Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Immunol ; 67(2 Pt B): 357-68, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26190308

RESUMO

Interleukin-26 (IL-26) is a member of the IL-10 family of cytokines. Though conserved across vertebrates, the IL-26 gene is functionally inactivated in a few mammals like rat, mouse and horse. We report here the identification, isolation and cloning of the cDNA of IL-26 from the dromedary camel. The camel cDNA contains a 516 bp open reading frame encoding a 171 amino acid precursor protein, including a 21 amino acid signal peptide. Sequence analysis revealed high similarity with other mammalian IL-26 homologs and the conservation of IL-10 cytokine family domain structure including key amino acid residues. We also report the identification and cloning of four novel transcript variants produced by alternative splicing at the Exon 3-Exon 4 regions of the gene. Three of the alternative splice variants had premature termination codons and are predicted to code for truncated proteins. The transcript variant 4 (Tv4) having an insertion of an extra 120 bp nucleotides in the ORF was predicted to encode a full length protein product with 40 extra amino acid residues. The mRNA transcripts of all the variants were identified in lymph node, where as fewer variants were observed in other tissues like blood, liver and kidney. The expression of Tv2 and Tv3 were found to be up regulated in mitogen induced camel peripheral blood mononuclear cells. IL-26-Tv2 expression was also induced in camel fibroblast cells infected with Camel pox virus in-vitro. The identification of the transcript variants of IL-26 from the dromedary camel is the first report of alternative splicing for IL-26 in a species in which the gene has not been inactivated.


Assuntos
Processamento Alternativo/genética , Camelus/genética , Interleucinas/genética , Processamento Alternativo/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Complementar/genética , Éxons/genética , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/virologia , Perfilação da Expressão Gênica , Interleucinas/metabolismo , Íntrons/genética , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Mitógenos/farmacologia , Dados de Sequência Molecular , Orthopoxvirus , Ovário/patologia , Filogenia , Infecções por Poxviridae/genética , Infecções por Poxviridae/patologia , Infecções por Poxviridae/virologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Alinhamento de Sequência , Homologia Estrutural de Proteína
2.
Eur J Immunol ; 42(10): 2655-66, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22706912

RESUMO

Cross-presentation is the pathway by which exogenous antigens are routed for presentation by MHC class I molecules leading to activation of antiviral CD8(+) T-cell responses. However, there is little information describing the modulation of cross-presentation and the impact of pathogen-derived signals associated with Japanese encephalitis virus (JEV), which is one of the most common causes of encephalitis in humans. In this study, we demonstrate that JEV infection could suppress in vivo cross-presentation of soluble and cell-associated antigens, thereby generating weak CD8(+) T-cell responses to exogenous antigens, as evaluated by CFSE dilution of adoptively transferred CD8(+) T cells and in vivo CTL killing activity. Furthermore, CD8α(+) CD11c(+) dendritic cells (DCs), which are known to be far more efficient at cross-presenting soluble antigens, played a specific role in contributing to JEV-mediated inhibition of the cross-presentation of exogenous antigens through interference with effective antigen uptake. Finally, this study provides evidence that TLR2-MyD88 and p38 MAPK signal pathway might be involved in JEV-mediated inhibition of cross-presentation of soluble and cell-associated antigens. These observations suggest that the modulation of cross-presentation of exogenous antigens through TLR signaling has important implications for antiviral immune responses against JEV infection and the development of effective vaccination strategies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada , Células Dendríticas/imunologia , Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Antígenos Virais/metabolismo , Antígeno CD11c/metabolismo , Antígenos CD8/metabolismo , Proliferação de Células , Citotoxicidade Imunológica , Células Dendríticas/virologia , Ativação Linfocitária , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
J Immunol ; 185(3): 1429-41, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20581148

RESUMO

Japanese encephalitis virus (JEV) is a frequent cause of acute and epidemic viral encephalitis. However, there is little information describing the mechanisms by which JEV subverts immune responses that may predispose the host to secondary infections. In this study, we found that JEV induced the subversion of CD8(+) T cell responses in a transient manner that was closely correlated with viral multiplication. Subsequently, analysis using a TCR-transgenic system revealed that CD8(+) T cells purified from JEV-infected mice showed impaired responses, and that naive CD8(+) T cells adoptively transferred into JEV-infected recipients showed less expanded responses. Furthermore, JEV altered the splenic dendritic cell (DC) subpopulation via preferential depletion of CD8alpha(+)CD11c(+) DCs without changing the plasmacytoid DCs and induced a significant reduction in the surface expression of MHC class II and CD40, but not MHC class I, CD80, and CD86 molecules. Finally, JEV was shown to inhibit the presentation of MHC class I-restricted Ag in DCs, and this immune suppression was ameliorated via the activation of DCs by TLR agonists. Collectively, our data indicate that JEV precludes the functions of Ag-presenting machinery, such as depletion of CD8alpha(+)CD11c(+) DCs and downregulation of MHC class I-restricted Ag presentation, thereby leading to immune subversion of CD8(+) T cells.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Vírus da Encefalite Japonesa (Espécie)/imunologia , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Evasão da Resposta Imune/imunologia , Animais , Antígeno CD11c/biossíntese , Antígeno CD11c/metabolismo , Antígenos CD8/biossíntese , Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/patologia , Chlorocebus aethiops , Testes Imunológicos de Citotoxicidade , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Encefalite Japonesa/imunologia , Encefalite Japonesa/patologia , Encefalite Japonesa/virologia , Epitopos de Linfócito T/imunologia , Herpesvirus Humano 1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Vero
4.
J Immunol ; 183(4): 2462-74, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19635909

RESUMO

Dendritic cells (DCs) are potent initiators of T cell-mediated immunity that undergo maturation during viral infections. However, few reports describing the interactions of DCs with Japanese encephalitis virus (JEV), which remains the most frequent cause of acute and epidemic viral encephalitis, are available. In this study, we investigated the interaction of JEV with DCs and macrophages. JEV replicated its viral RNA in both cells with different efficiency, and JEV infection of macrophages followed the classical activation pathway of up-regulation of tested costimulatory molecules and proinflammatory cytokine production (IL-6, TNF-alpha, and IL-12). On the contrary, JEV-infected DCs failed to up-regulate costimulatory molecules such as CD40 and MHC class II. Of more interest, along with production of proinflammatory cytokines, DCs infected by JEV released antiinflammatory cytokine IL-10, which was not detected in macrophages. Moreover, signaling through MyD88 molecule, a pan-adaptor molecule of TLRs, and p38 MAPK in JEV-infected DCs was found to play a role in the production of cytokines and subversion of primary CD4(+) and CD8(+) T cell responses. We also found that IL-10 released from JEV-infected DCs led to a reduction in the priming of CD8(+) T cells, but not CD4(+) T cells. Taken together, our data suggest that JEV induces functional impairment of DCs through MyD88-dependent and -independent pathways, which subsequently leads to poor CD4(+) and CD8(+) T cell responses, resulting in boosting viral survival and dissemination in the body.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Vírus da Encefalite Japonesa (Espécie)/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Fator 88 de Diferenciação Mieloide/fisiologia , Transdução de Sinais/imunologia , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Células Cultivadas , Células Dendríticas/metabolismo , Encefalite Japonesa/imunologia , Encefalite Japonesa/patologia , Encefalite Japonesa/virologia , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Replicação Viral/imunologia
5.
Immunol Cell Biol ; 87(1): 91-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18794906

RESUMO

The CC chemokine receptor 7 (CCR7) and cognate CCR7 ligands, CCL19 and CCL21, help establish microenvironments in lymphoid tissue that can facilitate encounters between naive T cells and mature dendritic cells (DCs). This study was conducted to determine if CCR7 ligands can augment the immunogenicity of a DNA vaccine that expresses glycoprotein B (gB) of the pseudorabies virus (PrV). The genetic co-transfer of CCR7 ligands along with a PrV DNA vaccine increased the levels of serum PrV-specific immunoglobulin (Ig) G by 2- to 2.5-fold. In addition, the level of PrV-specific IgG2a isotype was significantly enhanced by co-injection of CCR7 ligand DNA, which indicates that CCR7 ligand biases the humoral immunity toward the Th1-type pattern. The co-injection of CCR7 ligand DNA consistently enhanced the level of Th1-type cytokines (IL-2 and IFN-gamma) produced by stimulated immune cells when compared with a group that was vaccinated with the PrV DNA vaccine. Also, the genetic co-transfer of CCR7 ligand DNAs with PrV DNA vaccine provided prolonged survival against a virulent challenge by PrV. Moreover, the co-administration of CCR7 ligand DNA increased the number of mature DCs into the secondary lymphoid tissues, which appeared to enhance the proliferation of PrV-immune CD4(+) T cells. Taken together, these findings indicate that CCR7 ligands are an attractive adjuvant for a PrV DNA vaccine that can offer protective immunity against the PrV.


Assuntos
Quimiocina CCL19/imunologia , Quimiocina CCL21/imunologia , Vacinas contra Pseudorraiva/imunologia , Pseudorraiva/prevenção & controle , Receptores CCR7/imunologia , Vacinas de DNA/imunologia , Animais , Formação de Anticorpos , Quimiocina CCL19/genética , Quimiocina CCL21/genética , Citocinas/biossíntese , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Pseudorraiva/imunologia , Vacinas contra Pseudorraiva/administração & dosagem , Vacinas contra Pseudorraiva/genética , Transfecção , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas do Envelope Viral/imunologia
6.
J Microbiol Biotechnol ; 18(7): 1326-34, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18667863

RESUMO

The prime-boost vaccination with DNA vaccine and recombinant viral vector has emerged as an effective prophylactic strategy to control infectious diseases. Here, we compared the protective immunities induced by multiple alternating immunizations with DNA vaccine (pCIgB) and replication-incompetent adenovirus (Ad-gB) expressing glycoprotein gB of pseudorabies virus (PrV). The platform of pCIgB-prime and Ad-gB-boost induced the most effective immune responses and provided protection against virulent PrV infection. However, priming with pCIgB prior to vaccinating animals by the DNA vaccine-prime and Ad-boost protocol provided neither effective immune responses nor protection against PrV. Similarly, boosting with Ad-gB following immunization with DNA vaccine-prime and Ad-boost showed no significant responses. Moreover, whereas the administration of Ad-gB for primary immunization induced Th2-type-biased immunity, priming with pCIgB induced Th1-type-biased immunity, as judged by the production of PrV-specific IgG isotypes and cytokine IFN-gamma. These results indicate that the order and injection frequency of vaccine vehicles used for heterologous prime-boost vaccination affect the magnitude and nature of the immunity. Therefore, our demonstration implies that the prime-boost protocol should be carefully considered and selected to induce the desired immune responses.


Assuntos
Adenoviridae/metabolismo , Expressão Gênica , Herpesvirus Suídeo 1/imunologia , Vacinas contra Pseudorraiva/administração & dosagem , Pseudorraiva/prevenção & controle , Vacinação , Vacinas de DNA/administração & dosagem , Proteínas do Envelope Viral/imunologia , Adenoviridae/genética , Animais , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Citocinas/imunologia , Feminino , Herpesvirus Suídeo 1/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Pseudorraiva/imunologia , Pseudorraiva/virologia , Vacinas contra Pseudorraiva/genética , Vacinas contra Pseudorraiva/imunologia , Vacinação/métodos , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/genética , Replicação Viral
7.
Exp Mol Med ; 40(6): 583-95, 2008 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-19116444

RESUMO

Replication-incompetent adenoviruses expressing three major glycoproteins (gB, gC, and gD) of pseudorabies virus (PrV) were constructed and used to examine the ability of these glycoproteins to induce protective immunity against a lethal challenge. Among three constructs, recombinant adenovirus expressing gB (rAd-gB) was found to induce the most potent immunity biased to Th1-type, as determined by the IgG isotype ratio and the profile of the Th1/Th2 cytokine production. Conversely, the gC-expressing adenovirus (rAd-gC) revealed Th2-type immunity and the gD-expressing adenovirus (rAd-gD) induced lower levels of IFN-? and IL-4 production than other constructs, except IL-2 production. Mucosal delivery of rAd-gB induced mucosal IgA and serum IgG responses and biased toward Th2-type immune responses. However, these effects were not observed in response to systemic delivery of rAd-gB. In addition, rAd-gB appeared to induce effective protective immunity against a virulent viral infection, regardless of whether it was administered via the muscular or systemic route. These results suggest that administration of replication-incompetent adenoviruses can induce different types of immunity depending on the expressed antigen and that recombinant adenoviruses expressing gB induced the most potent Th1-biased humoral and cellular immunity and provided effective protection against PrV infection.


Assuntos
Adenoviridae/imunologia , Glicoproteínas/imunologia , Herpesvirus Suídeo 1/imunologia , Vacinas contra Pseudorraiva/imunologia , Pseudorraiva/imunologia , Replicação Viral , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Formação de Anticorpos , Linhagem Celular , Citocinas/imunologia , Feminino , Glicoproteínas/biossíntese , Glicoproteínas/genética , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/fisiologia , Imunidade Celular , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pseudorraiva/prevenção & controle , Vacinas contra Pseudorraiva/administração & dosagem , Suínos , Células Th1/imunologia , Células Th2/imunologia
8.
Res Vet Sci ; 83(1): 73-81, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17196231

RESUMO

To assess the correlation between the nature of immunity induced by different types of immunogens and the establishment of latent infection by wild-type pseudorabies virus (PrV), we used a murine model immunized with different immunogens, the PrV modified live vaccine (MLV), inactivated vaccine (IAV), and commercial oil-adjuvant subunit vaccine (OSV), via either intranasal (i.n.) or intramuscular (i.m.) route. Both MLV and IAV induced a different nature of immunity biased to Th1- and Th2-type, respectively, as judged by the ratio of PrV-specific IgG isotypes (IgG2a/IgG1) and the profile of cytokine IL-2, IL-4, and IFN-gamma production. In contrast, the OSV induced a lower isotype IgG2a to IgG1 ratio and higher level of IL-2 production. The MLV (inducing Th1-type) provided more effective protection against a virulent wild-type PrV challenge than IAV and OSV (inducing Th2- and mixed type, respectively). In addition, the MLV impeded the establishment of a latent infection with wild-type PrV, and the decrease in the PrV latency load by immunization with the MLV appeared to be mediated by the immune T-cells. These results demonstrate the substantial role of the immune responses driven by preceding vaccination in modulating the establishment of PrV latency caused by the post-infection of a field virus.


Assuntos
Herpesvirus Suídeo 1/imunologia , Vacinas contra Pseudorraiva/imunologia , Pseudorraiva/prevenção & controle , Pseudorraiva/virologia , Latência Viral/fisiologia , Animais , Anticorpos Antivirais/sangue , Vias de Administração de Medicamentos , Feminino , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos C57BL , Pseudorraiva/imunologia , Vacinas contra Pseudorraiva/administração & dosagem
9.
J Microbiol Biotechnol ; 17(12): 1955-64, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18167442

RESUMO

A recent report showed that analysis of CD154 expression in the presence of the secretion inhibitor Brefeldin A (Bref A) could be used to assess the entire repertoire of antigen-specific CD4(+) T helper cells. However, the capacity of intracellular CD154 expression to identify antigen-specific CD8(+) T cells has yet to be investigated. In this study, we compared the ability of intracellular CD154 expression to assess antigen-specific CD8(+) T cells with that of accepted standard assays, namely intracellular cytokine IFN-gamma staining (ICS) and MHC class I tetramer staining. The detection of intracellular CD154 molecules in the presence of Bref A reflected the kinetic trend of antigen-specific CD8+ T cell number, but unfortunately showed less sensitivity than ICS and tetramer staining. However, ICS levels peaked and saturated 8 h after antigenic stimulation in the presence of Bref A and then declined, whereas intracellular CD154 expression peaked by 8 h and maintained the saturated level up to 24 h post-stimulation. Moreover, intracellular CD154 expression in antigen-specific CD8+ T cells developed in the absence of CD4(+) T cells changed little, whereas the number of IFN-gamma-producing CD8(+) T cells decreased abruptly. These results suggest that intracellular CD154 could aid the assessment of antigen-specific CD8(+) T cells, but does not have as much ability to identify heterogeneous CD4(+) T helper cells. Therefore, the combined analytical techniques of ICS and tetramer staining together with intracellular CD154 assays may be able to provide useful information on the accurate phenotype and functionality of antigen-specific CD8(+) T cells.


Assuntos
Ligante de CD40/biossíntese , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Interferon gama/biossíntese , Transferência Adotiva , Animais , Antivirais/farmacologia , Brefeldina A/farmacologia , Linfócitos T CD4-Positivos/imunologia , Ligante de CD40/imunologia , Proteínas do Ovo/imunologia , Proteínas do Ovo/farmacologia , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/imunologia , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia , Ovalbumina/farmacologia , Fragmentos de Peptídeos , Organismos Livres de Patógenos Específicos
10.
J Interferon Cytokine Res ; 26(10): 730-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17032167

RESUMO

The immunomodulatory efficacy of interferon-gamma (IFN-gamma)-associated cytokines coadministered with a plasmid DNA vaccine has been investigated, with variable results. Therefore, to test the immunomodulatory effect of IFN-gamma-associated cytokines as vaccine adjuvant, the present study evaluated the immune responses induced by pseudorabies virus (PrV) gB-encoded plasmid DNA vaccine coadministered with IFN-gamma-associated cytokines and chemokines. These cytokines and chemokines included interleukin-12 (IL-12) and IL-18, as potent inducers of IFN-gamma, and IFN-gamma-inducible protein (IP-10), the production of which is IFN-gamma dependent. A coinjection of either IL-12 or IL-18 strongly suppressed the humoral antibody responses but increased the production of the Th1-type cytokines IFN-gamma and IL-2 from immune T cells. Such antibody suppression was closely related to the increased susceptibility against a virulent viral challenge. On the other hand, IP-10 exhibited enhanced immune responses in both antibody responses and IFN-gamma production of immune T cells and facilitated the prolonged survival of infected mice. In contrast, there was no significant change in the immune responses of the mice that received codelivery of IFN-gamma. Therefore, IFN-gamma-associated cytokines, as Th1-type inducers, can generate unexpected and unwanted effects, and their application as a vaccine adjuvant should be carefully evaluated depending on the target antigens.


Assuntos
Citocinas/genética , Herpesvirus Suídeo 1/imunologia , Vacinas contra Herpesvirus/imunologia , Proteínas do Envelope Viral/genética , Animais , Anticorpos Antivirais/sangue , Quimiocina CXCL10 , Quimiocinas CXC/genética , Citocinas/biossíntese , Feminino , Expressão Gênica , Vacinas contra Herpesvirus/genética , Interferon gama/biossíntese , Interferon gama/genética , Interleucina-12/genética , Interleucina-18/genética , Camundongos , Camundongos Endogâmicos C57BL , Pseudorraiva/imunologia , Linfócitos T/imunologia , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/metabolismo
11.
Immunol Cell Biol ; 84(6): 502-11, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16869937

RESUMO

A murine model immunized with plasmid DNA vaccine expressing three glycoproteins pCIgB, pCIgC and pCIgD were used to examine the relative potency of major glycoproteins as well as the contribution of immunological parameters in providing protective immunity against the pseudorabies virus (PrV). Among the three glycoprotein-encoded plasmid DNA vaccines, pCIgB produced the strongest response of PrV-specific IgG in the sera. pCIgB and pCIgD also induced a contrast pattern of immunity that was biased to the Th1 and Th2 types, respectively. pCIgC showed the potent inducer of CD8+ T-cell-mediated CTL activity against PrV. In addition, a cocktail vaccination of all three glycoprotein-encoded plasmid DNA vaccines induced the production of both cytokine types, Th1 and Th2 with levels that were the same as that of each immunogen. With regard to protective efficacy, pCIgB induced the most effective protection against a virulent virus challenge and a cocktail vaccination appeared to offer complete protection against a 5 LD50 challenge, but not a 10 LD50 one. pCIgD induced protection that was same as pCIgB, but pCIgC offered no effective protection. These results show the relative potency of the three glycoprotein-encoded PrV DNA vaccines in inducing protective immunity against PrV infection. The results in this study support previous results showing the importance of Th1-type CD4+ T cells and their antibodies in conferring protection.


Assuntos
Formação de Anticorpos , Glicoproteínas/imunologia , Herpesvirus Suídeo 1/imunologia , Vacinas contra Pseudorraiva/imunologia , Vacinas de DNA/imunologia , Animais , Feminino , Vetores Genéticos , Glicoproteínas/genética , Glicoproteínas/metabolismo , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
12.
Microbiol Immunol ; 50(2): 83-92, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16490926

RESUMO

Granulocyte/macrophage colony-stimulatory factor (GM-CSF) is an attractive adjuvant for a DNA vaccine on account of its ability to recruit antigen-presenting cells (APCs) to the site of antigen synthesis as well as its ability to stimulate the maturation of dendritic cells (DCs). This study evaluated the utility of GM-CSF cDNA as a DNA vaccine adjuvant for glycoprotein B (gB) of pseudorabies virus (PrV) in a murine model. The co-injection of GM-CSF DNA enhanced the levels of serum PrV-specific IgG with a 1.5-to 2-fold increase. Moreover, GM-CSF co-injection inhibited the production of IgG2a isotype. However, it enhanced production of an IgG1 isotype resulting in humoral responses biased to the Th2-type against PrV antigen. In contrast, the co-administration of GM-CSF DNA enhanced the T cell-mediated immunity biased to the Th1-type, as judged by the significantly higher level of cytokine IL-2 and IFN-gamma production but not IL-4. When challenged with a lethal dose of PrV, the GM-CSF co-injection enhanced the resistance against a PrV infection. This suggests that co-inoculation with a vector expressing GM-CSF enhanced the protective immunity against a PrV infection. This immunity was caused by the induction of increased humoral and cellular immunity in response to PrV antigen.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Herpesvirus Suídeo 1/imunologia , Vacinas contra Pseudorraiva/imunologia , Pseudorraiva/imunologia , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/genética , Animais , Anticorpos Antivirais/sangue , Encéfalo/virologia , Ensaio de Imunoadsorção Enzimática , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Isotipos de Imunoglobulinas , Interferon gama/imunologia , Interleucina-2/imunologia , Interleucina-4/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , Plasmídeos/imunologia , Pseudorraiva/prevenção & controle , Vacinas contra Pseudorraiva/genética , Vacinas contra Pseudorraiva/uso terapêutico , Vacinas de DNA/genética , Vacinas de DNA/uso terapêutico , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...