Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Arch Virol ; 150(9): 1797-812, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15883656

ABSTRACT

In recent years interest in the family Iridoviridae has been renewed by the identification of a number of viruses, particularly from the genus Ranavirus, associated with disease in a range of poikilotherms. Ranaviruses have been isolated from amphibian, piscine and reptilian species. Here we describe an open reading frame (ORF) identified in the genome of Bohle iridovirus (BIV) which contains a nucleotide binding motif conserved within the thymidine kinase (TK) genes of iridoviruses from other genera (lymphocystis disease virus, LCDV, type species of the genus Lymphocystivirus; Chilo iridescent virus, CIV, type species of the genus Iridovirus). The ability of this putative gene to express a functional TK was confirmed by rescue of a TK negative mutant vaccinia virus in the presence of selective media, when expression was controlled by a vaccinia virus promoter. The sequence of the BIV TK was compared with the homologous sequences from epizootic haematopoietic necrosis virus (EHNV), a virus associated with disease in fish, from Wamena iridovirus (WIV) associated with systemic disease in green pythons, and from frog virus 3 (FV3) the ranavirus type species. Comparisons between these sequences and those available from other ranaviruses, other iridoviruses, other DNA viruses and cellular TKs are presented.


Subject(s)
Iridoviridae/genetics , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , Gene Expression , Genetic Vectors , Molecular Sequence Data , Open Reading Frames , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Vaccinia virus/metabolism
2.
J Virol ; 78(24): 13819-28, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564490

ABSTRACT

Further advances are required in understanding protection from AIDS by T-cell immunity. We analyzed a set of multigenic simian/human immunodeficiency virus (SHIV) DNA and fowlpox virus priming and boosting vaccines for immunogenicity and protective efficacy in outbred pigtail macaques. The number of vaccinations required, the effect of DNA vaccination alone, and the effect of cytokine (gamma interferon) coexpression by the fowlpox virus boost was also studied. A coordinated induction of high levels of broadly reactive CD4 and CD8 T-cell immune responses was induced by sequential DNA and fowlpox virus vaccination. The immunogenicity of regimens utilizing fowlpox virus coexpressing gamma interferon, a single DNA priming vaccination, or DNA vaccines alone was inferior. Significant control of a virulent SHIV challenge was observed despite a loss of SHIV-specific proliferating T cells. The outcome of challenge with virulent SHIV(mn229) correlated with vaccine immunogenicity except that DNA vaccination alone primed for protection almost as effectively as the DNA/fowlpox virus regimen despite negligible immunogenicity by standard assays. These studies suggest that priming of immunity with DNA and fowlpox virus vaccines could delay AIDS in humans.


Subject(s)
AIDS Vaccines/immunology , Fowlpox virus/immunology , HIV Infections/prevention & control , Immunization , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV-1/genetics , HIV-1/immunology , Humans , Immunization Schedule , Immunization, Secondary , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation , Macaca nemestrina , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Treatment Outcome , Vaccines, DNA/administration & dosage
3.
J Wildl Dis ; 38(2): 239-52, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12038121

ABSTRACT

Ten juvenile green pythons (Chondropython viridis) died or were euthanized shortly after having been illegally imported into Australia from Indonesia in 1998. Histologic examination of two of the three snakes that died revealed moderately severe chronic ulceration of the nasal mucosa and focal or periacinar degeneration and necrosis of the liver. In addition there was severe necrotizing inflammation of the pharyngeal submucosa accompanied by numerous macrophages, heterophils, and edema. An iridovirus was isolated in culture from several tissues and characterized by immunohistochemistry, electron microscopy, enzyme-linked immunosorbent Assay, polyacrylamide gel electrophoresis, polymerase chain reaction and sequence analysis, restriction endonuclease digestion, and DNA hybridization. This is the first report of a systemic ranavirus infection in any species of snake and is a new member of the genus, Ranavirus.


Subject(s)
Boidae/virology , RNA Virus Infections/veterinary , Ranavirus/isolation & purification , Amino Acid Sequence , Animals , Base Sequence , Capsid/chemistry , Capsid/genetics , Cell Line , Cytopathogenic Effect, Viral , DNA, Viral/analysis , DNA, Viral/chemistry , Electrophoresis, Polyacrylamide Gel/veterinary , Enzyme-Linked Immunosorbent Assay/veterinary , Immunohistochemistry/veterinary , Indonesia , Liver/pathology , Microscopy, Electron/veterinary , Molecular Sequence Data , Nasal Mucosa/pathology , Pharynx/pathology , Phylogeny , Queensland , RNA Virus Infections/pathology , RNA Virus Infections/virology , Ranavirus/classification , Ranavirus/genetics , Ranavirus/ultrastructure , Restriction Mapping/veterinary , Sequence Alignment/veterinary
4.
Vet Microbiol ; 80(2): 101-19, 2001 May 21.
Article in English | MEDLINE | ID: mdl-11295331

ABSTRACT

Weaned pigs (6-week-old) and 7-day-old pre-weaned piglets were vaccinated with naked plasmid DNA expressing the gp55/E2 gene from classical swine fever virus (CSFV). Both groups of pigs were then given a booster dose of recombinant porcine adenovirus expressing the gp55 gene (rPAV-gp55). Following challenge with CSFV, 100% of weaned pigs and 75% pre-weaned piglets were protected from disease. Weaned pigs given a single dose of rPAV-gp55 were also protected, but showed a slight increase in temperature immediately post-challenge. However, weaned animals given a DNA prime before rPAV-gp55 showed no fluctuation in body temperature following challenge and no pathology in spleen or lymph nodes upon post-mortem. In addition, no CSFV could be re-isolated from the rPAV vaccinated group and from only one pig in the prime-boost group following challenge, suggesting that both vaccination regimes have the potential to reduce or prevent virus shedding following experimental challenge.


Subject(s)
Adenoviridae , Classical Swine Fever/prevention & control , Vaccination/veterinary , Vaccines, DNA , Animals , Body Temperature , DNA, Recombinant/administration & dosage , Enzyme-Linked Immunosorbent Assay/veterinary , Swine , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Weaning
5.
Immunol Cell Biol ; 79(1): 87-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11168628

ABSTRACT

Deoxyribonucleic acid (DNA) vaccination offers the prospect of novel, safe vaccines that can stimulate cell-mediated immunity as well as antibody. An understanding of how and where transfection occurs in vivo will assist strategies to enhance their efficacy. Plasmid DNA encoding for the expression of firefly luciferase was injected intramuscularly into sheep. The DNA was rapidly disseminated and could be detected in efferent lymph within 5 min. At the earliest time that cells were tested separately from lymph plasma (1 h), DNA was found to be present in both the lymph plasma and within lymph cells. Although plasmid DNA could be detected intracellularly in lymph and in muscle tissue at the site of inoculation, no luciferase gene expression could be detected. The results suggest that, in addition to the internalization of plasmid DNA, other factors may limit gene expression, possibly the rate of DNA migration into the nucleus.


Subject(s)
DNA/metabolism , Luciferases/metabolism , Vaccines, DNA/administration & dosage , Animals , DNA/genetics , Humans , Luciferases/genetics , Lymph/metabolism , Muscle, Skeletal/metabolism , Plasmids , Polymerase Chain Reaction , Sheep , Time Factors , Transfection , Vaccines, DNA/immunology , Vaccines, DNA/pharmacokinetics
6.
Vaccine ; 19(1): 68-74, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10924788

ABSTRACT

DNA vaccination has proven to be effective against a number of tumours and microbial diseases. As DNA vaccines are unable to replicate, plasmid copy number per cell is dependent on in vivo transfection efficiency, which is usually quite low. Consequently, immune responses generated are likely to be sub-optimal due to low antigen expression levels in transfected cells. During this study, replicating DNA vaccines delivered intra-epidermally by gene gun, were assessed for their ability to more efficiently generate immune responses in mice. The data demonstrate that, using a polyoma virus-based system of replication, 10-fold less DNA expressing the haemagglutinin gene of influenza virus, was required to stimulate a humoral immune response, compared to an equivalent non-replicating vaccine. This observation suggests that the use of replicating DNA vaccines in some delivery systems may enhance the effectiveness of immune responses.


Subject(s)
Antibodies, Viral/immunology , DNA, Viral/administration & dosage , Polyomavirus/immunology , Vaccines, DNA/administration & dosage , Animals , Antibodies, Viral/blood , Cells, Cultured , DNA, Viral/genetics , DNA, Viral/immunology , Female , Genes, Reporter , Immunity/drug effects , Luciferases/genetics , Mice , Mice, Inbred CBA , Polyomavirus/genetics , Replicon/genetics , Replicon/immunology , Transfection , Vaccines, DNA/immunology
7.
Vaccine ; 18(18): 1932-8, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10699343

ABSTRACT

Classical swine fever virus causes significant mortality and morbidity in commercial piggeries in many countries in Europe and Asia. The protective antigen, gp55, is highly conformation-dependent and thus killed virus or bacterially produced proteins are not protective. This report demonstrates that DNA vaccination with the gene encoding gp55 can provide protective immunity with inoculation of two doses of 25 microg DNA or a single shot of 200 microg. Furthermore, the DNA can be delivered intramuscularly or by a simple spring-loaded needleless inoculator. In addition it is shown that inoculation of the DNA at a single site conveys the same level of immunity as division of the dose between two sites.


Subject(s)
Classical Swine Fever Virus/immunology , Classical Swine Fever/prevention & control , Vaccines, DNA/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/genetics , Animals , Antibodies, Viral/biosynthesis , Classical Swine Fever/immunology , DNA, Viral/administration & dosage , DNA, Viral/genetics , DNA, Viral/immunology , DNA, Viral/therapeutic use , Injections, Intramuscular , Neutralization Tests , Swine , Vaccines, DNA/therapeutic use , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/therapeutic use , Viral Vaccines/administration & dosage
8.
J Gen Virol ; 81(Pt 2): 431-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644842

ABSTRACT

The expression of antigens or other molecules from recombinant vaccinia viruses requires the insertion of coding sequence at specific sites in the viral genome. Here we investigate the influence of two different sites on the level of protein expressed during a viral infection. The level of immune response in mice to vaccinia virus-expressed murine interleukin 2 (IL-2) or IL-4 varied depending on whether the coding sequence was inserted into the vaccinia virus thymidine kinase (tk) gene or into the HindIII F fragment of the viral genome where herpes simplex virus (HSV) tk was used as a selectable marker. In each case the intensity of the response was greater when the relevant gene was expressed from the HindIII F insertion site. In order to quantify these differences a series of recombinant viruses expressing luciferase was constructed. Luciferase activity from coding sequence inserted into the HindIII F fragment was significantly higher than that from the tk gene insertion, provided HSV tk(+) constructs were compared. Insertion of a marker gene (HSV tk) into the HindIII F site with disruption of the F7L open reading frame led to a reduced level of luciferase expressed from the tk insert, despite more than 45 kb of intervening sequence. In mice, luciferase expression was higher from the HindIII F inserted gene than from the tk insert in both lungs and ovaries.


Subject(s)
Vaccinia virus/genetics , Animals , Female , Gene Expression , Genetic Vectors , Interleukin-2/genetics , Interleukin-2/pharmacology , Interleukin-4/genetics , Interleukin-4/pharmacology , Luciferases/genetics , Lung/virology , Mice , Mice, Inbred CBA , Mice, Nude , Ovary/virology , Recombinant Proteins/genetics , Recombination, Genetic , Vaccinia virus/pathogenicity , Virulence/genetics
9.
Arch Virol ; 145(2): 301-31, 2000.
Article in English | MEDLINE | ID: mdl-10752555

ABSTRACT

A total of 30 iridoviruses collected from Australia, South-East Asia, North America, South America and Europe were characterised. With the exception of the South-East Asian iridoviruses all viruses were found to belong to the genus Ranavirus. All viruses, except those originating from South-East Asia, cross-reacted with antisera against epizootic haematopoietic necrosis virus (EHNV). Viruses or virus-infected cells were examined using electron microscopy, SDS PAGE, restriction endonuclease (RE) digestion, DNA hybridisation, and DNA sequencing. Data from RE digestion of genomic DNA, and from the sequencing of PCR products indicated that the viruses generally grouped according to their geographic and taxonomic (i.e. amphibian or fish) origin. The one exception to this was the viruses from the United Kingdom that grouped with the North American ranaviruses. The differences between specified genomic regions were small. To assess the validity of the differences in sequence homology, similar studies were performed with different isolates from two viruses (EHNV and Guatopo virus (GV), collected from different animals at different locations and time). The sequence data showed complete homology for the isolates for any one virus over the 200 and 586 bp regions examined. Collectively, the data showed that the coding region for the major coat protein (MCP) is stable for any one species (e.g. EHNV).


Subject(s)
Amphibians/virology , Fishes/virology , Iridoviridae/classification , Ranavirus/classification , Animals , Antigens, Viral/immunology , Base Sequence , Capsid/chemistry , Capsid/genetics , DNA Restriction Enzymes/metabolism , DNA, Viral/isolation & purification , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay/methods , Fish Diseases/virology , Iridoviridae/genetics , Iridoviridae/immunology , Iridoviridae/ultrastructure , Microscopy, Electron , Molecular Sequence Data , Nucleic Acid Hybridization , Polymerase Chain Reaction , Ranavirus/genetics , Ranavirus/immunology , Ranavirus/ultrastructure , Sequence Analysis, DNA , Viral Proteins/chemistry , Viral Proteins/genetics
10.
Res Vet Sci ; 67(3): 213-21, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10607500

ABSTRACT

This study uses recombinant vaccinia viruses expressing truncated or entire bluetongue virus (BTV) proteins to map the location of epitopes recognized by cytotoxic T lymphocytes (CTL) from Australian merino sheep. The non-structural protein, NS1, was recognised by CTL from all sheep, while VP2, VP3, VP5 and VP7 were recognised by CTL from only some sheep. The remaining proteins (except for VP1, which was not tested) did not contain CTL epitopes. When truncated genes were used to map the location of CTL epitopes, it was found that sheep often have CTL that recognise more than one epitope in NS1 or VP2. Overall there was considerable diversity in the CTL recognition patterns in the sheep tested.


Subject(s)
Bluetongue virus/immunology , Sheep/immunology , T-Lymphocytes, Cytotoxic/virology , Animals , Antigens, Viral/immunology , Capsid/immunology , Capsid Proteins , Cell Line , Epitope Mapping/veterinary , Peptide Mapping/veterinary , Sheep/virology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus , Viral Nonstructural Proteins/immunology
11.
Arch Virol ; 143(7): 1433-9, 1998.
Article in English | MEDLINE | ID: mdl-9722886

ABSTRACT

The coat protein of the potyvirus, Johnsongrass mosaic virus (JGMV), was expressed using a recombinant vaccinia virus (VV) system. Ultra-thin section electron microscopy demonstrated that the coat protein assembled into potyvirus-like particles (PVLPs) in recombinant VV infected cells. Infection of cells with two additional VV recombinants expressing coat protein plus N-terminal and N- and C-terminal extensions also resulted in the formation of PVLPs. These results suggest that the ability of VV to express the potyvirus coat protein at sufficient levels to allow PVLP formation in vitro, could make VV a suitable vector for the delivery of PVLPs displaying vaccine antigens in vivo without the need for particle purification and/or inclusion of adjuvant. Use of such a vaccine strategy would also benefit from the proven advantages of poxviruses as vaccines such as stability in a freeze dried form, resistance to environmental factors and the potential for oral administration.


Subject(s)
Capsid/genetics , Potyvirus/genetics , Potyvirus/physiology , Vaccinia virus/genetics , Animals , Capsid/ultrastructure , Cell Line , Gene Expression , Genetic Vectors , Inclusion Bodies, Viral/ultrastructure , Microscopy, Electron , Potyvirus/immunology , Recombinant Proteins/genetics , Recombinant Proteins/ultrastructure , Recombination, Genetic , Vaccinia virus/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology , Virus Replication
12.
J Virol ; 72(3): 2246-52, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9499082

ABSTRACT

The role of CD4+ and CD8+ cells in the generation of an effective immune response against viral infections is well established. Moreover, there is an increasing realization that subunit vaccines which include both CD4+- and CD8+-T-cell epitopes are highly effective in controlling viral infections, as opposed to those which are designed to activate a CD8+- or CD4+-T-cell response alone. One of the major limitations of epitope-based vaccines designed to stimulate virus-specific CD4+ T cells is that endogenously expressed class II-restricted minimal cytotoxic-T-lymphocyte (CTL) epitopes are poorly recognized by CD4+ CTLs. In the present study we attempted to enhance the efficiency of class II-restricted endogenous presentation of minimal class II-restricted CTL epitopes by specifically targeting a polyepitope protein to class II processing compartments through the endosomal and/or lysosomal pathway. A significantly enhanced stimulation of virus-specific CD4+-T-cell clones by antigen-presenting cells (APC) expressing the recombinant polyepitope protein targeted to the endocytic/secretory pathway was readily demonstrated in cytotoxicity assays. In addition, in vitro activation of Epstein-Barr virus- and influenza virus-specific CD4+ memory CTLs by the recombinant constructs encoding the polyepitope protein, specifically targeted to the lysosomal compartment, was also demonstrated. The enhanced stimulatory capacity of APC expressing a lysosome-targeted polyepitope protein has important implications for vaccine design.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Peptide Fragments/immunology , Proteins/immunology , Viral Vaccines , Antigen Presentation , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , CD4-Positive T-Lymphocytes/cytology , Cell Line, Transformed , Drug Design , Endocytosis , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Genetic Vectors , HeLa Cells , Humans , Immunologic Memory , Lysosomal Membrane Proteins , Lysosomes , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Peptide Fragments/biosynthesis , Protein Biosynthesis , Vaccinia virus
13.
Virology ; 235(2): 367-76, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9281517

ABSTRACT

For baculoviruses and herpesviruses, integration of transposons or retroviruses into the virus genome has been documented. We report here that field and vaccine strains of fowlpox virus (FPV) carry integrated sequences from the avian retrovirus, reticuloendotheliosis virus (REV). Using PCR and hybridization analysis we observed that vaccine and field strains of FPV carry REV sequences integrated into a previously uncharacterized region of the right 1/3 of the FPV genome. Long-range PCR, hybridization, and nucleotide sequence determination demonstrated that one vaccine strain (FPV S) and recently isolated field strains carry a near-full-length REV provirus. For another vaccine strain (FPV M) a rearranged remnant of the LTR was found at the same insertion site. By Western blotting and reverse transcriptase assays we were unable to demonstrate free REV in supernatants of FPV S cultures. The near-full-length REV provirus integrated into the FPV genome is infectious since FPV S DNA gave rise to REV upon transfection into chicken embryo fibroblasts. Upon infection of chickens with FPV S, all chickens developed high-titered antibodies to REV, and REV was isolated from the blood of half of the inoculated chickens. Our observations add to the list of targets for retrovirus integration into DNA virus genomes. The integration of a near-full-length, and apparently infectious, REV provirus into FPV provides additional transmission routes for the retrovirus by way of the infectious cycle of FPV, including the possibility of mechanical transmission by biting insects since FPV is believed to be transmitted by this route. For large DNA viruses, including the poxviruses, retrovirus integration with attendant possibilities of gene transduction may be an important mechanism for virus evolution, including the acquisition of cellular genes with the potential to modify virus virulence and pathogenicity.


Subject(s)
Fowlpox virus/genetics , Proviruses/genetics , Reticuloendotheliosis Viruses, Avian/genetics , Animals , Base Sequence , Blotting, Western , Chick Embryo , Chickens , Cloning, Molecular , Fibroblasts/virology , Molecular Sequence Data , Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid/genetics , Restriction Mapping , Reticuloendotheliosis Viruses, Avian/immunology , Reticuloendotheliosis Viruses, Avian/isolation & purification , Reticuloendotheliosis Viruses, Avian/pathogenicity , Retroviridae/pathogenicity , Sequence Alignment , Vaccines/genetics
14.
J Virol Methods ; 66(1): 135-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9220399

ABSTRACT

A synthetic vaccinia virus promoter (Psel) was constructed based upon sequences which increase activity of the P7.5 early/late promoter. Comparison of luciferase activity in lysates from cells infected with recombinant vaccinia viruses expressing the luciferase gene either under the control of the P7.5 promoter or Psel, demonstrated significantly enhanced activity mediated by Psel at both early and late times post infection. This promoter may be of considerable benefit in the construction of recombinant poxviruses where early foreign gene expression is important for generating a protective immune response in vaccinated animals, or in reporter/target gene expression in vitro.


Subject(s)
Promoter Regions, Genetic , Vaccinia virus/genetics , Genes, Reporter , Genes, Synthetic , Luciferases/genetics
15.
Arch Virol ; 142(4): 737-48, 1997.
Article in English | MEDLINE | ID: mdl-9170501

ABSTRACT

The mild fowlpox vaccine, FPV M, widely used in Australia is composed of two predominant genotypes based upon differences identifiable in restriction enzyme analyses of plaque purified derivatives of this vaccine. The differences, where identifiable, were in the end fragments of the genomes. Five field isolates of FPV from chickens in New South Wales showed restriction enzyme profiles closely related to the more virulent (standard) vaccine strain, FPV S. The FPV S strain differs from FPV M in both terminal genome fragments and in the presence of a PstI fragment of approximately 10kb (this fragment was also present in PstI digests of all of the field isolates). Plaque purified derivatives of FPV M showed similar lesion development upon inoculation into the wing web of chickens. The field isolates showed significantly higher virulence in day-old and three-week-old chickens in comparison with FPV M. One field isolate was similar to the FPV S vaccine. Two isolates had slowly developing wing web lesions, caused significant secondary lesions in three-week-old chickens and generalised poxvirus infection when inoculated into day-old chickens. For two isolates, the primary wing web lesion took even longer to develop and resolve although these isolates did not cause generalised poxvirus infection. It was possible to identify four virulence/pathogenicity types amongst these vaccine and field isolates of FPV. These strains may allow the characterisation of FPV encoded virulence factors. The field strains with higher virulence may be suitable as parent strains for the construction of FPV recombinants with enhanced immune responses to co-expressed vaccine antigens when compared with current FPV M strain based recombinants.


Subject(s)
Avipoxvirus/classification , Viral Vaccines , Animals , Antibodies, Viral/biosynthesis , Avipoxvirus/genetics , Avipoxvirus/immunology , Avipoxvirus/pathogenicity , Cells, Cultured , Chick Embryo , DNA, Viral , Enzyme-Linked Immunosorbent Assay , Restriction Mapping , Virulence
16.
Vet Immunol Immunopathol ; 59(3-4): 293-309, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9477479

ABSTRACT

The role of individual viral proteins in the immune response to bluetongue virus (BTV) is not clearly understood. To investigate the contributions of the outer capsid proteins, VP2 and VP5, and possible interactions between them, these proteins were expressed from recombinant vaccinia viruses either as individual proteins or together in double recombinants, or with the core protein VP7 in a triple recombinant. Comparison of the immunogenicity of the vaccinia expressed proteins with BTV expressed proteins was carried out by inoculation of rabbits and sheep. Each of the recombinants was capable of stimulating an anti-BTV antibody response, although there was a wide range in the level of response between animals and species. Vaccinia-expressed VP2 was poorly immunogenic, particularly in rabbits. VP5, on the whole, stimulated higher ELISA titers in rabbits and sheep and in some animals in both species was able to stimulate virus neutralizing antibodies. When the protective efficacy of VP2 and VP5 was tested in sheep, vaccinia-expressed VP2, VP5 and VP2 + VP5 were protective, with the most consistent protection being in groups immunized with both proteins.


Subject(s)
Antibodies, Viral/biosynthesis , Antigens, Viral/immunology , Bluetongue virus/immunology , Bluetongue/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Antigens, Viral/genetics , Bluetongue/prevention & control , Capsid/genetics , Capsid/immunology , Capsid Proteins , Immunity , Rabbits , Sheep , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Viral Vaccines/genetics
17.
J Immunol ; 157(2): 822-6, 1996 Jul 15.
Article in English | MEDLINE | ID: mdl-8752934

ABSTRACT

Development of epitope-based CD8 alpha beta CTL vaccines requires effective strategies for codelivery of large numbers of individual epitopes. We have designed an artificial "polyepitope" protein containing 10 contiguous minimal CTL epitopes, which were restricted by five MHC alleles and derived from five viruses, a parasite, and a tumor model. A recombinant vaccinia virus coding for this protein was capable of inducing MHC-restricted primary CTL responses to all 10 epitopes. Mice immunized with this recombinant vaccinia showed protection against murine cytomegalovirus, Sendai virus, and a tumor model. This simple generic approach to multiepitope delivery should find application in CTL-based vaccine design.


Subject(s)
Epitopes/genetics , Epitopes/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , Drug Design , Epitopes/administration & dosage , Genetic Vectors/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Protein Engineering/methods , Vaccines, Synthetic/administration & dosage
18.
Vet Immunol Immunopathol ; 48(3-4): 287-98, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8578687

ABSTRACT

A cDNA encoding ovine granulocyte-macrophage colony-stimulating factor (GM-CSF) was isolated and two forms of recombinant ovine GM-CSF were produced. A glycosylated form was produced in mammalian cells infected with a recombinant vaccinia virus encoding ovine GM-CSF. Recombinant ovine GM-CSF was also produced in Escherichia coli and purified by affinity chromatography. Both forms of the protein were detected by ovine GM-CSF-specific monoclonal antibodies, and exhibited activity on ovine bone marrow haemopoetic progenitor cells.


Subject(s)
Escherichia coli/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Sheep/immunology , Vaccinia virus/metabolism , Animals , Antibodies, Monoclonal , Base Sequence , Blotting, Western/veterinary , Bone Marrow Cells , Cell Division , Cell Line , Chromatography, Affinity/veterinary , Cloning, Molecular , Colony-Forming Units Assay/veterinary , DNA Primers/chemistry , DNA, Complementary/isolation & purification , Electrophoresis, Polyacrylamide Gel/veterinary , Enzyme-Linked Immunosorbent Assay/veterinary , Escherichia coli/genetics , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/isolation & purification , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Molecular Sequence Data , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Stem Cells/cytology , Transfection , Vaccinia virus/genetics
19.
Virology ; 198(1): 10-6, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8259644

ABSTRACT

We determined the capacity of vaccinia virus recombinants expressing individual rotavirus genes to induce virus-specific cytotoxic T lymphocytes (CTLs) in mice. Mice were orally inoculated with vaccinia virus recombinants containing genes which encode rotavirus outer capsid proteins vp4 or vp7, single-shelled virus proteins vp1, vp2, or vp6, or rotavirus nonstructural proteins NS53, NS35, NS28, or NS26/NS12. We found that (i) the greatest frequencies of virus-specific CTLs were induced by vaccinia virus recombinants expressing vp7, (ii) transport of vp7 beyond the endoplasmic reticulum was not necessary for induction of CTLs, (iii) recombinants expressing vp7 induced CTLs which reacted with different rotavirus serotypes, and (iv) CTLs were induced among both intestinal and nonintestinal lymphocytes after oral inoculation. These findings may be relevant to vaccine strategies which utilize vectors expressing individual rotavirus genes.


Subject(s)
Genes, Viral , Rotavirus/immunology , T-Lymphocytes, Cytotoxic/microbiology , Vaccinia virus/genetics , Administration, Oral , Animals , Cytotoxicity, Immunologic , Female , Gene Expression Regulation, Viral , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Recombinant Proteins/immunology , Recombination, Genetic , Rotavirus/genetics , Serotyping , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology
20.
Virology ; 189(2): 423-34, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1322583

ABSTRACT

The major inner capsid protein of rotavirus is VP6, a 42-kDa polypeptide that forms the icosahedral surface of the rotavirus single-shelled particle. A chimeric form of VP6 (VP6sc) was constructed containing an upstream leader sequence derived from the influenza virus hemagglutinin and a downstream membrane-spanning (anchor) domain from a mouse immunoglobulin gene. When VP6sc was expressed in cells using a recombinant vaccinia virus, the protein was transported, glycosylated, and anchored in the plasma membrane as a trimer with the major domains of the protein orientated externally. Immunofluorescence and immunolabeling with colloidal gold indicated that VP6sc also localized in patches on the cell surface; electron microscopy revealed that the protein assembled into two-dimensional arrays which exhibited the same periodicity as the paracrystalline arrays formed by purified (viral) VP6. Mice inoculated with a recombinant vaccinia virus that expressed VP6sc produced rotavirus-specific antibodies at a titer 10 times higher than that achieved when wild-type, intracellular VP6 was delivered in the same way. Presentation at the cell surface therefore may represent a general method for enhancing the immunogenicity of rotavirus proteins.


Subject(s)
Antigens, Viral , Capsid Proteins , Capsid/metabolism , Membrane Proteins/metabolism , Rotavirus/ultrastructure , Animals , Base Sequence , Capsid/chemistry , Capsid/immunology , Cell Membrane/metabolism , Cells, Cultured , Chlorocebus aethiops , In Vitro Techniques , Macromolecular Substances , Membrane Proteins/chemistry , Microscopy, Electron , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides/chemistry , Protein Binding , Recombinant Fusion Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...