Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 167(6): 3398-405, 2001 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11544331

RESUMO

Mucosally active vaccine adjuvants that will prime a full range of local and systemic immune responses against defined antigenic epitopes are much needed. Cholera toxin and lipophilic immune stimulating complexes (ISCOMS) containing Quil A can both act as adjuvants for orally administered Ags, possibly by targeting different APCs. Recently, we have been successful in separating the adjuvant and toxic effects of cholera toxin by constructing a gene fusion protein, CTA1-DD, that combines the enzymatically active CTA1-subunit with a B cell-targeting moiety, D, derived from Staphylococcus aureus protein A. Here we have extended this work by combining CTA1-DD with ISCOMS, which normally target dendritic cells and/or macrophages. ISCOMS containing a fusion protein comprising the OVA(323-339) peptide epitope linked to CTA1-DD were highly immunogenic when given in nanogram doses by the s.c., oral, or nasal routes, inducing a wide range of T cell-dependent immune responses. In contrast, ISCOMS containing the enzymatically inactive CTA1-R7K-DD mutant protein were much less effective, indicating that at least part of the activity of the combined vector requires the ADP-ribosylating property of CTA1. No toxicity was observed by any route. To our knowledge, this is the first report on the successful combination of two mechanistically different principles of adjuvant action. We conclude that rationally designed vectors consisting of CTA1-DD and ISCOMS may provide a novel strategy for the generation of potent and safe mucosal vaccines.


Assuntos
Adjuvantes Imunológicos , Antígenos/imunologia , ISCOMs/imunologia , Mucosa/imunologia , Administração Intranasal , Administração Oral , Animais , Anticorpos/sangue , Antígenos/administração & dosagem , Antígenos/química , Subpopulações de Linfócitos B/imunologia , Toxina da Cólera/administração & dosagem , Toxina da Cólera/genética , Toxina da Cólera/imunologia , Dimerização , Relação Dose-Resposta Imunológica , Vetores Genéticos/genética , ISCOMs/administração & dosagem , Imunização/métodos , Injeções Subcutâneas , Linfonodos/imunologia , Mesentério/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/administração & dosagem , Ovalbumina/química , Ovalbumina/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Nódulos Linfáticos Agregados/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Organismos Livres de Patógenos Específicos
2.
Vaccine ; 19(27): 3661-70, 2001 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-11395200

RESUMO

A vaccine based upon a recombinant plant virus (CPMV-PARVO1), displaying a peptide derived from the VP2 capsid protein of canine parvovirus (CPV), has previously been described. To date, studies with the vaccine have utilized viable plant chimaeric particles (CVPs). In this study, CPMV-PARVO1 was inactivated by UV treatment to remove the possibility of replication of the recombinant plant virus in a plant host after manufacture of the vaccine. We show that the inactivated CVP is able to protect dogs from a lethal challenge with CPV following parenteral immunization with the vaccine. Dogs immunized with the inactivated CPMV-PARVO1 in adjuvant displayed no clinical signs of disease and shedding of CPV in faeces was limited following CPV challenge. All immunized dogs elicited high titres of peptide-specific antibody, which neutralized CPV in vitro. Levels of protection, virus shedding and VP2-specific antibody were comparable to those seen in dogs immunized with the same VP2- peptide coupled to keyhole limpet hemocyanin (KLH). Since plant virus-derived vaccines have the potential for cost-effective manufacture and are not known to replicate in mammalian cells, they represent a viable alternative to current replicating vaccine vectors for development of both human and veterinary vaccines.


Assuntos
Comovirus/imunologia , Infecções por Parvoviridae/prevenção & controle , Parvovirus Canino/imunologia , Proteínas Recombinantes/uso terapêutico , Proteínas Virais/uso terapêutico , Vacinas Virais/uso terapêutico , Sequência de Aminoácidos , Animais , Capsídeo/uso terapêutico , Proteínas do Capsídeo , Comovirus/efeitos da radiação , Doenças do Cão/prevenção & controle , Doenças do Cão/virologia , Cães , Esquemas de Imunização , Dados de Sequência Molecular , Infecções por Parvoviridae/mortalidade , Infecções por Parvoviridae/veterinária , Parvovirus Canino/efeitos da radiação , Raios Ultravioleta , Vacinas de Produtos Inativados/uso terapêutico , Vacinas Sintéticas/uso terapêutico
3.
Vaccine ; 18(28): 3254-65, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10869770

RESUMO

Feline immunodeficiency virus (FIV) is a natural lentiviral pathogen of cats which can be experimentally transmitted via rectal and vaginal routes--the major routes of human immunodeficiency virus type 1 transmission in man. An important objective for lentiviral research is the development of vaccine strategies which generate good mucosal immune responses capable of giving protection from a mucosal virus challenge. The experimental vaccines employed in this study were based on (a) a peptide from the third variable region of the FIV envelope glycoprotein and (b) fixed whole FIV, Glasgow-8 strain. Adjuvants used were Quil A and cholera toxin for mucosal administration and incomplete Freund's adjuvant and immune stimulating complexes for subcutaneous injection. Mucosal immunization was given by rectal and intranasal routes. Both antibody and proliferative responses were elicited by mucosal immunization and cholera toxin was found to be a good mucosal adjuvant. The addition of a lipo thioester to the FIV peptide improved IgG and IgA responses upon parenteral administration. However, no protection from a rectal FIV challenge was achieved.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Imunodeficiência Felina/imunologia , Ativação Linfocitária , Reto/virologia , Vacinas Virais/imunologia , Administração Intranasal , Administração Retal , Sequência de Aminoácidos , Animais , Gatos , Imunização , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Dados de Sequência Molecular , Linfócitos T/imunologia , Vacinas Virais/administração & dosagem
4.
Vaccine ; 18(23): 2482-93, 2000 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-10775782

RESUMO

Semipurified Quil A and purified Quil A were used to prepare well-characterized subunit vaccine candidates against measles. Variation in the relative amounts of the measles virus (MV) fusion (F) protein, Quil A-components and lipids did not influence induction of antibody responses in mice, but had a pronounced effect on the capacity to induce cytotoxic T cell (CTL) activity of a CD8(+) MV F-protein specific human T cell clone in vitro. A characteristic MV iscom preparation based on the combined use of HPLC-purified Quil A-components QA-3 and QA-22 (QA-3/22) efficiently induced CTL activity in vitro. Comparable results were obtained by mixing beta-propiolactone inactivated MV with iscom-matrix QA-3/22 or free QA-22. On the basis of the data presented it was concluded that these three preparations are interesting MV vaccine candidates for further evaluation in pre-clinical experiments in a primate model.


Assuntos
Adjuvantes Imunológicos/química , Anticorpos Antivirais/biossíntese , Hemaglutininas Virais/imunologia , ISCOMs/imunologia , Ativação Linfocitária , Vacina contra Sarampo/imunologia , Vírus do Sarampo/imunologia , Saponinas/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais de Fusão/imunologia , Adjuvantes Imunológicos/isolamento & purificação , Animais , Células Cultivadas , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão , Citotoxicidade Imunológica , Humanos , ISCOMs/química , Vacina contra Sarampo/química , Vírus do Sarampo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Propiolactona/farmacologia , Saponinas de Quilaia , Saponinas/química , Vacinação , Vacinas Atenuadas/imunologia , Células Vero
5.
Vaccine ; 18(21): 2244-9, 2000 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-10717344

RESUMO

Immunisation against pathogens remains one of the most effective ways of preventing or reducing losses due to infectious diseases in animal husbandry. When inactivated vaccines are used, adjuvants are most often required to obtain satisfactory immune responses. One such type of adjuvant is saponin derived from the bark of Quillaja saponaria Molina, a tree of the rose family. A few different commercial sources exist, but due to the structural complexity and heterogeneity of these saponin preparations, it has been difficult to establish exactly which components are responsible for the adjuvant activity. By carefully selecting the bark source, we have succeeded in preparing a much less heterogeneous preparation of quillaja saponin. In this report we describe the preparation, in terms of structural complexity, hemolytic activity, adjuvant activity, and its ability to form ISCOM matrix. This new preparation could have implications for use per se, or as starting material for more effective preparation of pure substances.


Assuntos
Adjuvantes Imunológicos/isolamento & purificação , Plantas Medicinais/química , Saponinas/isolamento & purificação , Animais , Cromatografia Líquida de Alta Pressão , ISCOMs/isolamento & purificação , ISCOMs/farmacologia , Camundongos , Saponinas de Quilaia , Saponinas/farmacologia , Ovinos
6.
Virology ; 257(2): 449-59, 1999 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-10329555

RESUMO

African horse sickness virus (AHSV) causes a fatal disease in horses. The virus capsid is composed of a double protein layer, the outermost of which is formed by two proteins: VP2 and VP5. VP2 is known to determine the serotype of the virus and to contain the neutralizing epitopes. The biological function of VP5, the other component of the capsid, is unknown. In this report, AHSV VP5, expressed in insect cells alone or together with VP2, was able to induce AHSV-specific neutralizing antibodies. Moreover, two VP5-specific monoclonal antibodies (MAbs) that were able to neutralize the virus in a plaque reduction assay were generated. To dissect the antigenic structure of AHSV VP5, the protein was cloned in Escherichia coli using the pET3 system. The immunoreactivity of both MAbs, and horse and rabbit polyclonal antisera, with 17 overlapping fragments from VP5 was analyzed. The most immunodominant region was found in the N-terminal 330 residues of VP5, defining two antigenic regions, I (residues 151-200) and II (residues 83-120). The epitopes were further defined by PEPSCAN analysis with 12mer peptides, which determined eight antigenic sites in the N-terminal half of the molecule. Neutralizing epitopes were defined at positions 85-92 (PDPLSPGE) for MAb 10AE12 and at 179-185 (EEDLRTR) for MAb 10AC6. Epitope 10AE12 is highly conserved between the different orbiviruses. MAb 10AE12 was able to recognize bluetongue virus VP5 and epizootic hemorrhagic disease virus VP5 by several techniques. These data will be especially useful for vaccine development and diagnostic purposes.


Assuntos
Vírus da Doença Equina Africana/imunologia , Antígenos Virais/imunologia , Vírus Bluetongue/imunologia , Capsídeo/imunologia , Epitopos de Linfócito B/imunologia , Vírus da Doença Hemorrágica Epizoótica/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Antivirais/biossíntese , Antígenos Virais/genética , Capsídeo/genética , Proteínas do Capsídeo , Chlorocebus aethiops , Reações Cruzadas , Mapeamento de Epitopos , Epitopos de Linfócito B/genética , Escherichia coli , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Testes de Neutralização , Peptídeos/síntese química , Peptídeos/imunologia , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Sorotipagem , Células Vero
7.
Vaccine ; 17(9-10): 1057-64, 1999 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-10195615

RESUMO

Bovine Viral Diarrhea Virus (BVDV) is a major pathogen of cattle in most countries. The main reservoir of virus in herds are BVDV persistently infected animals, which arise as a result of infection of the bovine fetus early in gestation. The spread of virus to the unborn fetus may be prevented by vaccination of the dam. We describe in this report the production and initial testing of an inactivated subunit vaccine against BVDV. The vaccine is based on production of antigen in primary bovine cell cultures, extraction of antigens from infected cells with detergent, chromatographic purification, concentration, and insertion of antigens into immune stimulating complexes (ISCOMs). Vaccines based on two different Danish strains of BVDV were injected into calves and the antisera produced were tested for neutralising activity against a panel of Danish BVDV strains. The two vaccines induced different neutralisation responses, which seem to partly complement each other. The implication of these observations for successful vaccination against BVDV is discussed.


Assuntos
Adjuvantes Imunológicos/síntese química , Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Vírus da Diarreia Viral Bovina/imunologia , ISCOMs/imunologia , Animais , Anticorpos Antivirais/biossíntese , Bovinos , Células Cultivadas , Reações Cruzadas , Eletroforese em Gel de Poliacrilamida
8.
J Virol ; 73(2): 930-8, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9882293

RESUMO

The humoral immune responses to the D2 peptide of fibronectin-binding protein B (FnBP) of Staphylococcus aureus, expressed on the plant virus cowpea mosaic virus (CPMV), were evaluated after mucosal delivery to mice. Intranasal immunization of these chimeric virus particles (CVPs), either alone or in the presence of ISCOM matrix, primed CPMV-specific T cells and generated high titers of CPMV- and FnBP-specific immunoglobulin G (IgG) in sera. Furthermore, CPMV- and FnBP-specific IgA and IgG could also be detected in the bronchial, intestinal, and vaginal lavage fluids, highlighting the ability of CVPs to generate antibody at distant mucosal sites. IgG2a and IgG2b were the dominant IgG subclasses in sera to both CPMV and FnBP, demonstrating a bias in the response toward the T helper 1 type. The sera completely inhibited the binding of human fibronectin to the S. aureus FnBP. Oral immunization of the CVPs also generated CPMV- and FnBP-specific serum IgG; however, these titers were significantly lower and more variable than those generated by the intranasal route, and FnBP-specific intestinal IgA was undetectable. Neither the ISCOM matrix nor cholera toxin enhanced these responses. These studies demonstrate for the first time that recombinant plant viruses have potential as mucosal vaccines without the requirement for adjuvant and that the nasal route is most effective for the delivery of these nonreplicating particles.


Assuntos
Adesinas Bacterianas , Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Proteínas de Transporte/imunologia , Comovirus/imunologia , Fibronectinas/metabolismo , Imunidade nas Mucosas , Staphylococcus aureus/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Administração Oral , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/genética , Vacinas Bacterianas/genética , Proteínas de Transporte/genética , Toxina da Cólera/imunologia , Comovirus/genética , Feminino , Humanos , ISCOMs/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Imunoglobulina G/imunologia , Intestinos/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Staphylococcus aureus/genética , Linfócitos T/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vagina/imunologia , Vírion
9.
Vet Q ; 20 Suppl 3: S92-5, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9689737

RESUMO

The ultimate vaccine is an oral vaccine which given once protects against a multitude of diseases. Furthermore this ultimate vaccine needs to be very stable and inexpensive to produce. Probably this latter condition can be met only if the vaccines are produced in plants. Such vaccines are called 'edible vaccines'. Edible vaccines can be produced in plants in many ways. Using recombinant plantvirus, CPMV, it was shown that plants can produce massive amounts of chimaeric virus particles which protect after a single injection the target animal against disease. The final step, oral administration, is being addressed at present. Preliminary experiments by others suggest that this step may be solved sooner than expected.


Assuntos
Plantas Comestíveis/imunologia , Vacinação/tendências , Vacinas Sintéticas/normas , Sequência de Aminoácidos , Animais , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Capsídeo/química , Capsídeo/imunologia , Comovirus/genética , Comovirus/imunologia , Doenças do Cão/prevenção & controle , Cães , Vírus da Panleucopenia Felina/genética , Vírus da Panleucopenia Felina/imunologia , Engenharia Genética , Humanos , Vison , Dados de Sequência Molecular , Infecções por Parvoviridae/prevenção & controle , Infecções por Parvoviridae/veterinária , Parvovirus Canino/genética , Parvovirus Canino/imunologia , Plantas Geneticamente Modificadas/imunologia , Vacinas Sintéticas/administração & dosagem
10.
J Gen Virol ; 79 ( Pt 8): 1901-9, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9714238

RESUMO

Rabbit haemorrhagic disease virus (RHDV) causes an important disease in rabbits. The virus capsid is composed of a single 60 kDa protein. The capsid protein gene was cloned in Escherichia coli using the pET3 system, and the antigenic structure of RHDV VP60 was dissected using 11 monoclonal antibodies (MAbs) and 12 overlapping fragments of the protein expressed in E. coli. Two antigenic regions were found. Ten out of the 11 MAbs recognized different discontinuous epitopes in the most immunodominant region of the viral capsid. This domain was located between residues 31 and 250 of the VP60 N terminus. The other MAb revealed the presence of an antigenic site within 102 aa of the C terminus. This MAb did not recognize the major cleavage product of the full-length 60 kDa protein. These results indicate that, in contrast to other caliciviruses such as Norwalk virus (NV), the 36 kDa cleavage product probably forms the N-terminal region of VP60. However, as in NV, the cleavage region appears to be the most immunodominant region.


Assuntos
Antígenos Virais/imunologia , Capsídeo/imunologia , Epitopos de Linfócito B/imunologia , Vírus da Doença Hemorrágica de Coelhos/imunologia , Proteínas Estruturais Virais/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Antígenos Virais/genética , Ligação Competitiva , Capsídeo/genética , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Immunoblotting , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Spodoptera , Proteínas Estruturais Virais/genética
11.
Virus Res ; 53(2): 163-73, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9620208

RESUMO

The antigenic structure of the capsid proteins of porcine parvovirus (PPV) was investigated. A total of nine linear epitopes were identified by Pepscan using porcine or rabbit anti-PPV antisera. No sites were identified with a panel of neutralising monoclonal antibodies (MAbs). All epitopes were located in the region corresponding to the major capsid protein VP2. Based on this information, and on analogy to other autonomous parvoviruses, 24 different peptides were synthesised, coupled to keyhole limpet haemocyanin (KLH) and used to immunise rabbits. Most antisera were able to bind viral protein. Only peptides from the N-terminal part of VP2 were able to induce virus-neutralising antibodies, although at low levels. A similar neutralising activity could be obtained in pigs. The exposure of the N-terminus was shown in full virions, both by immunoelectron microscopy and absorption experiments. It is concluded that in PPV, the VP2 N-terminus is involved in virus neutralisation (VN) and peptides from this region are therefore primary targets for developing peptide-based vaccines against this virus.


Assuntos
Capsídeo/imunologia , Mapeamento de Epitopos , Parvovirus/imunologia , Peptídeos/imunologia , Suínos/virologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Proteínas do Capsídeo , Ensaio de Imunoadsorção Enzimática , Imunização , Dados de Sequência Molecular , Peptídeos/síntese química , Coelhos
12.
J Pept Res ; 50(5): 357-64, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9401920

RESUMO

Synthetic peptides have frequently been used to immunize animals. However, peptides less than about 20 to 30 amino acids long are poor immunogens. In general, to increase its immunogenicity, the presentation of the peptide should be improved, and molecular weight needs to be increased. Many attempts have been made to couple peptide immunogens to different carrier proteins [e.g. keyhole limpet haemocyanin (KLH) or ovalbumin]. This leads to very complex structures, however. We used a controlled conjugation of a peptide to a single long-chain fatty acid like palmitic acid by a thioester or an amide bond. It was found that these S-palmitoylated peptides were much more immunogenic than N-palmitoylated peptides and at least similar to KLH-conjugated peptides with respect to appearance and magnitude of induced antibodies (canine parvovirus) or immunocastration effect (gonadotropin-releasing hormone). For chemical synthesis of thioesters, we established conditions for solution and solid-phase synthesis. In both phases, Cys(SBut) could only be deprotected efficiently using phosphines, and S-acylation was accomplished using standard coupling at pH 5. We speculate that, in vivo, the presence of an appropriate fatty acid chain, chemically linked through a labile thioester bond, greatly enhances immunogenicity, because it represents a favourable substrate for cleavage by cellular thioesterases in cells of the immune system.


Assuntos
Antígenos/imunologia , Ácido Palmítico/metabolismo , Peptídeos/imunologia , Compostos de Sulfidrila/metabolismo , Vacinas Sintéticas/imunologia , Sequência de Aminoácidos , Animais , Ésteres , Cobaias , Imunização , Masculino , Dados de Sequência Molecular , Peptídeos/síntese química , Suínos
13.
Nat Biotechnol ; 15(3): 248-52, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9062924

RESUMO

The successful expression of animal or human virus epitopes on the surface of plant viruses has recently been demonstrated. These chimeric virus particles (CVPs) could represent a cost-effective and safe alternative to conventional animal cell-based vaccines. We report the insertion of oligonucleotides coding for a short linear epitope from the VP2 capsid protein of mink enteritis virus (MEV) into an infectious cDNA clone of cowpea mosaic virus and the successful expression of the epitope on the surface of CVPs when propagated in the black-eyed bean, Vigna unguiculata. The efficacy of the CVPs was established by the demonstration that one subcutaneous injection of 1 mg of the CVPs in mink conferred protection against clinical disease and virtually abolished shedding of virus after challenge with virulent MEV, demonstrating the potential utility of plant CVPs as the basis for vaccine development. The epitope used occurs in three different virus species-MEV, canine parvovirus, and feline panleukopenia virus- and thus the same vaccine could be used in three economically important viral hosts-mink, dogs, and cats, respectively.


Assuntos
Infecções por Parvoviridae/prevenção & controle , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar , Vírus da Panleucopenia Felina , Microscopia Eletrônica , Vison , Dados de Sequência Molecular , Vacinas Sintéticas/genética , Vacinas Virais/genética
14.
Vaccine ; 13(11): 1033-7, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8525686

RESUMO

Two recently developed vaccine--one based on synthetic peptide and one based on recombinant capsid protein--fully protected dogs against heavy experimental canine parvovirus (CPV) infection. The high sequence homology ( > 98%) and antigenic similarity between CPV and mink enteritis virus (MEV), feline panleukopenia virus, and raccoon parvovirus, suggest that both vaccines could protect mink, cats and raccoons against these respective host range variants. This was tested in mink and turned out to be the case. The two vaccines were fully protective and as effective as a conventional commercial vaccine based on inactivated virus. Surprisingly, this protection was obtained after only a single injection. Furthermore, the vaccinal dose of 150 micrograms of conjugated peptide or 3 micrograms of recombinant VP2 particles per animal, are sufficiently low to be cost-effective and applicable on a large scale.


Assuntos
Proteínas do Capsídeo , Vírus da Panleucopenia Felina/imunologia , Vison/virologia , Infecções por Parvoviridae/prevenção & controle , Infecções por Parvoviridae/veterinária , Parvovirus Canino/imunologia , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Capsídeo/genética , Capsídeo/imunologia , Dados de Sequência Molecular , Testes de Neutralização , Peptídeos/imunologia , Proteínas Recombinantes/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Virais/administração & dosagem
17.
Vaccine ; 12(15): 1473-80, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7887026

RESUMO

Fourteen synthetic peptides corresponding to previously mapped antigenic sites in VP2 of canine parvovirus (CPV) were used for immunization of rabbits to identify antiviral properties favourable for inclusion into a vaccine. Most antipeptide antisera obtained were reactive with viral protein, and with one of them it was possible to locate the hypothetical amino terminus of VP3 within positions 15-31 of VP2. Virus-neutralizing antibodies were only obtained with two overlapping 15-mer peptides corresponding in sequence to the amino terminus of VP2 (MSDGAVQPDGGQPAVRNERAT). Antibodies in the neutralizing sera bound most strongly to amino acids of the sequence DGGQPAV within the N-terminus of VP2, indicating that efforts to develop a synthetic vaccine against CVP should be focused on this stretch of amino acids. The two peptides induced long-lasting immunity (at least 8 months) using either Freund's adjuvant or aluminium hydroxide plus Quil A. Thus, this approach delineated the exact peptide sequence useful for vaccines applied to the amino-terminal region of VP2. These findings in experimental animals form a solid basis for exploration of a synthetic peptide vaccine against parvovirus infection in dogs, minks or cats.


Assuntos
Anticorpos Antivirais/biossíntese , Capsídeo/imunologia , Parvovirus Canino/imunologia , Peptídeos/imunologia , Adjuvantes Imunológicos/fisiologia , Sequência de Aminoácidos , Animais , Capsídeo/síntese química , Proteínas do Capsídeo , Feminino , Cobaias , Soros Imunes , Cinética , Dados de Sequência Molecular , Testes de Neutralização , Peptídeos/síntese química , Coelhos
18.
J Virol ; 68(7): 4506-13, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8207825

RESUMO

A synthetic peptide vaccine which protects dogs against challenge with virulent canine parvovirus is described. The amino acid sequence used was discovered in previous studies on the immunogenic properties of previously mapped antigenic sites and represents the amino-terminal region of viral protein VP2. As with marker vaccines, it is possible to discriminate between vaccinated dogs that have not been exposed to the virus and dogs that have been infected with the virus. The protective mechanism can be explained by a humoral response against the peptide aided by T-cell epitopes contained in the carrier protein used for peptide coupling. This is the first example of a synthetic peptide vaccine that induces protection in target animals.


Assuntos
Infecções por Parvoviridae/veterinária , Parvovirus Canino/imunologia , Peptídeos/imunologia , Vacinas Virais/uso terapêutico , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/biossíntese , Especificidade de Anticorpos , Divisão Celular/imunologia , Cães , Ensaio de Imunoadsorção Enzimática/veterinária , Masculino , Dados de Sequência Molecular , Testes de Neutralização , Infecções por Parvoviridae/prevenção & controle , Parvovirus Canino/patogenicidade , Peptídeos/síntese química , Coelhos , Linfócitos T/citologia , Linfócitos T/imunologia , Vacinação/veterinária , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , Vacinas Virais/imunologia
19.
J Gen Virol ; 74 ( Pt 9): 2005-10, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7690844

RESUMO

The distribution of epitopes defined by monoclonal antibodies (MAbs) on the surface of canine parvovirus (CPV) virions and recombinant VP2-capsids was established using immunoelectron microscopy. A correlation appeared to exist between the linear position, neutralizing activity and immunogold staining. Both viral capsids and recombinant capsids gave similar patterns of immunostaining. The neutralizing MAbs that recognized epitopes not previously identified by Pepscan or immunoblotting gave a clear staining. However, MAbs 3C9 and 3C10, identified by Pepscan and immunoblotting as recognizing linear epitopes, did not show any labelling (3C9) or only scattered labelling (3C10). MAb 3C9 recognizes an N-terminal domain of VP2. MAb 4AG6, which recognizes the same linear epitope as 3C10, did not bind to the capsids, indicating a different orientation. An immunofluorescence assay was performed to supplement the B cell epitope characterization. In contrast to other MAbs that gave nuclear and cytoplasmic staining, MAb 3C9 gave a preferential nuclear staining. Based on these results, it is hypothesized that the N terminus of VP2 is barely, or not at all, exposed on the surface of the native virions, but becomes accessible after some virion steric change (e.g. after attachment to the cell receptor).


Assuntos
Antígenos Virais/análise , Capsídeo/análise , Parvoviridae/ultraestrutura , Proteínas Recombinantes/análise , Vírion/ultraestrutura , Animais , Anticorpos Monoclonais , Capsídeo/imunologia , Proteínas do Capsídeo , Células Cultivadas , Cães , Epitopos/análise , Imunofluorescência , Immunoblotting , Microscopia Imunoeletrônica , Testes de Neutralização , Proteínas Recombinantes/imunologia , Frações Subcelulares/ultraestrutura
20.
J Virol ; 67(2): 765-72, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7678305

RESUMO

Ten antigenic sites on canine parvovirus (CPV) were mapped with a complete set of overlapping nonapeptides of the capsid proteins VP1 and VP2: five of these sites were recognized by sera from CPV-infected dogs, three were recognized by a rabbit anti-CPV antiserum, and two were recognized by murine monoclonal anti-CPV antibodies. A region covering the first 21 amino-terminal amino acid residues of VP2 was recognized by three sera from infected dogs, one neutralizing rabbit antiserum, and one neutralizing murine monoclonal antibody. Immunoabsorption experiments with full virions indicated that at least 6 of the 10 antigenic sites are located on the surface. Of these six, three sites occur in the amino terminus of VP2. When superimposed on the three-dimensional structure of canine parvovirus (J. Tsao, M. S. Chapman, M. Agbandje, W. Keller, K. Smith, H. Wu, M. Luo, T. J. Smith, M. G. Rossmann, R. W. Compans, and C. R. Parrish, Science 251:1456-1464, 1991), the other three epitopes are located on two loops of VP2 which form the highly exposed "spike" around the threefold-symmetry axis of the virus. Thus, these regions (amino terminus and loops 1 and 3) are of interest as major target sites for induction of neutralizing antibodies.


Assuntos
Antígenos Virais/imunologia , Linfócitos B/imunologia , Capsídeo/imunologia , Parvoviridae/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo , Células Cultivadas , Reações Cruzadas , Cães , Epitopos/imunologia , Cobaias , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Oligopeptídeos/imunologia , Coelhos , Relação Estrutura-Atividade , Propriedades de Superfície , Vírion/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...