Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Tipo de estudo
Intervalo de ano de publicação
1.
Viruses ; 15(11)2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-38005821

RESUMO

Classical swine fever (CSF) remains one of the most economically significant viral diseases affecting domestic pigs and wild boars worldwide. To develop a safe and effective vaccine against CSF, we have constructed a triple gene-deleted pseudorabies virus (PRVtmv)-vectored bivalent subunit vaccine against porcine circovirus type 2b (PCV2b) and CSFV (PRVtmv+). In this study, we determined the protective efficacy of the PRVtmv+ against virulent CSFV challenge in pigs. The results revealed that the sham-vaccinated control group pigs developed severe CSFV-specific clinical signs characterized by pyrexia and diarrhea, and became moribund on or before the seventh day post challenge (dpc). However, the PRVtmv+-vaccinated pigs survived until the day of euthanasia at 21 dpc. A few vaccinated pigs showed transient diarrhea but recovered within a day or two. One pig had a low-grade fever for a day but recovered. The sham-vaccinated control group pigs had a high level of viremia, severe lymphocytopenia, and thrombocytopenia. In contrast, the vaccinated pigs had a low-moderate degree of lymphocytopenia and thrombocytopenia on four dpc, but recovered by seven dpc. Based on the gross pathology, none of the vaccinated pigs had any CSFV-specific lesions. Therefore, our results demonstrated that the PRVtmv+ vaccinated pigs are protected against virulent CSFV challenge.


Assuntos
Circovirus , Vírus da Febre Suína Clássica , Peste Suína Clássica , Herpesvirus Suídeo 1 , Linfopenia , Trombocitopenia , Vacinas Virais , Suínos , Animais , Herpesvirus Suídeo 1/genética , Vacinas Virais/genética , Proteínas do Envelope Viral , Anticorpos Antivirais , Sus scrofa , Diarreia
2.
Viruses ; 15(11)2023 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-38005861

RESUMO

Rift Valley fever virus (RVFV) is considered to be a high biodefense priority based on its threat to livestock and its ability to cause human hemorrhagic fever. RVFV-infected livestock are also a significant risk factor for human infection by direct contact with contaminated blood, tissues, and aborted fetal materials. Therefore, livestock vaccination in the affected regions has the direct dual benefit and one-health approach of protecting the lives of millions of animals and eliminating the risk of severe and sometimes lethal human Rift Valley fever (RVF) disease. Recently, we have developed a bovine herpesvirus type 1 (BoHV-1) quadruple gene mutant virus (BoHV-1qmv) vector that lacks virulence and immunosuppressive properties due to the deletion of envelope proteins UL49.5, glycoprotein G (gG), gE cytoplasmic tail, and US9 coding sequences. In the current study, we engineered the BoHV-1qmv further by incorporating a chimeric gene sequence to express a proteolytically cleavable polyprotein: RVFV envelope proteins Gn ectodomain sequence fused with bovine granulocyte-macrophage colony-stimulating factor (GMCSF) and Gc, resulting in a live BoHV-1qmv-vectored subunit vaccine against RVFV for livestock. In vitro, the resulting recombinant virus, BoHV-1qmv Sub-RVFV, was replicated in cell culture with high titers. The chimeric Gn-GMCSF and Gc proteins expressed by the vaccine virus formed the Gn-Gc complex. In calves, the BoHV-1qmv Sub-RVFV vaccination was safe and induced moderate levels of the RVFV vaccine strain, MP12-specific neutralizing antibody titers. Additionally, the peripheral blood mononuclear cells from the vaccinated calves had six-fold increased levels of interferon-gamma transcription compared with that of the BoHV-1qmv (vector)-vaccinated calves when stimulated with heat-inactivated MP12 antigen in vitro. Based on these findings, we believe that a single dose of BoHV-1qmv Sub-RVFV vaccine generated a protective RVFV-MP12-specific humoral and cellular immune response. Therefore, the BoHV-1qmv sub-RVFV can potentially be a protective subunit vaccine for cattle against RVFV.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Vacinas Virais , Animais , Bovinos , Humanos , Vírus da Febre do Vale do Rift/genética , Anticorpos Neutralizantes , Anticorpos Antivirais , Leucócitos Mononucleares , Imunidade Celular , Vacinas Atenuadas/genética , Vacinas de Subunidades Antigênicas
3.
Viruses ; 12(9)2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32937797

RESUMO

Bovine herpesvirus 1 (BHV-1) causes respiratory infection and abortion in cattle. Following a primary infection, BHV-1 establishes lifelong latency in the trigeminal ganglia (TG). Periodic reactivation of the latent virus in TG neurons results in anterograde virus transport to nerve endings in the nasal mucosa and nasal virus shedding. The BHV-1 glycoprotein E cytoplasmic tail (gE-CT) is necessary for virus cell-to-cell spread in epithelial cells and neuronal anterograde transport. Recently, we identified two tyrosine residues, Y467 and Y563, within the tyrosine-based motifs 467YTSL470 and 563YTVV566, which, together, account for the gE CT-mediated efficient cell-to-cell spread of BHV-1 in epithelial cells. Here, we determined that in primary neuron cultures in vitro, the individual alanine exchange Y467A or Y563A mutants had significantly diminished anterograde axonal spread. Remarkably, the double-alanine-exchanged Y467A/Y563A mutant virus was not transported anterogradely. Following intranasal infection of rabbits, both wild-type (wt) and the Y467A/Y563A mutant viruses established latency in the TG. Upon dexamethasone-induced reactivation, both wt and the mutant viruses reactivated and replicated equally efficiently in the TG. However, upon reactivation, only the wt, not the mutant, was isolated from nasal swabs. Therefore, the gE-CT tyrosine residues Y467 and Y563 together are required for gE CT-mediated anterograde neuronal transport.


Assuntos
Transporte Biológico/fisiologia , Glicoproteínas/metabolismo , Herpesvirus Bovino 1/fisiologia , Neurônios/virologia , Tirosina/metabolismo , Animais , Bovinos , Doenças dos Bovinos/virologia , Linhagem Celular , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Dispositivos Lab-On-A-Chip , Coelhos , Gânglio Trigeminal/virologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Ativação Viral , Latência Viral , Eliminação de Partículas Virais
4.
J Virol ; 92(13)2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669828

RESUMO

Alphaherpesvirus envelope glycoprotein N (gN) and gM form a covalently linked complex. Bovine herpesvirus type 1 (BHV-1) UL49.5 (a gN homolog) contains two predicted cysteine residues, C42 and C78. The C42 is highly conserved among the alphaherpesvirus gN homologs (e.g., herpes simplex virus 1 and pseudorabies virus). To identify which cysteine residue is required for the formation of the UL49.5/gM complex and to characterize the functional significance of the UL49.5/gM complex, we constructed and analyzed C42S and C78S substitution mutants in either a BHV-1 wild type (wt) or BHV-1 UL49.5 cytoplasmic tail-null (CT-null) virus background. The results demonstrated that BHV-1 UL49.5 residue C42 but not C78 was essential for the formation of the covalently linked functional UL49.5/gM complex, gM maturation in the Golgi compartment, and efficient cell-to-cell spread of the virus. Interestingly, the C42S and CT-null mutations separately did not affect mutant UL49.5 virion incorporation. However, when both of the mutations were introduced simultaneously, the UL49.5 C42S/CT-null protein virion incorporation was severely reduced. Incidentally, the anti-VP22 antibody coimmunoprecipitated the UL49.5 C42S/CT-null mutant protein at a noticeably reduced level compared to that of the individual UL49.5 C42S and CT-null mutant proteins. As expected, in a dual UL49.5 C42S/VP22Δ virus with deletion of VP22 (VP22Δ), the UL49.5 C42S virion incorporation was also severely reduced while in a gMΔ virus, UL49.5 virion incorporation was affected only slightly. Together, these results suggested that UL49.5 virion incorporation is mediated redundantly, by both UL49.5/gM functional complex and VP22, through a putative gM-independent novel UL49.5 and VP22 interaction.IMPORTANCE Bovine herpesvirus 1 (BHV-1) envelope protein UL49.5 is an important virulence determinant because it downregulates major histocompatibility complex class I (MHC-I). UL49.5 also forms a covalently linked complex with gM. The results of this study demonstrate that UL49.5 regulates gM maturation and virus cell-to-cell spread since gM maturation in the Golgi compartment depends on covalently linked UL49.5/gM complex. The results also show that the UL49.5 residue cysteine 42 (C42) mediates the formation of the covalently linked UL49.5-gM interaction. Furthermore, a C42S mutant virus in which UL49.5 cannot interact with gM has defective cell-to-cell spread. Interestingly, UL49.5 also interacts with the tegument protein VP22 via its cytoplasmic tail (CT). The putative UL49.5 CT-VP22 interaction is essential for a gM-independent UL49.5 virion incorporation and is revealed when UL49.5 and gM are not linked. Therefore, UL49.5 virion incorporation is mediated by UL49.5-gM complex interaction and through a gM-independent interaction between UL49.5 and VP22.


Assuntos
Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/fisiologia , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Vírion/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Chlorocebus aethiops , Infecções por Herpesviridae/metabolismo , Homologia de Sequência , Células Vero , Replicação Viral
5.
Vet Immunol Immunopathol ; 147(3-4): 223-9, 2012 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-22578851

RESUMO

Bovine herpesvirus type 1 (BHV-1) envelope protein U(L)49.5 inhibits transporter associated with antigen processing (TAP) and down-regulates cell-surface expression of major histocompatibility complex (MHC) class I molecules to promote immune evasion. Earlier, we have constructed a BHV-1U(L)49.5Δ30-32 CT-null virus and determined that in the infected cells, TAP inhibition and MHC-I down regulation properties of the virus are abolished. In this study, we compared the pathogenicity and immune responses in calves infected with BHV-1U(L)49.5Δ30-32 CT-null and BHV-1 wt viruses. Following primary infection, both BHV-1 wt and BHV-1U(L)49.5Δ30-32 CT-null virus replicated in the nasal epithelium with very similar yields. BHV-1 antigen-specific CD8+ T cell proliferation as well as CD8+ T cell cytotoxicity in calves infected with the BHV-1U(L)49.5Δ30-32 CT-null virus peaked by 7 dpi (P<0.05) which is 7 days earlier than that of BHV-1 wt-infected calves. Further, virus neutralizing antibody (VN Ab) titers and IFN-γ producing peripheral blood mononuclear cells (PBMCs) in the U(L)49.5 mutant virus-infected calves, also peaked 7 days (IFN-γ; P<0.05) and 14 days (VN Ab; P<0.05) earlier, respectively. Therefore, relative to wt in the BHV-1U(L)49.5 mutant virus-infected calves, primary neutralizing antibody and cellular immune responses were induced significantly more rapidly.


Assuntos
Bovinos/imunologia , Bovinos/virologia , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD8-Positivos/imunologia , Doenças dos Bovinos/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 1/patogenicidade , Imunidade Celular , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Mutação , Estrutura Terciária de Proteína , Deleção de Sequência , Proteínas do Envelope Viral/química , Vacinas Virais/genética , Vacinas Virais/imunologia , Replicação Viral
6.
PLoS One ; 6(10): e25742, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22046246

RESUMO

Bovine herpesvirus type 1 (BHV-1) U(L)49.5 inhibits transporter associated with antigen processing (TAP) and down-regulates cell-surface expression of major histocompatibility complex (MHC) class I molecules to promote immune evasion. We have constructed a BHV-1 U(L)49.5 cytoplasmic tail (CT) null and several U(L)49.5 luminal domain mutants in the backbone of wild-type BHV-1 or BHV-1 U(L)49.5 CT- null viruses and determined their relative TAP mediated peptide transport inhibition and MHC-1 down-regulation properties compared with BHV-1 wt. Based on our results, the U(L)49.5 luminal domain residues 30-32 and U(L)49.5 CT residues, together, promote efficient TAP inhibition and MHC-I down-regulation functions. In vitro, BHV-1 U(L)49.5 Δ30-32 CT-null virus growth property was similar to that of BHV-1 wt and like the wt U(L)49.5, the mutant U(L)49.5 was incorporated in the virion envelope and it formed a complex with gM in the infected cells.


Assuntos
Herpesvirus Bovino 1/patogenicidade , Antígenos de Histocompatibilidade Classe I/genética , Evasão da Resposta Imune , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/fisiologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Regulação para Baixo/genética , Herpesvirus Bovino 1/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Alótipos Gm de Imunoglobulina/metabolismo , Proteínas Estruturais Virais/metabolismo
7.
Pesqui. vet. bras ; 30(1): 57-62, jan. 2010. tab, ilus
Artigo em Inglês | LILACS | ID: lil-540328

RESUMO

The immunogenicity of an inactivated, experimental vaccine based on a bovine herpesvirus type 5 strain defective in thymidine kinase and glycoprotein E (BoHV-5 gE/TKΔ) was evaluated in cattle and the results were compared with a vaccine containing the parental BoHV-5 strain (SV507/99). To formulate the vaccines, each virus (wildtype SV507/99 and BoHV-5 gE/TK∆) was multiplied in cell culture and inactivated with binary ethyleneimine (BEI). Each vaccine dose contained approximately of 10(7.5) TCID50 of inactivated virus mixed with an oil-based adjuvant (46:54). Forty calves, 6 to 9-months-old, were allocated into two groups of 20 animals each and vaccinated twice (days 0 and 22pv) by the subcutaneous route with either vaccine. Serum samples collected at day 0 and at different intervals after vaccination were tested for virus neutralizing (VN) antibodies against the parental virus and against heterologous BoHV-5 and BoHV-1 isolates. The VN assays demonstrated seroconversion to the respective homologous viruses in all vaccinated animals after the second vaccine dose (mean titers of 17.5 for the wildtype vaccine; 24.1 for the recombinant virus). All animals remained reagents up to day 116 pv, yet showing a gradual reduction in VN titers. Animals from both vaccine groups reacted in similar VN titers to different BoHV-1 and BoHV-5 isolates, yet the magnitude of serological response of both groups was higher against BoHV-5 field isolates. Calves vaccinated with the recombinant virus did not develop antibodies to gE as verified by negative results in a gE-specific ELISA, what would allow serological differentiation from naturally infected animals. Taken together, these results indicate that inactivated antigens of BoHV-5 gE/TK recombinant virus induced an adequate serological response against BoHV-5 and BoHV-1 and thus can be used as an alternative, differential vaccine candidate.


A imunogenicidade de vacina experimental inativada, produzida com uma cepa do herpesvírus bovino tipo 5 defectiva nos genes da timidina quinase e glicoproteína E (BoHV-5 gE/TKΔ) foi avaliada em bovinos e o resultado foi comparado com a resposta induzida pela cepa parental do BoHV-5 (SV507/99). Para a formulação da vacina, cultivos de células infectados com cada um dos vírus (SV507/99 ou BoHV-5 gE/TKΔ) foram inativados com etilenamina binária. Cada dose de vacina continha aproximadamente 107,5 TCID50 de um dos vírus inativados emulsionado em adjuvante oleoso (46:54). Quarenta bezerros de raças cruzadas, com idade entre seis a nove meses, foram alocados em dois grupos de 20 animais cada e vacinados duas vezes (dia 0 e 22 pv) pela via subcutânea com uma das vacinas. Amostras de soro foram coletadas no dia 0 e a vários intervalos após vacinação para a pesquisa de anticorpos neutralizantes frente ao vírus homólogo ou frente a isolados de BoHV-5 e BoHV-1. Os testes de soroneutralização (SN) demonstraram que todos os animais soroconverteram após a segunda dose da vacina (títulos médios de 17,5 para o grupo SV507/99; 24,1 para o grupo BoHV-5 gE/TKΔ). Todos os animais mantiveram níveis de anticorpos neutralizantes até o dia 116 pv, no entanto foi observada uma redução gradual no títulos. A sorologia cruzada com amostras heterólogas do BoHV-5 e BoHV-1 indicou que ambos os grupos vacinais reagiram em níveis similares frente ao mesmo vírus, no entanto a magnitude da resposta sorológica foi maior frente a amostras de BoHV-5. Os animais vacinados com a cepa recombinante não desenvolveram anticorpos contra a gE detectáveis por um ELISA específico, o que permitiria a sua diferenciação sorológica de animais infectados naturalmente. Esses resultados demonstram que a vacina contendo antígenos inativados do vírus recombinante BoHV-5 gE/TKΔ induziu resposta sorológica em níveis satisfatórios, constituindo-se, assim, em alternativa a cepa ...


Assuntos
Animais , Bovinos , Encefalite Viral , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/veterinária , Meningoencefalite , Vacinas Sintéticas , Vacinas Sintéticas/uso terapêutico , Bovinos , Vacinação/veterinária
8.
PLoS Pathog ; 4(5): e1000080, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18516302

RESUMO

Cytotoxic T-lymphocytes play an important role in the protection against viral infections, which they detect through the recognition of virus-derived peptides, presented in the context of MHC class I molecules at the surface of the infected cell. The transporter associated with antigen processing (TAP) plays an essential role in MHC class I-restricted antigen presentation, as TAP imports peptides into the ER, where peptide loading of MHC class I molecules takes place. In this study, the UL 49.5 proteins of the varicelloviruses bovine herpesvirus 1 (BHV-1), pseudorabies virus (PRV), and equine herpesvirus 1 and 4 (EHV-1 and EHV-4) are characterized as members of a novel class of viral immune evasion proteins. These UL 49.5 proteins interfere with MHC class I antigen presentation by blocking the supply of antigenic peptides through inhibition of TAP. BHV-1, PRV, and EHV-1 recombinant viruses lacking UL 49.5 no longer interfere with peptide transport. Combined with the observation that the individually expressed UL 49.5 proteins block TAP as well, these data indicate that UL 49.5 is the viral factor that is both necessary and sufficient to abolish TAP function during productive infection by these viruses. The mechanisms through which the UL 49.5 proteins of BHV-1, PRV, EHV-1, and EHV-4 block TAP exhibit surprising diversity. BHV-1 UL 49.5 targets TAP for proteasomal degradation, whereas EHV-1 and EHV-4 UL 49.5 interfere with the binding of ATP to TAP. In contrast, TAP stability and ATP recruitment are not affected by PRV UL 49.5, although it has the capacity to arrest the peptide transporter in a translocation-incompetent state, a property shared with the BHV-1 and EHV-1 UL 49.5. Taken together, these results classify the UL 49.5 gene products of BHV-1, PRV, EHV-1, and EHV-4 as members of a novel family of viral immune evasion proteins, inhibiting TAP through a variety of mechanisms.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , Herpesvirus Bovino 1/imunologia , Herpesvirus Equídeo 1/imunologia , Herpesvirus Suídeo 1/imunologia , Varicellovirus/fisiologia , Proteínas do Envelope Viral/imunologia , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Apresentação de Antígeno , Bovinos , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Cães , Herpesvirus Bovino 1/genética , Herpesvirus Equídeo 1/genética , Herpesvirus Suídeo 1/genética , Cavalos , Humanos , Transporte Proteico , Recombinação Genética , Suínos , Transdução Genética , Varicellovirus/patogenicidade , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA