Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
PLoS Pathog ; 16(2): e1008279, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32023327

RESUMO

IFN-γ is an enigmatic cytokine that shows direct anti-viral effects, confers upregulation of MHC-II and other components relevant for antigen presentation, and that adjusts the composition and balance of complex cytokine responses. It is produced during immune responses by innate as well as adaptive immune cells and can critically affect the course and outcome of infectious diseases, autoimmunity, and cancer. To selectively analyze the function of innate immune cell-derived IFN-γ, we generated conditional IFN-γOFF mice, in which endogenous IFN-γ expression is disrupted by a loxP flanked gene trap cassette inserted into the first intron of the IFN-γ gene. IFN-γOFF mice were intercrossed with Ncr1-Cre or CD4-Cre mice that express Cre mainly in NK cells (IFN-γNcr1-ON mice) or T cells (IFN-γCD4-ON mice), respectively. Rosa26RFP reporter mice intercrossed with Ncr1-Cre mice showed selective RFP expression in more than 80% of the NK cells, while upon intercrossing with CD4-Cre mice abundant RFP expression was detected in T cells, but also to a minor extent in other immune cell subsets. Previous studies showed that IFN-γ expression is needed to promote survival of vaccinia virus (VACV) infection. Interestingly, during VACV infection of wild type and IFN-γCD4-ON mice two waves of serum IFN-γ were induced that peaked on day 1 and day 3/4 after infection. Similarly, VACV infected IFN-γNcr1-ON mice mounted two waves of IFN-γ responses, of which the first one was moderately and the second one profoundly reduced when compared with WT mice. Furthermore, IFN-γNcr1-ON as well as IFN-γCD4-ON mice survived VACV infection, whereas IFN-γOFF mice did not. As expected, ex vivo analysis of splenocytes derived from VACV infected IFN-γNcr1-ON mice showed IFN-γ expression in NK cells, but not T cells, whereas IFN-γOFF mice showed IFN-γ expression neither in NK cells nor T cells. VACV infected IFN-γNcr1-ON mice mounted normal cytokine responses, restored neutrophil accumulation, and showed normal myeloid cell distribution in blood and spleen. Additionally, in these mice normal MHC-II expression was detected on peripheral macrophages, whereas IFN-γOFF mice did not show MHC-II expression on such cells. In conclusion, upon VACV infection Ncr1 positive cells including NK cells mount two waves of early IFN-γ responses that are sufficient to promote the induction of protective anti-viral immunity.


Assuntos
Antígenos Ly/imunologia , Regulação da Expressão Gênica/imunologia , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Vírus Vaccinia/imunologia , Vaccinia/imunologia , Animais , Antígenos Ly/genética , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Interferon gama/genética , Células Matadoras Naturais/patologia , Camundongos , Camundongos Transgênicos , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Linfócitos T/imunologia , Linfócitos T/patologia , Vaccinia/genética , Vaccinia/patologia , Vírus Vaccinia/genética
2.
J Leukoc Biol ; 107(6): 941-952, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31985117

RESUMO

Receptor interacting protein kinase 1 (RIP1) is a critical effector of inflammatory responses and cell death activation. Cell death pathways regulated by RIP1 include caspase-dependent apoptosis and caspase-independent necroptosis. The kinase activity of RIP1 has been associated with a number of inflammatory, neurodegenerative, and oncogenic diseases. In this study, we use the RIP1 kinase inhibitor GNE684 to demonstrate that RIP1 inhibition can effectively block skin inflammation and immune cell infiltrates in livers of Sharpin mutant (Cpdm; chronic proliferative dermatitis) mice in an interventional setting, after disease onset. On the other hand, genetic inactivation of RIP1 (RIP1 KD) or ablation of RIP3 (RIP3 KO) or MLKL (MLKL KO) did not affect testicular pathology of aging male mice. Likewise, infection with vaccinia virus or with mouse gammaherpesvirus MHV68 resulted in similar viral clearance in wild-type, RIP1 KD, and RIP3 KO mice. In summary, this study highlights the benefits of inhibiting RIP1 in skin inflammation, as opposed to its lack of relevance for testicular longevity and the response to certain viral infections.


Assuntos
Dermatite/genética , Infecções por Herpesviridae/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Pele/imunologia , Vaccinia/genética , Animais , Doença Crônica , Dermatite/imunologia , Dermatite/patologia , Dermatite/virologia , Modelos Animais de Doenças , Gammaherpesvirinae/imunologia , Gammaherpesvirinae/patogenicidade , Regulação da Expressão Gênica , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Inflamação , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/deficiência , Proteínas Quinases/genética , Proteínas Quinases/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Transdução de Sinais , Pele/patologia , Pele/virologia , Testículo/imunologia , Testículo/patologia , Testículo/virologia , Vaccinia/imunologia , Vaccinia/patologia , Vaccinia/virologia , Vírus Vaccinia/imunologia , Vírus Vaccinia/patogenicidade , Replicação Viral/imunologia
3.
PLoS One ; 14(7): e0219449, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31283790

RESUMO

Signaling through the inducible costimulator ICOS is required for the homeostasis and function of various immune cell populations, with an outstanding role in the generation and maintenance of germinal centers. Very recently, it has been suggested that the clinical phenotype of ICOS-deficient patients is much broader than initially anticipated and the innate immune response might be also affected. However, the role of the ICOS/ICOS-Ligand axis in the homeostasis and development of innate NK cells is not known, and reports on its participation in NK cell activation are scarce. NK cells may express low levels of ICOS that are markedly enhanced upon activation. We show here that ICOS-deficient (ICOS-KO) mice present low NK cell numbers and defects in the homeostasis of these cells, with delayed maturation and altered expression of the developmental NK cell markers CD122, NK1.1, CD11b or CD27. Our experiments in mixed bone marrow chimera mice indicate that, both, cell-intrinsic defects of ICOS-KO NK and deficiencies in the milieu of these mice contribute to the altered phenotype. ICOS-deficient NK cells show impaired production of IFN-γ and cytotoxicity, and a final outcome of defects in NK cell-mediated effector function during the response to poly(I:C) or vaccinia virus infection in vivo. Interestingly, we show that murine innate cells like IL-2-cultured NK and bone marrow-derived dendritic cells can simultaneously express ICOS and ICOS-Ligand; both molecules are functional in NK intracellular signaling, enhancing early phosphorylation of Akt and Erk, or IFN-γ secretion in IL-2-activated NK cells. Our study shows the functional importance of the ICOS/ICOS-L pair in NK cell homeostasis, differentiation and activity and suggests novel therapeutic targets for NK manipulation.


Assuntos
Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Células Matadoras Naturais/metabolismo , Animais , Apoptose , Antígeno CD11b/metabolismo , Diferenciação Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Ligante Coestimulador de Linfócitos T Induzíveis/genética , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/deficiência , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interferon gama/metabolismo , Interleucina-2/farmacologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Poli I-C/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Vaccinia/imunologia , Vaccinia/patologia
4.
Nat Microbiol ; 4(2): 216-225, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30420785

RESUMO

Cell motility is essential for viral dissemination1. Vaccinia virus (VACV), a close relative of smallpox virus, is thought to exploit cell motility as a means to enhance the spread of infection1. A single viral protein, F11L, contributes to this by blocking RhoA signalling to facilitate cell retraction2. However, F11L alone is not sufficient for VACV-induced cell motility, indicating that additional viral factors must be involved. Here, we show that the VACV epidermal growth factor homologue, VGF, promotes infected cell motility and the spread of viral infection. We found that VGF secreted from early infected cells is cleaved by ADAM10, after which it acts largely in a paracrine manner to direct cell motility at the leading edge of infection. Real-time tracking of cells infected in the presence of EGFR, MAPK, FAK and ADAM10 inhibitors or with VGF-deleted and F11-deleted viruses revealed defects in radial velocity and directional migration efficiency, leading to impaired cell-to-cell spread of infection. Furthermore, intravital imaging showed that virus spread and lesion formation are attenuated in the absence of VGF. Our results demonstrate how poxviruses hijack epidermal growth factor receptor-induced cell motility to promote rapid and efficient spread of infection in vitro and in vivo.


Assuntos
Movimento Celular , Interações Hospedeiro-Patógeno , Peptídeos/metabolismo , Transdução de Sinais , Vírus Vaccinia/fisiologia , Vaccinia/virologia , Proteína ADAM10/antagonistas & inibidores , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Efeito Citopatogênico Viral/genética , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Deleção de Genes , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Peptídeos/deficiência , Peptídeos/genética , Transdução de Sinais/efeitos dos fármacos , Vaccinia/metabolismo , Vaccinia/patologia , Vírus Vaccinia/genética , Vírus Vaccinia/crescimento & desenvolvimento , Vírus Vaccinia/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
Cell Death Dis ; 9(1): 9, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311549

RESUMO

Differentiation of naive CD4+ T-cells into functionally distinct T helper (Th) subsets is critical to immunity against pathogen infection. Little is known about the role of signals emanating from the nuclear envelope for T-cell differentiation. The nuclear envelope protein lamin A/C is induced in naive CD4+ T-cells upon antigen recognition and acts as a link between the nucleus and the plasma membrane during T-cell activation. Here we demonstrate that the absence of lamin A/C in naive T-cell reduces Th1 differentiation without affecting Th2 differentiation in vitro and in vivo. Moreover, Rag1 -/- mice reconstituted with Lmna -/- CD4+CD25 - T-cells and infected with vaccinia virus show weaker Th1 responses and viral removal than mice reconstituted with wild-type T-cells. Th1 responses and pathogen clearance upon Leishmania major infection were similarly diminished in mice lacking lamin A/C in the complete immune system or selectively in T-cells. Lamin A/C mediates Th1 polarization by a mechanism involving T-bet and IFNγ production. Our results reveal a novel role for lamin A/C as key regulator of Th1 differentiation in response to viral and intracellular parasite infections.


Assuntos
Lamina Tipo A/genética , Leishmaniose Cutânea/patologia , Células Th1/metabolismo , Vaccinia/patologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular , Suscetibilidade a Doenças , Sistema Imunitário/metabolismo , Interferon gama/metabolismo , Interleucina-4/metabolismo , Lamina Tipo A/deficiência , Leishmania major/patogenicidade , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/veterinária , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Proteínas com Domínio T/metabolismo , Células Th1/citologia , Células Th1/imunologia , Vaccinia/imunologia , Vaccinia/veterinária , Vírus Vaccinia/patogenicidade
6.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490586

RESUMO

All viruses strategically alter the antiviral immune response to their benefit. The vaccinia virus (VACV) K1 protein has multiple immunomodulatory effects in tissue culture models of infection, including NF-κB antagonism. However, the effect of K1 during animal infection is poorly understood. We determined that a K1L-less vaccinia virus (vΔK1L) was less pathogenic than wild-type VACV in intranasal and intradermal models of infection. Decreased pathogenicity was correlated with diminished virus replication in intranasally infected mice. However, in intradermally inoculated ears, vΔK1L replicated to levels nearly identical to those of VACV, implying that the decreased immune response to vΔK1L infection, not virus replication, dictated lesion size. Several lines of evidence support this theory. First, vΔK1L induced slightly less edema than vK1L, as revealed by histopathology and noninvasive quantitative ultrasound technology (QUS). Second, infiltrating immune cell populations were decreased in vΔK1L-infected ears. Third, cytokine and chemokine gene expression was decreased in vΔK1L-infected ears. While these results identified the biological basis for smaller lesions, they remained puzzling; because K1 antagonizes NF-κB in vitro, antiviral gene expression was expected to be higher during vΔK1L infection. Despite these diminished innate immune responses, vΔK1L vaccination induced a protective VACV-specific CD8+ T cell response and protected against a lethal VACV challenge. Thus, vΔK1L is the first vaccinia virus construct reported that caused a muted innate immune gene expression profile and decreased immune cell infiltration in an intradermal model of infection yet still elicited protective immunity.IMPORTANCE The vaccinia virus (VACV) K1 protein inhibits NF-κB activation among its other antagonistic functions. A virus lacking K1 (vΔK1L) was predicted to be less pathogenic because it would trigger a more robust antiviral immune response than VACV. Indeed, vΔK1L was less pathogenic in intradermally infected mouse ear pinnae. However, vΔK1L infection unexpectedly elicited dramatically reduced infiltration of innate immune cells into ears. This was likely due to decreased expression of cytokine and chemokine genes in vΔK1L-infected ears. As such, our finding contradicted observations from cell culture systems. Interestingly, vΔK1L conferred protective immunity against lethal VACV challenge. This suggests that the muted immune response triggered during vΔK1L infection remained sufficient to mount an effective protective response. Our results highlight the complexity and unpredictable nature of virus-host interactions, a relationship that must be understood to better comprehend virus pathogenesis or to manipulate viruses for use as vaccines.


Assuntos
Deleção de Genes , Imunidade Inata , Vírus Vaccinia/patogenicidade , Vaccinia/patologia , Proteínas Virais/genética , Fatores de Virulência/genética , Animais , Modelos Animais de Doenças , Camundongos , Vaccinia/virologia , Vírus Vaccinia/genética , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo
7.
Antiviral Res ; 144: 8-20, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28495463

RESUMO

Bioluminescence imaging (BLI) was used to follow dissemination of recombinant vaccinia virus (VACV) expressing luciferase (IHD-J-Luc) in BALB/c nu/nu mice treated post-challenge with monoclonal antibodies (MAbs) against L1 and B5 VACV proteins in a model of Progressive Vaccinia (PV). Areas Under the flux Curve (AUC) were calculated for viral loads in multiple organs in individual mice. Following scarification with 105 pfu, IHD-J-Luc VACV undergoes fast replication at the injection site and disseminates rapidly to the inguinal lymph nodes followed by spleen, liver, and axillary lymph nodes within 2-3 days and before primary lesions are visible at the site of scarification. Extension of survival in nude mice treated with a combination of anti-B5 and anti-L1 MAbs 24 h post challenge correlated with a significant reduction in viral load at the site of scarification and delayed systemic dissemination. Nude mice reconstituted with 104 T cells prior to challenge with IHD-J-Luc, and treated with MAbs post-challenge, survived infection, cleared the virus from all organs and scarification site, and developed anti-VACV IgG and VACV-specific polyfunctional CD8+ T cells that co-expressed the degranulation marker CD107a, and IFNγ and TNFα cytokines. All T cell reconstituted mice survived intranasal re-challenge with IHD-J-Luc (104 pfu) two months after the primary infection. Thus, using BLI to monitor VACV replication in a PV model, we showed that anti-VACV MAbs administered post challenge extended survival of nude mice and protected T cell reconstituted nude mice from lethality by reducing replication at the site of scarification and systemic dissemination of VACV.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antivirais/administração & dosagem , Modelos Animais de Doenças , Vírus Vaccinia/crescimento & desenvolvimento , Vaccinia/patologia , Vaccinia/terapia , Estruturas Animais/virologia , Animais , Fatores Imunológicos/administração & dosagem , Medições Luminescentes , Camundongos Endogâmicos BALB C , Camundongos Nus , Análise de Sobrevida , Resultado do Tratamento , Carga Viral , Proteínas Virais/imunologia , Imagem Corporal Total
8.
J Virol ; 91(13)2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28424281

RESUMO

Poxviruses use a complex strategy to escape immune control, by expressing immunomodulatory proteins that could limit their use as vaccine vectors. To test the role of poxvirus NF-κB pathway inhibitors A52, B15, and K7 in immunity, we deleted their genes in an NYVAC (New York vaccinia virus) strain that expresses HIV-1 clade C antigens. After infection of mice, ablation of the A52R, B15R, and K7R genes increased dendritic cell, natural killer cell, and neutrophil migration as well as chemokine/cytokine expression. Revertant viruses with these genes confirmed their role in inhibiting the innate immune system. To different extents, enhanced innate immune responses correlated with increased HIV Pol- and Gag-specific polyfunctional CD8 T cell and HIV Env-specific IgG responses induced by single-, double-, and triple-deletion mutants. These poxvirus proteins thus influence innate and adaptive cell-mediated and humoral immunity, and their ablation offers alternatives for design of vaccine vectors that regulate immune responses distinctly.IMPORTANCE Poxvirus vectors are used in clinical trials as candidate vaccines for several pathogens, yet how these vectors influence the immune system is unknown. We developed distinct poxvirus vectors that express heterologous antigens but lack different inhibitors of the central host-cell signaling pathway. Using mice, we studied the capacity of these viruses to induce innate and adaptive immune responses and showed that these vectors can distinctly regulate the magnitude and quality of these responses. These findings provide important insights into the mechanism of poxvirus-induced immune response and alternative strategies for vaccine vector design.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Subunidade p50 de NF-kappa B/antagonistas & inibidores , Vírus Vaccinia/imunologia , Vírus Vaccinia/patogenicidade , Proteínas Virais/metabolismo , Animais , Movimento Celular , Citocinas/biossíntese , Células Dendríticas/imunologia , Modelos Animais de Doenças , Deleção de Genes , Células Matadoras Naturais/imunologia , Camundongos , Neutrófilos/imunologia , Vaccinia/patologia , Vaccinia/virologia , Proteínas Virais/genética
9.
PLoS One ; 12(1): e0170070, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28081250

RESUMO

RATIONALE: Defects in filaggrin and STAT3 are associated with atopic dermatitis (AD) and susceptibility to severe skin infection. METHODS: We evaluated skin infection with the current smallpox vaccine, ACAM-2000, in immunosuppressed mice with combined cutaneous deficiency in filaggrin and STAT3. In parallel, early events post-infection with ACAM-2000 were investigated in cultured keratinocytes in which filaggrin expression was knocked down via siRNA. RESULTS: Immunosuppressed, filaggrin-deficient mice, treated with the topical STAT3 inhibitor Stattic® prior to ACAM-2000 infection, demonstrated rapid weight loss, prolonged vaccinia burden in skin, and dermatitis. The TGF-ß family ligand activin A was upregulated ten-fold in infected skin. Topically-applied ALK5/TGßR1 signaling inhibitor synergized with vaccinia immune globulin (VIG) to promote vaccinia clearance and limit weight loss. In cultured keratinocytes, filaggrin-directed siRNA inhibited programmed necrosis and inflammatory cytokine release induced by ACAM-2000, while viral growth was increased. CONCLUSIONS: Our findings may point to a novel role for filaggrin in early antiviral responses in skin. In wounded skin with underlying barrier defects, chronically elevated activin A levels may contribute to skin remodeling and cutaneous pathogen persistence. Inhibition of ALK5/TGFßR1 signaling may provide a novel co-therapeutic approach, together with VIG, to limit cutaneous spread of vaccinia.


Assuntos
Proteínas de Filamentos Intermediários/genética , Fator de Transcrição STAT3/genética , Vaccinia/patologia , Ativinas/análise , Ativinas/metabolismo , Animais , Anticorpos/imunologia , Citocinas/metabolismo , Dermatite/etiologia , Dermatite/metabolismo , Dermatite/virologia , Proteínas Filagrinas , Proteínas de Filamentos Intermediários/antagonistas & inibidores , Proteínas de Filamentos Intermediários/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Índice de Gravidade de Doença , Pele/metabolismo , Regulação para Cima , Vaccinia/complicações , Vaccinia/virologia , Vírus Vaccinia/imunologia
10.
Int J Parasitol ; 47(1): 1-10, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28003150

RESUMO

The aim of this work was to elucidate the immunopathological mechanisms of how helminths may influence the course of a viral infection, using a murine model. Severe virulence, a relevant increase in the virus titres in the lung and a higher mortality rate were observed in Ascaris and Vaccinia virus (VACV) co-infected mice, compared with VACV mono-infected mice. Immunopathological analysis suggested that the ablation of CD8+ T cells, the marked reduction of circulating CD4+ T cells producing IFN-γ, and the robust pulmonary inflammation were associated with the increase of morbidity/mortality in co-infection and subsequently with the negative impact of concomitant pulmonary ascariasis and respiratory VACV infection for the host. On the other hand, when evaluating the impact of the co-infection on the parasitic burden, co-infected mice presented a marked decrease in the total number of migrating Ascaris lung-stage larvae in comparison with Ascaris mono-infection. Taken together, our major findings suggest that Ascaris and VACV co-infection may potentiate the virus-associated pathology by the downmodulation of the VACV-specific immune response. Moreover, this study provides new evidence of how helminth parasites may influence the course of a coincident viral infection.


Assuntos
Ascaríase/virologia , Ascaris/imunologia , Coinfecção/imunologia , Pneumonia/parasitologia , Vírus Vaccinia/imunologia , Vaccinia/etiologia , Animais , Ascaríase/imunologia , Linfócitos T CD8-Positivos/imunologia , Coinfecção/parasitologia , Coinfecção/virologia , Citocinas/imunologia , Modelos Animais de Doenças , Feminino , Interferon gama/imunologia , Larva/parasitologia , Pulmão/imunologia , Pulmão/parasitologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/imunologia , Pneumonia/virologia , Suínos , Vaccinia/imunologia , Vaccinia/patologia , Vaccinia/virologia , Carga Viral
11.
Equine Vet J ; 49(2): 221-224, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26875684

RESUMO

REASONS FOR PERFORMING STUDY: In August 2014, an outbreak of oral exanthematous disease in equids was reported in Brazil, affecting 11 donkeys and 3 mules. OBJECTIVES: To investigate if Vaccinia virus (VACV) was the aetiological agent in this outbreak. STUDY DESIGN: Investigation of clinical cases using serological, molecular and phylogenetic approaches. METHODS: To analyse the presence of neutralising antibodies against VACV, samples were submitted in triplicate to a plaque-reduction neutralisation test (PRNT50% ). On the basis of previous studies which detected VACV DNA in sera, we submitted extracted DNA samples to different polymerase chain reaction (PCR) platforms targeting Orthopoxvirus (OPV) genes (C11R, A56R and A26L). The PCR products were directly sequenced in both orientations using specific primers and capillary electrophoresis. The alignment and phylogenetic analysis of the A26L and A56R nucleotide sequences (maximum likelihood) were prepared with the obtained nucleotide fragments. RESULTS: Serological and molecular data suggested VACV as the aetiological agent. The neutralising antibodies against OPV were detected in 5 (55.5%) of the equids, with titres ≥40 neutralising u/ml. Based on the results obtained from all PCR platforms, all samples were positive for OPV: 9 (100%) for A56R, 4 (44.4%) for C11R and 3 (33.3%) for A26L. The alignment of the nucleotide sequences of the A26L and A56R fragments revealed that the samples were highly similar to the homologous genes from other Brazilian VACV Group 1 isolates (98.8% identity on average). Furthermore, both the A26L and A56R sequences showed signature deletions also present in the sequences of Group 1 VACV isolates from Brazil. CONCLUSIONS: Our data raises questions about the role of equids in the chain of VACV epidemiology. The surveillance of equids in VACV-affected areas worldwide is relevant.


Assuntos
Surtos de Doenças/veterinária , Equidae , Exantema/veterinária , Doenças da Boca/veterinária , Vírus Vaccinia/isolamento & purificação , Vaccinia/veterinária , Animais , Brasil/epidemiologia , Exantema/patologia , Exantema/virologia , Doenças da Boca/epidemiologia , Doenças da Boca/patologia , Doenças da Boca/virologia , Vaccinia/diagnóstico , Vaccinia/epidemiologia , Vaccinia/patologia
12.
J Exp Med ; 213(13): 3075-3086, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27899444

RESUMO

Tissue-resident memory CD8+ T cells (TRM) constitute a major component of the immune-surveillance system in nonlymphoid organs. Local, noncognate factors are both necessary and sufficient to support the programming of TRM cell fate in tissue-infiltrating T cells. Recent evidence suggests that TCR signals received in infected nonlymphoid tissues additionally contribute to TRM cell formation. Here, we asked how antigen-dependent pathways influence the generation of skin-resident memory T cells that arise from a polyclonal repertoire of cells induced by infection with an antigenically complex virus and recombinant vaccine vector. We found that CD8+ T cells of different specificities underwent antigen-dependent competition in the infected tissue, which shaped the composition of the local pool of TRM cells. This local cross-competition was active for T cells recognizing antigens that are coexpressed by infected cells. In contrast, TRM cell development remained largely undisturbed by the presence of potential competitors when antigens expressed in the same tissue were segregated through infection with antigenically distinct viral quasispecies. Functionally, local cross-competition might serve as a gatekeeping mechanism to regulate access to the resident memory niche and to fine-tune the local repertoire of antiviral TRM cells.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Vírus Vaccinia/imunologia , Vaccinia/imunologia , Animais , Antígenos Virais/genética , Linfócitos T CD8-Positivos/patologia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética , Transdução de Sinais/genética , Vaccinia/genética , Vaccinia/patologia , Vírus Vaccinia/genética
13.
Viruses ; 8(12)2016 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-27973399

RESUMO

Vaccinia virus (VACV) has been implicated in infections of dairy cattle and humans, and outbreaks have substantially impacted local economies and public health in Brazil. During a 2005 outbreak, a VACV strain designated Serro 2 virus (S2V) was collected from a 30-year old male milker. Our aim was to phenotypically and genetically characterize this VACV Brazilian isolate. S2V produced small round plaques without associated comets when grown in BSC40 cells. Furthermore, S2V was less virulent than the prototype strain VACV-Western Reserve (WR) in a murine model of intradermal infection, producing a tiny lesion with virtually no surrounding inflammation. The genome of S2V was sequenced by primer walking. The coding region spans 184,572 bp and contains 211 predicted genes. Mutations in envelope genes specifically associated with small plaque phenotypes were not found in S2V; however, other alterations in amino acid sequences within these genes were identified. In addition, some immunomodulatory genes were truncated in S2V. Phylogenetic analysis using immune regulatory-related genes, besides the hemagglutinin gene, segregated the Brazilian viruses into two clusters, grouping the S2V into Brazilian VACV group 1. S2V is the first naturally-circulating human-associated VACV, with a low passage history, to be extensively genetically and phenotypically characterized.


Assuntos
Genoma Viral , Filogenia , Análise de Sequência de DNA , Vírus Vaccinia/genética , Vírus Vaccinia/isolamento & purificação , Vaccinia/virologia , Adulto , Animais , Brasil , Linhagem Celular , Modelos Animais de Doenças , Genes Virais , Humanos , Masculino , Camundongos , Homologia de Sequência , Vaccinia/patologia , Vírus Vaccinia/classificação , Vírus Vaccinia/fisiologia , Ensaio de Placa Viral , Virulência , Fatores de Virulência/genética
14.
Cell Rep ; 16(7): 1800-9, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27498869

RESUMO

The nature and anatomic location of the protective memory CD8(+) T cell subset induced by intranasal vaccination remain poorly understood. We developed a vaccination model to assess the anatomic location of protective memory CD8(+) T cells and their role in lower airway infections. Memory CD8(+) T cells elicited by local intranasal, but not systemic, vaccination with an engineered non-replicative CD8(+) T cell-targeted antigen confer enhanced protection to a lethal respiratory viral challenge. This protection depends on a distinct CXCR3(LO) resident memory CD8(+) T (Trm) cell population that preferentially localizes to the pulmonary interstitium. Because they are positioned close to the mucosa, where infection occurs, interstitial Trm cells act before inflammation can recruit circulating memory CD8(+) T cells into the lung tissue. This results in a local protective immune response as early as 1 day post-infection. Hence, vaccine strategies that induce lung interstitial Trm cells may confer better protection against respiratory pathogens.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Infecções Respiratórias/prevenção & controle , Vaccinia/prevenção & controle , Vacinas Virais/administração & dosagem , Administração Intranasal , Sequência de Aminoácidos , Animais , Antígenos Virais/química , Peso Corporal/efeitos dos fármacos , Linfócitos T CD8-Positivos/virologia , Expressão Gênica , Imunidade nas Mucosas/efeitos dos fármacos , Imunofenotipagem , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Transgênicos , Receptores CXCR3/genética , Receptores CXCR3/imunologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Vacinação , Vaccinia/imunologia , Vaccinia/patologia , Vaccinia/virologia , Vírus Vaccinia/química , Vírus Vaccinia/efeitos dos fármacos , Vírus Vaccinia/crescimento & desenvolvimento , Vírus Vaccinia/patogenicidade , Carga Viral/efeitos dos fármacos , Vacinas Virais/biossíntese
15.
J Exp Med ; 213(6): 951-66, 2016 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-27217536

RESUMO

Tissue-resident memory (Trm) CD8(+) T cells are functionally distinct from their circulating counterparts and are potent mediators of host protection against reinfection. Whether local recognition of antigen in nonlymphoid tissues during infection can impact the formation of Trm populations remains unresolved. Using skin infections with vaccinia virus (VacV)-expressing model antigens, we found that local antigen recognition had a profound impact on Trm formation. Activated CD8(+) T cells trafficked to VacV-infected skin in an inflammation-dependent, but antigen-independent, manner. However, after viral clearance, there was a subsequent ∼50-fold increase in Trm formation when antigen was present in the tissue microenvironment. Secondary antigen stimulation in nonlymphoid tissue caused CD8(+) T cells to rapidly express CD69 and be retained at the site of infection. Finally, Trm CD8(+) T cells that formed during VacV infection in an antigen-dependent manner became potent stimulators of localized antigen-specific inflammatory responses in the skin. Thus, our studies indicate that the presence of antigen in the nonlymphoid tissue microenvironment plays a critical role in the formation of functional Trm CD8(+) T cell populations, a finding with relevance for both vaccine design and prevention of inflammatory disorders.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Modelos Imunológicos , Vírus Vaccinia/imunologia , Vaccinia/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos Virais/genética , Linfócitos T CD8-Positivos/patologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Camundongos , Camundongos Transgênicos , Pele/imunologia , Pele/patologia , Vaccinia/genética , Vaccinia/patologia , Vírus Vaccinia/genética
16.
Immunity ; 43(4): 660-73, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26453379

RESUMO

Humans with Wiskott-Aldrich syndrome display a progressive immunological disorder associated with compromised Wiskott-Aldrich Syndrome Interacting Protein (WIP) function. Mice deficient in WIP recapitulate such an immunodeficiency that has been attributed to T cell dysfunction; however, any contribution of B cells is as yet undefined. Here we have shown that WIP deficiency resulted in defects in B cell homing, chemotaxis, survival, and differentiation, ultimately leading to diminished germinal center formation and antibody production. Furthermore, in the absence of WIP, several receptors, namely the BCR, BAFFR, CXCR4, CXCR5, CD40, and TLR4, were impaired in promoting CD19 co-receptor activation and subsequent PI3 kinase (PI3K) signaling. The underlying mechanism was due to a distortion in the actin and tetraspanin networks that lead to altered CD19 cell surface dynamics. In conclusion, our findings suggest that, by regulating the cortical actin cytoskeleton, WIP influences the function of CD19 as a general hub for PI3K signaling.


Assuntos
Antígenos CD19/fisiologia , Linfócitos B/imunologia , Proteínas de Transporte/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/imunologia , Citoesqueleto de Actina/ultraestrutura , Actinas/análise , Animais , Formação de Anticorpos , Linfócitos B/efeitos dos fármacos , Linfócitos B/enzimologia , Linfócitos B/ultraestrutura , Proteínas de Transporte/genética , Células Cultivadas , Quimiocinas/farmacologia , Quimiocinas/fisiologia , Quimiotaxia/efeitos dos fármacos , Proteínas do Citoesqueleto , Centro Germinativo/imunologia , Centro Germinativo/patologia , Haptenos , Hemocianinas/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Linfopoese , Proteínas de Membrana/imunologia , Camundongos , Fosforilação , Plasmócitos/imunologia , Processamento de Proteína Pós-Traducional , Quimera por Radiação , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Quimiocinas/fisiologia , Tetraspaninas/análise , Vaccinia/imunologia , Vaccinia/patologia
17.
BMC Cancer ; 15: 704, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26471278

RESUMO

BACKGROUND: JX594 is an oncolytic poxvirus derived from Wyeth strain vaccinia virus. We reported the presentation of cutaneous and mucosal pustules containing laboratory-confirmed JX594 in a patient following injection of JX594. CASE PRESENTATION: A 36-year-old man was diagnosed hepatitis B virus-associated hepatocellular carcinoma on September 19, 2011. Despite treatment with entecavir, radiofrequency ablation and transarterial chemoembolization for recurrent local tumors, the tumors recurred in both lobes and lung metastases were detected by computed tomography on September 12, 2012. The patient was treated with JX594 (Pexa-vec®) via intravenous injection on December 19, 2012. No apparent adverse effects were observed following intravenous injection other than a single fever episode. However, pustular lesions were detected on both sides of the tongue dorsum and on the proximal interphalangeal joint of the right middle finger on December 25, 1012. Biopsy samples analyzed by PCR identified the presence of the JX-594-specific hGM-CSF transgene and the disrupted viral thymidine kinase gene. Following aspiration of the lesion a scab formed that resolved within 14 days without necessitating additional treatment. CONCLUSION: Our case completely recovered and did not develop systemic or recurrent disease, the presentation of a few pustules may not necessarily require that treatment with JX594 be interrupted for patients with advanced hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Vírus Oncolíticos/genética , Poxviridae/genética , Adulto , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virologia , Vírus da Hepatite B/patogenicidade , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Masculino , Terapia Viral Oncolítica/métodos , Timidina Quinase/genética , Vaccinia/patologia , Vaccinia/virologia , Vírus Vaccinia/genética
18.
PLoS Pathog ; 11(9): e1005151, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26334635

RESUMO

Vaccinia virus (VACV) is the prototypic orthopoxvirus and the vaccine used to eradicate smallpox. Here we show that VACV strain Western Reserve protein 169 is a cytoplasmic polypeptide expressed early during infection that is excluded from virus factories and inhibits the initiation of cap-dependent and cap-independent translation. Ectopic expression of protein 169 causes the accumulation of 80S ribosomes, a reduction of polysomes, and inhibition of protein expression deriving from activation of multiple innate immune signaling pathways. A virus lacking 169 (vΔ169) replicates and spreads normally in cell culture but is more virulent than parental and revertant control viruses in intranasal and intradermal murine models of infection. Intranasal infection by vΔ169 caused increased pro-inflammatory cytokines and chemokines, infiltration of pulmonary leukocytes, and lung weight. These alterations in innate immunity resulted in a stronger CD8+ T-cell memory response and better protection against virus challenge. This work illustrates how inhibition of host protein synthesis can be a strategy for virus suppression of innate and adaptive immunity.


Assuntos
Imunidade Adaptativa , Interações Hospedeiro-Patógeno , Imunidade Inata , Iniciação Traducional da Cadeia Peptídica , Vírus Vaccinia/fisiologia , Vaccinia/virologia , Proteínas Virais/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Memória Imunológica , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Vaccinia/imunologia , Vaccinia/metabolismo , Vaccinia/patologia , Vírus Vaccinia/imunologia , Vírus Vaccinia/patogenicidade , Proteínas Virais/genética , Virulência
19.
Vaccine ; 33(41): 5396-5405, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26319070

RESUMO

Buffalopox virus (BPXV), an Indian variant of vaccinia virus (VACV), is a zoonotic agent and affects buffaloes, cattle and humans. A27L is one of the conserved major immuno-dominant envelope proteins of orthopox viruses (OPVs) involved in viral entry/maturation and elicits neutralizing antibodies. In this study, the A27L gene of BPXV-Vij/96 strain encoding recombinant mature A27L (21S to E110) and C-terminal truncated A27L-LZD (21S to N84aa) proteins were cloned and over-expressed in Escherichia coli as fusion proteins. Structurally, A27L of BPXV was similar to that of VACV and found to contain four regions including a potential coiled-coil motif (CCM) in the centre (43 to 84aa). Oligomerization of recombinant A27L fusion protein (∼30 kDa) leads to the formation of dimer/trimers/tetramers under non-reducing conditions. Further, the purified rA27L protein was used for active immunization of rabbit (250 µg/rabbit) and adult mice (10 µg and 50 µg/mice) with or without adjuvants (FCA, alum and CpG). Immune response measured by using indirect-ELISA and SNT revealed a gradual increase in antigen specific serum IgG as well as neutralization antibody titers. Upon challenge with virulent BPXV strain, a protection of 60% was observed in suckling mice passively administered with anti-rA27L sera. No cross-reactivity of rA27L protein with hyperimmune sera against ORFV, GTPV, SPPV, PPRV, FMDV and BTV was noticed in indirect-ELISA. The study indicated that the rA27L protein is a safe and potential prophylactic as well as diagnostic antigen for buffalopox.


Assuntos
Proteínas Recombinantes , Vírus Vaccinia/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Bovinos , Sequência Conservada , Soros Imunes/imunologia , Imunização , Camundongos , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Terciária de Proteína , Coelhos , Sensibilidade e Especificidade , Vaccinia/diagnóstico , Vaccinia/imunologia , Vaccinia/patologia , Vírus Vaccinia/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/isolamento & purificação
20.
Immunology ; 145(1): 34-49, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25382035

RESUMO

Factors influencing T-cell responses are important for vaccine development but are incompletely understood. Here, vaccinia virus (VACV) protein N1 is shown to impair the development of both effector and memory CD8(+) T cells and this correlates with its inhibition of nuclear factor-κB (NF-κB) activation. Infection with VACVs that either have the N1L gene deleted (vΔN1) or contain a I6E mutation (vN1.I6E) that abrogates its inhibition of NF-κB resulted in increased central and memory CD8(+) T-cell populations, increased CD8(+) T-cell cytotoxicity and lower virus titres after challenge. Furthermore, CD8(+) memory T-cell function was increased following infection with vN1.I6E, with more interferon-γ production and greater protection against VACV infection following passive transfer to naive mice, compared with CD8(+) T cells from mice infected with wild-type virus (vN1.WT). This demonstrates the importance of NF-κB activation within infected cells for long-term CD8(+) T-cell memory and vaccine efficacy. Further, it provides a rationale for deleting N1 from VACV vectors to enhance CD8(+) T-cell immunogenicity, while simultaneously reducing virulence to improve vaccine safety.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , NF-kappa B/antagonistas & inibidores , Vírus Vaccinia/imunologia , Vaccinia/imunologia , Proteínas Virais/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Feminino , Camundongos , Mutação de Sentido Incorreto , NF-kappa B/genética , NF-kappa B/imunologia , Vaccinia/genética , Vaccinia/patologia , Vírus Vaccinia/genética , Proteínas Virais/genética , Vacinas Virais/genética , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...