserved non-HA influenza virus (INFV) antigens may maintain protection with less frequent immunizations. Conserved antigens of influenza A virus (INFV A) that can generate cross protection against multiple INFV strains were evaluated in BALB/c mice using modified Vaccinia virus Ankara (MVA)-vectored vaccines that expressed INFV A antigens hemagglutinin (HA), matrix protein 1 (M1), nucleoprotein (NP), matrix protein 2 (M2), repeats of the external portion of M2 (M2e) or as tandem repeats (METR), and M2e with transmembrane region and cytoplasmic loop (M2eTML). Protection by combinations of non-HA antigens was equivalent to that of subtype-matched HA. Combinations of NP and forms of M2e generated serum antibody responses and protected mice against lethal INFV A challenge using PR8, pandemic H1N1 A/Mexico/4108/2009 (pH1N1) or H5N1 A/Vietnam/1203/2004 (H5N1) viruses, as demonstrated by reduced lung viral burden and protection against weight loss. The highest levels of protection were obtained with NP and M2e antigens delivered as MVA inserts, resulting in broadly protective immunity in mice and enhancement of previous natural immunity to INFV A.


Subject(s)
Antibodies, Viral/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Nucleocapsid Proteins/immunology , Orthomyxoviridae Infections/prevention & control , Viral Matrix Proteins/immunology , Viroporin Proteins/immunology , Animals , Antigens, Viral/immunology , Cross Protection , Female , Genetic Vectors , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Orthomyxoviridae Infections/immunology , Pandemics , Vaccination , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics , Viroporin Proteins/administration & dosage
2.
J Microbiol Biotechnol ; 31(2): 304-316, 2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33263336

ABSTRACT

Vaccination is the most effective way to prevent influenza virus infections. However, conventional vaccines based on hemagglutinin (HA) have to be annually updated because the HA of influenza viruses constantly mutates. In this study, we produced a 3M2e-3HA2-NP chimeric protein as a vaccine antigen candidate using an Escherichia coli expression system. The vaccination of chimeric protein (15 µg) conferred complete protection against A/Puerto Rico/8/1934 (H1N1; PR8) in mice. It strongly induced influenza virus-specific antibody responses, cytotoxic T lymphocyte activity, and antibody-dependent cellular cytotoxicity. To spare the dose and enhance the cross-reactivity of the chimeric, we used a complex of poly-γ-glutamic acid and alum (PGA/alum) as an adjuvant. PGA/alum-adjuvanted, low-dose chimeric protein (1 or 5 µg) exhibited higher cross-protective effects against influenza A viruses (PR8, CA04, and H3N2) compared with those of chimeric alone or alum-adjuvanted proteins in vaccinated mice. Moreover, the depletion of CD4+ T, CD8+ T, and NK cells reduced the survival rate and efficacy of the PGA/alum-adjuvanted chimeric protein. Collectively, the vaccination of PGA/alum-adjuvanted chimeric protein induced strong protection efficacy against homologous and heterologous influenza viruses in mice, which suggests that it may be a promising universal influenza vaccine candidate.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Nucleocapsid Proteins/immunology , Viral Matrix Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , Antibodies, Viral/immunology , Cross Reactions , Female , Hemagglutinins, Viral , Humans , Immunity, Humoral , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Polyglutamic Acid/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics
3.
Viruses ; 12(2)2020 02 20.
Article in English | MEDLINE | ID: mdl-32093256

ABSTRACT

T cells play an essential role in the immune response against the human respiratory syncytial virus (hRSV). It has been described that both CD4+ and CD8+ T cells can contribute to the clearance of the virus during an infection. However, for some individuals, such an immune response can lead to an exacerbated and detrimental inflammatory response with high recruitment of neutrophils to the lungs. The receptor of most T cells is a heterodimer consisting of α and ß chains (αßTCR) that upon antigen engagement induces the activation of these cells. The αßTCR molecule displays a broad sequence diversity that defines the T cell repertoire of an individual. In our laboratory, a recombinant Bacille Calmette-Guérin (BCG) vaccine expressing the nucleoprotein (N) of hRSV (rBCG-N-hRSV) was developed. Such a vaccine induces T cells with a Th1 polarized phenotype that promote the clearance of hRSV infection without causing inflammatory lung damage. Importantly, as part of this work, the T cell receptor (TCR) repertoire of T cells expanded after hRSV infection in naïve and rBCG-N-hRSV-immunized mice was characterized. A more diverse TCR repertoire was observed in the lungs from rBCG-N-hRSV-immunized as compared to unimmunized hRSV-infected mice, suggesting that vaccination with the recombinant rBCG-N-hRSV vaccine triggers the expansion of T cell populations that recognize more viral epitopes. Furthermore, differential expansion of certain TCRVß chains was found for hRSV infection (TCRVß+8.3 and TCRVß+5.1,5.2) as compared to rBCG-N-hRSV vaccination (TCRVß+11 and TCRVß+12). Our findings contribute to better understanding the T cell response during hRSV infection, as well as the functioning of a vaccine that induces a protective T cell immunity against this virus.


Subject(s)
BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Nucleocapsid Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Respiratory Syncytial Virus, Human/immunology , Animals , BCG Vaccine/genetics , Immunity, Cellular , Lung/immunology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Receptors, Antigen, T-Cell/classification , Receptors, Antigen, T-Cell, alpha-beta/genetics , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Specific Pathogen-Free Organisms , Vaccines, Synthetic/immunology
4.
PLoS Negl Trop Dis ; 12(7): e0006628, 2018 07.
Article in English | MEDLINE | ID: mdl-30011277

ABSTRACT

Crimean-Congo hemorrhagic fever (CCHF) is an acute, often fatal viral disease characterized by rapid onset of febrile symptoms followed by hemorrhagic manifestations. The etiologic agent, CCHF orthonairovirus (CCHFV), can infect several mammals in nature but only seems to cause clinical disease in humans. Over the past two decades there has been an increase in total number of CCHF case reports, including imported CCHF patients, and an expansion of CCHF endemic areas. Despite its increased public health burden there are currently no licensed vaccines or treatments to prevent CCHF. We here report the development and assessment of the protective efficacy of an adenovirus (Ad)-based vaccine expressing the nucleocapsid protein (N) of CCHFV (Ad-N) in a lethal immunocompromised mouse model of CCHF. The results show that Ad-N can protect mice from CCHF mortality and that this platform should be considered for future CCHFV vaccine strategies.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever, Crimean/prevention & control , Nucleocapsid Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Female , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/immunology , Hemorrhagic Fever, Crimean/virology , Humans , Male , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
5.
J Immunol Res ; 2014: 160316, 2014.
Article in English | MEDLINE | ID: mdl-24982920

ABSTRACT

Schmallenberg virus (SBV), discovered in continental Europe in late 2011, causes mild clinical signs in adult ruminants, including diarrhoea and reduced milk yield. However, fetal infection can lead to severe malformation in newborn offspring. To develop improved reagents for SBV serology, a high-level yeast expression system was employed to produce recombinant SBV nucleocapsid (N) protein. Recombinant SBV N protein was investigated as an antigen in SBV-specific IgG enzyme immunoassay and used for generation of monoclonal antibodies (MAbs). Yeast-expressed SBV N protein was reactive with anti-SBV IgG-positive cow serum specimens collected from different farms of Lithuania. After immunization of mice with recombinant SBV N protein, four MAbs were generated. The MAbs raised against recombinant SBV N protein reacted with native viral nucleocapsids in SBV-infected BHK cells by immunofluorescence assay. The reactivity of recombinant N protein with SBV-positive cow serum specimens and the ability of the MAbs to recognize virus-infected cells confirm the antigenic similarity between yeast-expressed SBV N protein and native viral nucleocapsids. Our study demonstrates that yeast expression system is suitable for high-level production of recombinant SBV N protein and provides the first evidence on the presence of SBV-specific antibodies in cow serum specimens collected in Lithuania.


Subject(s)
Bunyaviridae Infections/veterinary , Cattle Diseases/diagnosis , Nucleocapsid Proteins/biosynthesis , Orthobunyavirus/isolation & purification , Recombinant Proteins/biosynthesis , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Bunyaviridae Infections/diagnosis , Bunyaviridae Infections/epidemiology , Bunyaviridae Infections/immunology , Cattle , Cattle Diseases/epidemiology , Cattle Diseases/immunology , Cattle Diseases/virology , Cell Line , Cricetinae , Female , Gene Expression , Immune Sera , Immunization , Immunoglobulin G/biosynthesis , Lithuania/epidemiology , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Orthobunyavirus/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
6.
Viral Immunol ; 25(1): 55-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22225471

ABSTRACT

Infectious bronchitis virus (IBV) is prevented primarily by the use of live attenuated vaccines, which are known to have a limited strain range of protection. Alternative vaccines against the emerging new virus strains can improve control of the disease. The aim of this study was to evaluate the immunogenic potential of two recombinant viral proteins, when administered by eyedrop, without the assistance of a vector. The recombinant S1 (rS1) and N (rN) proteins of the M41 strain expressed in E. coli were tested, and the live attenuated vaccine H120 was used as a positive control. Protection was evaluated by re-isolation of virus from tracheas of vaccinated chickens after challenge with strain M41. After three immunizations, rS1 glycoprotein induced 40% protection, while vaccination with rN provided no protection. Vaccination with rS1, rN, or H120 induced a cellular immune response as demonstrated by in vitro ChIFN-γ production by splenocytes of vaccinated birds. Vaccination with H120, and to a lesser extent rS1, induced HI and virus-specific IgG antibody production. These findings indicate that recombinant viral proteins administered through the mucosal route can evoke an immune response without the assistance of a vector.


Subject(s)
Infectious bronchitis virus/immunology , Membrane Glycoproteins/immunology , Nucleocapsid Proteins/immunology , Poultry Diseases/immunology , Recombinant Proteins/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Administration, Mucosal , Animals , Antibodies, Viral/blood , Chickens , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Nucleocapsid Proteins , Infectious bronchitis virus/metabolism , Interferon-gamma/biosynthesis , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/genetics , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Poultry Diseases/prevention & control , Poultry Diseases/virology , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Spike Glycoprotein, Coronavirus , Vaccination , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
7.
Appl Microbiol Biotechnol ; 93(6): 2437-46, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22134641

ABSTRACT

To develop effective mucosal vaccine formulation against porcine epidemic diarrhea virus (PEDV) infection, the DNA fragments encoding spike protein immunodominant region S1 and nucleocapsid N of PEDV were inserted into pPG1 (surface-displayed) or pPG2 (secretory) plasmids followed by electrotransformation into Lactobacillus casei (Lc) to yield four recombinant strains: PG1-S1, PG2-S1, PG1-N, and PG2-N. After intragastric administration, it was observed that live Lc-expressing S1 protein combined with Lc-expressing N protein could elicit much more potent mucosal and systemic immune responses than the former alone (P < 0.001), however slightly inferior to the latter alone (P > 0.05). Furthermore, the surface-displayed mixture (PG1-S1+ PG1-N) revealed stronger immunogenicity than the secretory mixture (PG2-S1+ PG2-N) as well as PEDV-neutralizing potency in vitro (P < 0.001). On 49th day after the last immunization, splenocytes were prepared from mice immunized with surface-displayed mixture, secretory mixture and negative control to be stimulated by purified N and S protein, respectively. The results of ELISA analysis showed that N protein was capable of inducing a higher level of IL-4 (P < 0.001) and IFN-γ (P < 0.001) than S1 protein in the immunized mice. Taken together, Lc-expressed N protein as molecular adjuvant or immunoenhancer was able to effectively facilitate the induction of mucosal and systemic immune responses by Lc-expressing S1 region.


Subject(s)
Coronavirus Infections/veterinary , Lactobacillus/genetics , Membrane Glycoproteins/immunology , Nucleocapsid Proteins/immunology , Porcine epidemic diarrhea virus/immunology , Swine Diseases/immunology , Viral Envelope Proteins/immunology , Administration, Oral , Animals , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins , Female , Gene Expression , Immunity, Mucosal , Lactobacillus/metabolism , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Porcine epidemic diarrhea virus/genetics , Spike Glycoprotein, Coronavirus , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
8.
Clin Vaccine Immunol ; 18(1): 113-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21084464

ABSTRACT

Human coronaviruses (HCoVs) are responsible for respiratory tract infections ranging from common colds to severe acute respiratory syndrome. HCoV-NL63 and HCoV-229E are two of the four HCoVs that circulate worldwide and are close phylogenetic relatives. HCoV infections can lead to hospitalization of children, elderly individuals, and immunocompromised patients. Globally, approximately 5% of all upper and lower respiratory tract infections in hospitalized children are caused by HCoV-229E and HCoV-NL63. The latter virus has recently been associated with the childhood disease croup. Thus, differentiation between the two viruses is relevant for epidemiology studies. The aim of this study was to develop a double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA) as a potential tool for identification and differentiation between HCoV-NL63 and HCoV-229E. The nucleocapsid (N) proteins of HCoV-NL63 and HCoV-229E were expressed in an Escherichia coli system and used to immunize mice in order to obtain monoclonal antibodies (MAbs) specific for each virus. Three specific MAbs to HCoV-NL63, one MAb specific to HCoV-229E, and four MAbs that recognized both viruses were obtained. After their characterization, three MAbs were selected in order to develop a differential DAS-ELISA. The described assay could detect up to 3 ng/ml of N protein and 50 50% tissue culture infective doses/ml of virus stock. No cross-reactivity with other human coronaviruses or closely related animal coronaviruses was found. The newly developed DAS-ELISA was species specific, and therefore, it could be considered a potential tool for detection and differentiation of HCoV-NL63 and HCoV-229E infections.


Subject(s)
Antibodies, Monoclonal , Antibody Specificity , Coronavirus 229E, Human/classification , Coronavirus Infections/diagnosis , Coronavirus NL63, Human/classification , Nucleocapsid Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Coronavirus 229E, Human/immunology , Coronavirus Infections/immunology , Coronavirus Infections/microbiology , Coronavirus NL63, Human/immunology , Coronavirus Nucleocapsid Proteins , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Immunization , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/chemistry , Species Specificity
9.
J Immunol ; 181(9): 6337-48, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941225

ABSTRACT

The details of the mechanism by which severe acute respiratory syndrome-associated coronavirus (SARS-CoV) causes severe pneumonia are unclear. We investigated the immune responses and pathologies of SARS-CoV-infected BALB/c mice that were immunized intradermally with recombinant vaccinia virus (VV) that expressed either the SARS-CoV spike (S) protein (LC16m8rVV-S) or simultaneously all the structural proteins, including the nucleocapsid (N), membrane (M), envelope (E), and S proteins (LC16m8rVV-NMES) 7-8 wk before intranasal SARS-CoV infection. The LC16m8rVV-NMES-immunized group exhibited as severe pneumonia as the control groups, although LC16m8rVV-NMES significantly decreased the pulmonary SARS-CoV titer to the same extent as LC16m8rVV-S. To identify the cause of the exacerbated pneumonia, BALB/c mice were immunized with recombinant VV that expressed the individual structural proteins of SARS-CoV (LC16mOrVV-N, -M, -E, -S) with or without LC16mOrVV-S (i.e., LC16mOrVV-N, LC16mOrVV-M, LC16mOrVV-E, or LC16mOrVV-S alone or LC16mOrVV-N + LC16mOrVV-S, LC16mOrVV-M + LC16mOrVV-S, or LC16mOrVV-E + LC16mOrVV-S), and infected with SARS-CoV more than 4 wk later. Both LC16mOrVV-N-immunized mice and LC16mOrVV-N + LC16mOrVV-S-immunized mice exhibited severe pneumonia. Furthermore, LC16mOrVV-N-immunized mice upon infection exhibited significant up-regulation of both Th1 (IFN-gamma, IL-2) and Th2 (IL-4, IL-5) cytokines and down-regulation of anti-inflammatory cytokines (IL-10, TGF-beta), resulting in robust infiltration of neutrophils, eosinophils, and lymphocytes into the lung, as well as thickening of the alveolar epithelium. These results suggest that an excessive host immune response against the nucleocapsid protein of SARS-CoV is involved in severe pneumonia caused by SARS-CoV infection. These findings increase our understanding of the pathogenesis of SARS.


Subject(s)
Nucleocapsid Proteins/immunology , Pneumonia, Viral/immunology , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Cell Line , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins , Cytokines/biosynthesis , Female , Humans , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , RNA, Messenger/biosynthesis , Rabbits , Severe Acute Respiratory Syndrome/pathology , Severe Acute Respiratory Syndrome/virology , Severity of Illness Index , Vero Cells , Viral Vaccines/administration & dosage
10.
Viral Immunol ; 21(1): 49-60, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18355122

ABSTRACT

In Europe, Puumala virus and Dobrava virus are the major hantaviruses that cause hemorrhagic fever with renal syndrome in humans. As hantaviruses can cause diseases with high morbidity and mortality rates, and as to date there is no specific treatment, efforts are concentrated on the development of vaccines. In this study we characterized the immunogenicity of recombinant nucleocapsid proteins of Puumala virus (PUUV) linked to a carrier protein corresponding to the outer membrane protein A from Klebsiella pneumoniae (rP40). The rP40 molecule is a novel carrier protein that facilitates exogenous antigen uptake by dendritic cells. We cloned and expressed the recombinant PUUV proteins in the E. coli mutant ICONE 200 using the tryptophan promoter-controlled pTEXmp18 expression vector. All recombinant PUUV proteins were found to be highly immunogenic in NMRI mice after three immunizations of 10 microg each of the protein. Only the truncated construct, P40-Puu118, gave high antibody titers after two vaccinations of 0.2 microg each. Likewise in the challenge experiments in NMRI mice, only the truncated construct P40-Puu118 resulted in 100% protection after three immunizations of 10 microg each. The results suggest that P40-Puu118 in particular is a good candidate for a recombinant vaccine against Puumala virus. All recombinant proteins linked to rP40 induced high antibody responses, indicating that rP40 is a carrier protein with potential for use in other vaccines.


Subject(s)
Hemorrhagic Fever with Renal Syndrome/prevention & control , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Puumala virus/immunology , Animals , Antibodies, Viral/blood , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Hemorrhagic Fever with Renal Syndrome/immunology , Immunization, Secondary , Immunoglobulin G/blood , Lung/virology , Male , Mice , Nucleocapsid Proteins/administration & dosage , Puumala virus/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology
11.
J Virol ; 81(11): 6079-88, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17392374

ABSTRACT

By using a series of overlapping synthetic peptides covering 98% of the amino acid sequence of the nucleocapsid protein (NP) of severe acute respiratory syndrome coronavirus (SARS-CoV), four helper T-cell (Th) epitopes (NP11, residues 11 to 25; NP51, residues 51 to 65; NP61, residues 61 to 75; and NP111, residues 111 to 125) in C57BL mice (H-2(b)), four (NP21, residues 21 to 35; NP91, residues 91 to 105; NP331, residues 331 to 345; and NP351, residues 351 to 365) in C3H mice (H-2(k)), and two (NP81, residues 81 to 95; and NP351, residues 351 to 365) in BALB/c mice (H-2(d)) have been identified. All of these peptides were able to stimulate the proliferation of NP-specific T-cell lines or freshly isolated lymph node cells from mice immunized with recombinant NP. Immunization of mice with synthetic peptides containing appropriate Th epitopes elicited strong cellular immunity in vivo, as evidenced by delayed-type hypersensitivity. Priming with the helper peptides (e.g., NP111 and NP351) significantly accelerated the immune response induced by recombinant NP, as determined by the production of NP-specific antibodies. When fused with a conserved neutralizing epitope (SP1143-1157) from the spike protein of SARS-CoV, NP111 and NP351 assisted in the production of high-titer neutralizing antibodies in vivo. These data provide useful insights regarding immunity against SARS-CoV and have the potential to help guide the design of peptide-based vaccines.


Subject(s)
Epitopes, T-Lymphocyte/chemistry , Immunodominant Epitopes/chemistry , Nucleocapsid Proteins/chemistry , Severe acute respiratory syndrome-related coronavirus/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/isolation & purification , Female , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/isolation & purification , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/isolation & purification , Severe acute respiratory syndrome-related coronavirus/chemistry , Vero Cells
12.
Biotechnol Bioeng ; 96(6): 1183-90, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17004271

ABSTRACT

Previously, we found that baculoviral polyhedrin (Polh) can successfully be used in Escherichia coli as a fusion partner for the expression of special foreign proteins as inclusion bodies, and the resulting, easily isolatable Polh-induced fusion inclusion bodies had almost the same characteristics as the native Polh. Here, we investigated the effects of co-expression of baculoviral FP25 protein on Polh-induced inclusion-body production in an E. coli expression system, as FP25 is known to be involved specifically in polyhedra formation. Using several analytical tools, including SDS-PAGE, pronase proteolysis, solubilization under alkaline conditions, and electron microscopy, we found that co-expressed FP25 was associated with Polh-induced inclusion bodies and that its co-expression led to formation of compact inclusion bodies as well as high production levels. We confirmed that FP25 co-expression induced higher production levels of other heterologous protein, antimicrobial peptide Hal18, fused with aggregation-prone Polh. Therefore, co-expression of baculoviral FP25 can be promisingly used to increase the levels of baculoviral Polh-fused foreign proteins, especially harmful proteins, expressed as inclusion bodies in an E. coli expression system.


Subject(s)
Baculoviridae , Escherichia coli/metabolism , Inclusion Bodies, Viral/metabolism , Nucleocapsid Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/metabolism , Gene Expression Regulation, Bacterial , Protein Engineering/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification
13.
Vaccine ; 24(16): 3100-8, 2006 Apr 12.
Article in English | MEDLINE | ID: mdl-16494977

ABSTRACT

The recombinant nucleocapsid (rN) protein of the coronavirus (CoV) responsible for severe acute respiratory syndrome (SARS) was cloned and expressed in Escherichia coli, extracted from cell lysates containing 6M urea, then purified by Ni(2+)-affinity chromatography. In animal immunogenicity studies, we found that most anti-rN protein antibodies were IgG2a in BALB/c mice vaccinated with rN emulsified in Montanide ISA-51 containing the synthetic oligodeoxynucleotide, CpG. In contrast, anti-rN protein antibodies of mice immunized with rN protein in PBS were found to mainly be IgG1. These results indicated that ISA-51/CpG-formulated rN protein was dramatically biased toward a Th1 immune response. To identify the B-cell immunodominant epitopes of the rN protein in the mouse and monkey, the reactivities of antisera raised against purified rN proteins formulated in ISA-51/CpG were tested with a panel of overlapping synthetic peptides covering the entire N protein sequence. Three immunodominant linear B-cell epitope regions were mapped to residues 166-180, 356-375, and 396-410 of the rN protein. When the reactivities of these peptides were screened with human sera from five SARS patients, peptides corresponding to residues 156-175 reacted strongly with sera from two of the SARS patients. These results indicated that the region around residues 156-175 of the N protein is immunogenic in the mouse, monkey, and human. We found that peptides corresponding to residues 1-30, 86-100, 306-320, and 351-365 contained murine immunodominant T-cell epitopes. To identify functional CTL epitopes of the N protein, BALB/c mice were immunized with peptides containing the H-2K(d) CTL motif emulsified in adjuvant ISA-51/CpG. Using an IFN-gamma secretion cell assay and analysis by flow cytometry, peptides containing residues 81-95 were found to be capable of stimulating both CD4(+) and CD8(+) cell proliferation in vitro. We also only observed that peptides corresponding to residues 336-350 were capable of stimulating IFN-gamma production in T-cell cultures derived from peripheral blood mononuclear cells (PBMCs) of macaques immunized with the rN protein emulsified in ISA/CpG adjuvant. Our current results together with those of others suggest that some immunodominant B-cell and T-cell epitopes are conserved in the mouse, monkey, and human. This information is very important for the development SARS diagnostic kits and a vaccine.


Subject(s)
Nucleocapsid Proteins/immunology , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Escherichia coli/genetics , Female , Humans , Immunodominant Epitopes , Immunoglobulin G/blood , Lymphocyte Subsets/immunology , Macaca , Mannitol/analogs & derivatives , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/isolation & purification , Oleic Acids , Oligodeoxyribonucleotides , Severe acute respiratory syndrome-related coronavirus/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
14.
Vaccine ; 22(29-30): 4029-34, 2004 Sep 28.
Article in English | MEDLINE | ID: mdl-15364453

ABSTRACT

Dobrava hantavirus (DOBV) causes a severe form of hemorrhagic fever with renal syndrome (HFRS) for which there is no therapy or vaccine available. We compared the immunogenicity and protective efficacy of recombinant DOBV nucleocapsid protein (rDOBV N) given with Alum or Freund's as adjuvant, or PBS, in C57/BL6 mice. All mice given Alum or Freund's seroconverted as did 6/8 mice given rDOBV N with PBS. Reciprocal geometric mean total IgG-titers were 5380, 18,100, and 800, respectively, while the mean IgG1/IgG2a ratios were 17.5, 9.25, and 12, respectively. Furthermore, ELIspot assays showed higher levels of IL-4 producing peripheral blood mononuclear cells (PBMCs) in the group given Alum as compared to the other groups. Interestingly, only mice receiving rDOBV N with Freund's adjuvant were protected from challenge (75% protected), indicating that the strong Th2-type of immune response induced by Alum against rDOBV N did not induce protection in mice.


Subject(s)
Freund's Adjuvant , Hantavirus Infections/prevention & control , Nucleocapsid Proteins/immunology , Orthohantavirus/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic , Alum Compounds , Animals , Antibodies, Viral/blood , Disease Models, Animal , Female , Hantavirus Infections/immunology , Immunoglobulin G/blood , Interleukin-2/analysis , Interleukin-4/analysis , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Vaccination , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage
15.
Vopr Virusol ; 47(2): 28-31, 2002.
Article in Russian | MEDLINE | ID: mdl-12046464

ABSTRACT

Recombinant fragment of Lassa virus (strain Josiah) nucleocapsid protein (corresponding to amino acid residues 141 + 569) constructed by Dr. Jan ter Meulen (Tropenmedizine, Hamburg) was used for immunizing CBA/calac mice. The preparation was injected intraperitoneally twice with 2-week interval in a dose of 10 micrograms. The parameters of the resultant specific humoral and cell-mediated immunity were comparable to those in reference animals immunized with inactivated Lassa virus. Challenge with Lassa virus (10,000 PFU) resulted in 100% death of the reference animals, while of 15 animals immunized with the recombinant NP protein 8 survived.


Subject(s)
Antibodies, Viral/analysis , Lassa Fever/prevention & control , Lassa virus/immunology , Nucleocapsid Proteins/immunology , Spleen/immunology , Animals , Immunization Schedule , Immunoenzyme Techniques , Injections, Intraperitoneal , Lassa Fever/immunology , Lymphocyte Activation , Mice , Mice, Inbred CBA , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Recombinant Proteins/immunology
16.
J Virol ; 74(24): 11935-49, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090194

ABSTRACT

Molecular clones were constructed that express nucleocapsid (NC) deletion mutant simian immunodeficiency viruses (SIVs) that are replication defective but capable of completing virtually all of the steps of a single viral infection cycle. These steps include production of particles that are viral RNA deficient yet contain a full complement of processed viral proteins. The mutant particles are ultrastructurally indistinguishable from wild-type virus. Similar to a live attenuated vaccine, this approach should allow immunological presentation of a full range of viral epitopes, without the safety risks of replicating virus. A total of 11 Macaca nemestrina macaques were inoculated with NC mutant SIV expressing DNA, intramuscularly (i.m.) in one study and i.m. and subcutaneously in another study. Six control animals received vector DNA lacking SIV sequences. Only modest and inconsistent humoral responses and no cellular immune responses were observed prior to challenge. Following intravenous challenge with 20 animal infectious doses of the pathogenic SIV(Mne) in a long-term study, all control animals became infected and three of four animals developed progressive SIV disease leading to death. All 11 NC mutant SIV DNA-immunized animals became infected following challenge but typically showed decreased initial peak plasma SIV RNA levels compared to those of control animals (P = 0.0007). In the long-term study, most of the immunized animals had low or undetectable postacute levels of plasma SIV RNA, and no CD4(+) T-cell depletion or clinical evidence of progressive disease, over more than 2 years of observation. Although a subset of immunized and control animals were boosted with SIV(Mne) proteins, no apparent protective benefit was observed. Immunization of macaques with DNA that codes for replication-defective but structurally complete virions appears to protect from or at least delay the onset of AIDS after infection with a pathogenic immunodeficiency virus. With further optimization, this may be a promising approach for vaccine development.


Subject(s)
Macaca nemestrina/immunology , Macaca nemestrina/virology , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus , Animals , DNA, Viral/genetics , DNA, Viral/immunology , Mutation , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , SAIDS Vaccines/genetics , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology
17.
Virology ; 246(1): 134-44, 1998 Jun 20.
Article in English | MEDLINE | ID: mdl-9657001

ABSTRACT

DNA vaccines expressing the envelope glycoprotein (GP) or nucleocapsid protein (NP) genes of Ebola virus were evaluated in adult, immunocompetent mice. The vaccines were delivered into the skin by particle bombardment of DNA-coated gold beads with the Powderject-XR gene gun. Both vaccines elicited antibody responses as measured by ELISA and elicited cytotoxic T cell responses as measured by chromium release assays. From one to four vaccinations with 0.5 microgram of the GP DNA vaccine resulted in a dose-dependent protection from Ebola virus challenge. Maximal protection (78% survival) was achieved after four vaccinations. Mice were completely protected with a priming dose of 0.5 microgram of GP DNA followed by three or four subsequent vaccinations with 1.5 micrograms of DNA. Partial protection could be observed for at least 9 months after three immunizations with 0.5 microgram of the GP DNA vaccine. Comparing the GP and NP vaccines indicated that approximately the same level of protection could be achieved with either vaccine.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola/prevention & control , Vaccines, DNA , Viral Structural Proteins , Viral Vaccines , Animals , Antibodies, Viral/blood , Cloning, Molecular , Ebolavirus/genetics , Ebolavirus/immunology , Female , Gene Expression , Genes, Viral/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Sequence Analysis, DNA , T-Lymphocytes, Cytotoxic , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Plaque Assay , Viral Structural Proteins/administration & dosage , Viral Structural Proteins/genetics , Viral Structural Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , Viremia/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...