Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Transbound Emerg Dis ; 67(1): 80-97, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31379093

RESUMO

The genus Capripoxvirus in the subfamily Chordopoxvirinae, family Poxviridae, comprises sheeppox virus (SPPV), goatpox virus (GTPV) and lumpy skin disease virus (LSDV), which cause the eponymous diseases across parts of Africa, the Middle East and Asia. These diseases cause significant economic losses and can have a devastating impact on the livelihoods and food security of small farm holders. So far, only live classically attenuated SPPV, GTPV and LSDV vaccines are commercially available and the history, safety and efficacy of many have not been well established. Here, we report 13 new capripoxvirus genome sequences, including the hairpin telomeres, from both pathogenic field isolates and vaccine strains. We have also updated the genome annotations to incorporate recent advances in our understanding of poxvirus biology. These new genomes and genes grouped phenetically with other previously sequenced capripoxvirus strains, and these new alignments collectively identified several recurring alterations in genes thought to modulate virulence and host range. In particular, some of the many large capripoxvirus ankyrin and kelch-like proteins are commonly mutated in vaccine strains, while the variola virus B22R-like gene homolog has also been disrupted in many vaccine isolates. Among these vaccine isolates, frameshift mutations are especially common and clearly present a risk of reversion to wild type in vaccines bearing these mutations. A consistent pattern of gene inactivation from LSDV to GTPV and then SPPV is also observed, much like the pattern of gene loss in orthopoxviruses, but, rather surprisingly, the overall genome size of ~150 kbp remains relatively constant. These data provide new insights into the evolution of capripoxviruses and the determinants of pathogenicity and host range. They will find application in the development of new vaccines with better safety, efficacy and trade profiles.


Assuntos
Capripoxvirus/genética , Variação Genética , Genoma Viral/genética , Especificidade de Hospedeiro/genética , Infecções por Poxviridae/veterinária , Doenças dos Ovinos/virologia , África , Animais , Ásia , Evolução Biológica , Capripoxvirus/imunologia , Capripoxvirus/patogenicidade , Capripoxvirus/fisiologia , Células Cultivadas , Especiação Genética , Índia , Masculino , Oriente Médio , Mutação , Infecções por Poxviridae/prevenção & controle , Infecções por Poxviridae/virologia , Ovinos , Doenças dos Ovinos/prevenção & controle , Testículo/virologia , Vacinas Virais/imunologia , Virulência
2.
J Virol ; 93(4)2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30518648

RESUMO

The life cycle of influenza A virus (IAV) is modulated by various cellular host factors. Although previous studies indicated that IAV infection is controlled by HDAC6, the deacetylase involved in the regulation of PA remained unknown. Here, we demonstrate that HDAC6 acts as a negative regulator of IAV infection by destabilizing PA. HDAC6 binds to and deacetylates PA, thereby promoting the proteasomal degradation of PA. Based on mass spectrometric analysis, Lys(664) of PA can be deacetylated by HDAC6, and the residue is crucial for PA protein stability. The deacetylase activity of HDAC6 is required for anti-IAV activity, because IAV infection was enhanced due to elevated IAV RNA polymerase activity upon HDAC6 depletion and an HDAC6 deacetylase dead mutant (HDAC6-DM; H216A, H611A). Finally, we also demonstrate that overexpression of HDAC6 suppresses IAV RNA polymerase activity, but HDAC6-DM does not. Taken together, our findings provide initial evidence that HDAC6 plays a negative role in IAV RNA polymerase activity by deacetylating PA and thus restricts IAV RNA transcription and replication.IMPORTANCE Influenza A virus (IAV) continues to threaten global public health due to drug resistance and the emergence of frequently mutated strains. Thus, it is critical to find new strategies to control IAV infection. Here, we discover one host protein, HDAC6, that can inhibit viral RNA polymerase activity by deacetylating PA and thus suppresses virus RNA replication and transcription. Previously, it was reported that IAV can utilize the HDAC6-dependent aggresome formation mechanism to promote virus uncoating, but HDAC6-mediated deacetylation of α-tubulin inhibits viral protein trafficking at late stages of the virus life cycle. These findings together will contribute to a better understanding of the role of HDAC6 in regulating IAV infection. Understanding the molecular mechanisms of HDAC6 at various periods of viral infection may illuminate novel strategies for developing antiviral drugs.


Assuntos
Desacetilase 6 de Histona/metabolismo , Vírus da Influenza A/metabolismo , Células A549 , Acetilação , Animais , Antivirais/farmacologia , Linhagem Celular , RNA Polimerases Dirigidas por DNA/metabolismo , Cães , Células HEK293 , Desacetilase 6 de Histona/fisiologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Vírus da Influenza A/patogenicidade , Influenza Humana/genética , Influenza Humana/imunologia , Células Madin Darby de Rim Canino , RNA Viral/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/genética
3.
Immunology ; 153(4): 532-544, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29068058

RESUMO

The concept of dendritic cell (DC) maturation generally refers to the changes in morphology and function of DCs. Conventionally, DC maturity is based on three criteria: loss of endocytic ability, gain of high-level capacity to present antigens and induce proliferation of T cells, and mobility of DCs toward high concentrations of CCL19. Impairment of DC maturation has been suggested as the main reason for infectivity or chronicity of several infectious agents. In the case of hepatitis C virus, this has been a matter of controversy for the last two decades. However, insufficient attention has been paid to the method of ex vivo maturation as the possible source of such controversies. We previously reported striking differences between DCs matured with different methods, so we propose the use of a standard quantitative index to determine the level of maturity in DCs as an approach to compare results from different studies. We designed and formulated a mathematically calculated index to numerically define the level of maturity based on experimental data from ex vivo assays. This introduces a standard maturation index (SMI) and weighted maturation index (WMI) based on strictly standardized mean differences between different methods of generating mature DCs. By calculating an SMI and a WMI, numerical values were assigned to the level of maturity achieved by DCs matured with different methods. SMI and WMI could be used as a standard tool to compare diversely generated mature DCs and so better interpret outcomes of ex vivo and in vivo studies with mature DCs.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Modelos Estatísticos , Citometria de Fluxo , Voluntários Saudáveis , Humanos
4.
Nanomedicine ; 13(7): 2169-2178, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28579436

RESUMO

The most promising strategy to sustainably prevent infectious diseases is vaccination. However, emerging as well as re-emerging diseases still constitute a considerable threat. Furthermore, lack of compliance and logistic constrains often result in the failure of vaccination campaigns. To overcome these hurdles, novel vaccination strategies need to be developed, which fulfill maximal safety requirements, show maximal efficiency and are easy to administer. Mucosal vaccines constitute promising non-invasive approaches able to match these demands. Here we demonstrate that nanoparticle (polyphosphazenes)-based vaccine formulations including c-di-AMP as adjuvant, cationic innate defense regulator peptides (IDR) and ovalbumin (OVA) as model antigen were able to stimulate strong humoral and cellular immune responses, which conferred protection against the OVA expressing influenza strain A/WSN/OVAI (H1N1). The presented results confirm the potency of nanoparticle-based vaccine formulations to deliver antigens across the mucosal barrier, but also demonstrate the necessity to include adjuvants to stimulate efficient antigen-specific immune responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Fosfatos de Dinucleosídeos/administração & dosagem , Vacinas contra Influenza/administração & dosagem , Nanopartículas/química , Compostos Organofosforados/química , Infecções por Orthomyxoviridae/prevenção & controle , Ovalbumina/administração & dosagem , Polímeros/química , Adjuvantes Imunológicos/uso terapêutico , Administração Intranasal , Animais , Fosfatos de Dinucleosídeos/uso terapêutico , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/uso terapêutico , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Ovalbumina/uso terapêutico , Vacinação/métodos
5.
Infect Immun ; 85(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28167667

RESUMO

Maternal vaccination represents a potential strategy to protect both the mother and the offspring against life-threatening infections. This protective role has mainly been associated with antibodies, but the role of cell-mediated immunity, in particular passively transferred cytokines, is not well understood. Here, using a pertussis model, we have demonstrated that immunization of pregnant sows with heat-inactivated bacteria leads to induction of a wide range of cytokines (e.g., tumor necrosis factor alpha [TNF-α], gamma interferon [IFN-γ], interleukin-6 [IL-6], IL-8, and IL-12/IL-23p40) in addition to pertussis-specific antibodies. These cytokines can be detected in the sera and colostrum/milk of vaccinated sows and subsequently were detected at significant levels in the serum and bronchoalveolar lavage fluid of piglets born to vaccinated sows together with pertussis-specific antibodies. In contrast, active vaccination of newborn piglets with heat-inactivated bacteria induced high levels of specific IgG and IgA but no cytokines. Although the levels of antibodies in vaccinated piglets were comparable to those of passively transferred antibodies, no protection against Bordetella pertussis infection was observed. Thus, our results demonstrate that a combination of passively transferred cytokines and antibodies is crucial for disease protection. The presence of passively transferred cytokines/antibodies influences the cytokine secretion ability of splenocytes in the neonate, which provides novel evidence that maternal immunization can influence the newborn's cytokine milieu and may impact immune cell differentiation (e.g., Th1/Th2 phenotype). Therefore, these maternally derived cytokines may play an essential role both as mediators of early defense against infections and possibly as modulators of the immune repertoire of the offspring.


Assuntos
Bordetella pertussis/imunologia , Citocinas/administração & dosagem , Imunidade Materno-Adquirida , Imunização Passiva , Coqueluche/imunologia , Coqueluche/microbiologia , Animais , Animais Recém-Nascidos , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Citocinas/biossíntese , Citocinas/sangue , Feminino , Pulmão/patologia , Gravidez , Baço/imunologia , Baço/metabolismo , Suínos , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia
6.
Zebrafish ; 13 Suppl 1: S153-63, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27248438

RESUMO

The zebrafish model system is helping researchers improve the health and welfare of people and animals and has become indispensable for advancing biomedical research. As genetic engineering is both resource intensive and time-consuming, sharing successfully developed genetically modified zebrafish lines throughout the international community is critical to research efficiency and to maximizing the millions of dollars in research funding. New restrictions on importation of zebrafish into Canada based on putative susceptibility to infection by the spring viremia of carp virus (SVCV) have been imposed on the scientific community. In this commentary, we review the disease profile of SVCV in fish, discuss the findings of the Canadian government's scientific assessment, how the interpretations of their assessment differ from that of the Canadian research community, and describe the negative impact of these regulations on the Canadian research community and public as it pertains to protecting the health of Canadians.


Assuntos
Comércio/legislação & jurisprudência , Doenças dos Peixes/prevenção & controle , Doenças dos Peixes/transmissão , Regulamentação Governamental , Infecções por Rhabdoviridae/veterinária , Peixe-Zebra , Animais , Canadá , Doenças dos Peixes/virologia , Rhabdoviridae/fisiologia , Infecções por Rhabdoviridae/prevenção & controle , Infecções por Rhabdoviridae/transmissão , Infecções por Rhabdoviridae/virologia
7.
Vaccine ; 34(7): 923-32, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26776470

RESUMO

BACKGROUND: Therapeutic vaccination is a novel treatment approach for chronic hepatitis B, but only had limited success so far. We hypothesized that optimized vaccination schemes have increased immunogenicity, and aimed at increasing therapeutic hepatitis B vaccine efficacy. METHODS: Modified Vaccinia virus Ankara (MVA) expressing hepatitis B virus (HBV) antigens was used to boost protein-prime vaccinations in wildtype and HBV-transgenic (HBVtg) mice. RESULTS: Protein-prime/MVA-boost vaccination was able to overcome HBV-specific tolerance in HBVtg mice with low and medium but not with high antigenemia. HBV-specific antibody titers, CD8+ T-cell frequencies and polyfunctionality inversely correlated with HBV antigen levels. However, optimization of the adjuvant formulation, increasing the level of antigen expression and utilization of HBsAg of heterologous subtype induced HBV-specific CD8+ and CD4+ T-cells and neutralizing antibodies even in high-antigenemic HBVtg mice. CONCLUSIONS: Our results indicate that high HBV antigen levels limit the immunological responsiveness to therapeutic vaccination but optimization of the vaccine formulation can overcome tolerance even in the presence of high antigenemia. These findings have important implications for the development of future therapeutic hepatitis B vaccination strategies and potentially also for the stratification of chronic hepatitis B patients for therapeutic vaccination.


Assuntos
Vacinas contra Hepatite B/imunologia , Hepatite B/prevenção & controle , Tolerância Imunológica , Vaccinia virus , Animais , Anticorpos Neutralizantes/sangue , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Anticorpos Anti-Hepatite B/sangue , Antígenos do Núcleo do Vírus da Hepatite B/imunologia , Antígenos de Superfície da Hepatite B/sangue , Antígenos de Superfície da Hepatite B/imunologia , Antígenos E da Hepatite B/sangue , Imunização Secundária , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Testes de Neutralização
8.
Antiviral Res ; 123: 39-49, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26341190

RESUMO

Sheep and goat pox continue to be important livestock diseases that pose a major threat to the livestock industry in many regions in Africa and Asia. Currently, several live attenuated vaccines are available and used in endemic countries to control these diseases. One of these is a partially attenuated strain of lumpy skin disease virus (LSDV), KS-1, which provides cross-protection against both sheep pox and goat pox. However, when used in highly stressed dairy cattle to protect against lumpy skin disease (LSD) the vaccine can cause clinical disease. In order to develop safer vaccines effective against all three diseases, a pathogenic strain of LSDV (Warmbaths [WB], South Africa) was attenuated by removing a putative virulence factor gene (IL-10-like) using gene knockout (KO) technology. This construct (LSDV WB005KO) was then evaluated as a vaccine for sheep and goats against virulent capripoxvirus challenge. Sheep and goats were vaccinated with the construct and the animals were observed for 21days. The vaccine appeared to be safe, and did not cause disease, although it induced minor inflammation at the injection site similar to that caused by other attenuated sheep and goat pox vaccines. In addition, no virus replication was detected in blood, oral or nasal swabs using real-time PCR following vaccination and low levels of neutralising antibodies were detected in both sheep and goats. Leukocytes isolated from vaccinated animals following vaccination elicited capripoxvirus-specific IFN-γ secretion, suggesting that immunity was also T-cell mediated. Following challenge with virulent capripoxvirus, vaccinated sheep and goats were found to be completely protected and exhibited no clinical disease. Furthermore, real-time PCR of blood samples at various time points suggested that viremia was absent in both groups of vaccinated animals, as opposed to capripoxvirus-related clinical disease and viremia observed in the unvaccinated animals. These findings suggest that this novel knockout strain of LSDV has potential as a vaccine to protect livestock against sheep pox and goat pox.


Assuntos
Doenças das Cabras/prevenção & controle , Interleucina-10/deficiência , Vírus da Doença Nodular Cutânea/imunologia , Infecções por Poxviridae/veterinária , Doenças dos Ovinos/prevenção & controle , Proteínas Virais/genética , Vacinas Virais/imunologia , Animais , Técnicas de Inativação de Genes , Doenças das Cabras/imunologia , Doenças das Cabras/virologia , Cabras , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Vírus da Doença Nodular Cutânea/genética , Infecções por Poxviridae/imunologia , Infecções por Poxviridae/prevenção & controle , Ovinos , Doenças dos Ovinos/imunologia , Doenças dos Ovinos/virologia , Análise de Sobrevida , Resultado do Tratamento , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Fatores de Virulência/deficiência
9.
Virus Res ; 197: 116-26, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25529439

RESUMO

The UL47 gene product, VP8, is one of the most abundant tegument proteins of bovine herpesvirus-1 (BoHV-1). Deletion of VP8 leads to impaired growth in tissue culture, and VP8 is indispensable for BHV-1 replication in cattle. To elucidate the biological functions of VP8, we explored its interaction with mRNAs of immediate early (bICP0), early (gB, gD) and late (gC) genes of BoHV-1. FLAG-tagged VP8 was pulled down from COS-7 cells co-transfected with plasmids encoding VP8 and either gB, gC, gD or bICP0. This was followed by RNA extraction, cDNA synthesis and qPCR, which demonstrated binding of VP8 to bICP0, gB, gC and gD mRNAs in the cytoplasm and nucleus. These results were supported by co-localization of VP8 with bICP0, gB, gC and gD mRNAs in the nucleus as determined by confocal microscopy. Amino acids 259-342, located in the conserved portion of UL47 homologues, were found to contain the RNA binding region on VP8. To further characterize these interactions, Northwestern blotting was performed by immobilizing VP8 on a nitrocellulose membrane followed by hybridization with in vitro transcribed bICP0 mRNA. The results demonstrated binding of VP8 to intron-less mRNA but not intron-containing mRNA of bICP0. In addition, the interaction of VP8 with bICP0 mRNA was confirmed in vitro by RNA electrophoretic mobility shift assay, which also showed that the zinc finger and acidic domains both interact with VP8. Based on these results, we concluded that VP8 binds to intron-less mRNAs of bICP0, gB, gC and gD.


Assuntos
Proteínas do Capsídeo/metabolismo , Herpesvirus Bovino 1/fisiologia , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Células COS , Proteínas do Capsídeo/genética , Núcleo Celular/virologia , Chlorocebus aethiops , Citoplasma/virologia , Ensaio de Desvio de Mobilidade Eletroforética , Microscopia Confocal , Plasmídeos , Ligação Proteica , Proteínas de Ligação a RNA/genética
10.
Clin Vaccine Immunol ; 22(1): 79-90, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25378352

RESUMO

Protective efficacy against bovine herpesvirus 1 (BoHV-1) has been demonstrated to be induced by a plasmid encoding bovine neutrophil beta-defensin 3 (BNBD3) as a fusion construct with truncated glycoprotein D (tgD). However, in spite of the increased cell-mediated immune responses induced by this DNA vaccine, the clinical responses of BoHV-1-challenged cattle were not reduced over those observed in animals vaccinated with the plasmid encoding tgD alone; this might have been because the vaccine failed to improve humoral responses. We hypothesized that an alternative vaccine design strategy that utilized the DNA vaccine pMASIA-tgD as a complex with BNBD3 might improve humoral responses while maintaining robust Th1-type cell-mediated responses. C57BL/6 mice were vaccinated with pMASIA-tgD complexed with 0, 0.01875, 0.1875, or 1.875 nmol of a stable synthesized analog of BNBD3 (aBNBD3). The best results were seen in mice immunized with the vaccine composed of pMASIA-tgD complexed to 0.1875 nmol aBNBD3. In this group, humoral responses were improved, as evidenced by increased virus neutralization, tgD-specific early IgG1, and later IgG2a titers, while the strong cell-mediated immune responses, measured based on specific gamma interferon (IFN-γ)-secreting cells, were maintained relative to pMASIA-tgD. Modulation of the immune response might have been due in part to the effect of BNBD3 on dendritic cells (DCs). In vitro studies showed that murine bone marrow-derived DCs (BMDCs) pretreated with aBNBD3 were activated, as evidenced by CD11c downregulation, and were functionally mature, as shown by increased allostimulatory ability. Native, synthetic, and analog forms of BNBD3 were equally capable of inducing functional maturation of BMDCs.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Herpesvirus/imunologia , Vacinas de DNA/imunologia , Proteínas Virais/imunologia , beta-Defensinas/genética , Adjuvantes Imunológicos/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Herpesvirus/administração & dosagem , Vacinas contra Herpesvirus/genética , Imunoglobulina G/sangue , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Virais/genética
11.
Mol Biotechnol ; 57(1): 58-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25173687

RESUMO

One of the impediments in the development of safe and cost effective vaccines for veterinary use has been the availability of appropriate delivery vehicle. We have chosen to develop and use bovine adenovirus (BAdV)-3 as vaccine delivery vector in cattle. Here, we describe the construction of recombinant E3 deleted BAdV-3 vectors expressing single vaccine antigen (BAV360; bovine respiratory syncytial virus G) or two vaccine antigens (BAV851; bovine herpesvirus-1gDt and bovine respiratory syncytial virus G). Recombinant proteins expressed by BAV360 or BAV851 were recognized by protein-specific monoclonal antibodies. Moreover, intranasal immunization of cotton rats with BAV360 (expressing a single vaccine antigen) or BAV851 (expressing two vaccine antigens) induced strong antigen-specific immune responses. These results suggest that single replication-competent BAdV-3 expressing vaccine antigens of two economically important respiratory pathogens of calves has potential to act as a feasible approach in the development of economically effective veterinary vaccines for cattle.


Assuntos
Herpesvirus Bovino 1/metabolismo , Imunidade , Proteínas Recombinantes/metabolismo , Vírus Sincicial Respiratório Bovino/metabolismo , Sigmodontinae/imunologia , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/metabolismo , Animais , Formação de Anticorpos/imunologia , Bovinos , Linhagem Celular , Vacinas Virais/imunologia
12.
PLoS One ; 9(10): e109778, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333720

RESUMO

Innate immunity represents the first line of defense against invading pathogens in the respiratory tract. Innate immune cells such as monocytes, macrophages, dendritic cells, NK cells, and granulocytes contain specific pathogen-recognition molecules which induce the production of cytokines and subsequently activate the adaptive immune response. c-di-GMP is a ubiquitous second messenger that stimulates innate immunity and regulates biofilm formation, motility and virulence in a diverse range of bacterial species with potent immunomodulatory properties. In the present study, c-di-GMP was used to enhance the innate immune response against pertussis, a respiratory infection mainly caused by Bordetella pertussis. Intranasal treatment with c-di-GMP resulted in the induction of robust innate immune responses to infection with B. pertussis characterized by enhanced recruitment of neutrophils, macrophages, natural killer cells and dendritic cells. The immune responses were associated with an earlier and more vigorous expression of Th1-type cytokines, as well as an increase in the induction of nitric oxide in the lungs of treated animals, resulting in significant reduction of bacterial numbers in the lungs of infected mice. These results demonstrate that c-di-GMP is a potent innate immune stimulatory molecule that can be used to enhance protection against bacterial respiratory infections. In addition, our data suggest that priming of the innate immune system by c-di-GMP could further skew the immune response towards a Th1 type phenotype during subsequent infection. Thus, our data suggest that c-di-GMP might be useful as an adjuvant for the next generation of acellular pertussis vaccine to mount a more protective Th1 phenotype immune response, and also in other systems where a Th1 type immune response is required.


Assuntos
GMP Cíclico/análogos & derivados , Imunidade Inata/efeitos dos fármacos , Coqueluche/tratamento farmacológico , Animais , Bordetella pertussis , GMP Cíclico/farmacologia , GMP Cíclico/uso terapêutico , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/imunologia , Coqueluche/imunologia
13.
Vet Immunol Immunopathol ; 160(3-4): 209-17, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25012000

RESUMO

The process of virus replication in host cells is greatly influenced by the set of cytokines, chemokines and antiviral substances activated as a result of host-virus interaction. Alteration of cytokines profiles through manipulation of the innate immune system by innate immune stimulants may be helpful in inhibiting virus replication in otherwise permissive cells. The aim of present studies was to characterize innate immune responses capable of inhibiting infectious bronchitis virus (IBV) replication in chicken lungs after in ovo administration of CpG ODN. In our experiments, CpG ODN 2007 or PBS solution was injected on 18th embryonic day (ED) via the chorioallontoic route. CpG ODN and PBS inoculated embryos were challenged with virulent IBV on the 19th ED. Lung tissue samples from experimental chicks were analysed for cytokines/chemokines gene expression at 24h, 48h, and 72h, post infection. Our data showed significant differential up-regulation of IFN-γ, IL-8 (CXCLi2) and MIP-1ß genes and suppression of IL-6 gene expression being associated with inhibition of IBV replication in lungs tissue retrieved from embryos pre-treated with CpG ODN. It is expected that understanding of the innate immune modulation of target tissues by the virus and innate immune stimulants will be helpful in identification of valuable targets for development of novel, safe, effective and economical control strategies against IBV infection in chickens.


Assuntos
Quimiocinas/genética , Galinhas/imunologia , Galinhas/virologia , Citocinas/genética , Vírus da Bronquite Infecciosa/imunologia , Animais , Proteínas Aviárias/genética , Quimiocina CCL4/genética , Embrião de Galinha , Galinhas/genética , Ilhas de CpG , Expressão Gênica , Genes Virais , Imunidade Inata/genética , Vírus da Bronquite Infecciosa/genética , Vírus da Bronquite Infecciosa/fisiologia , Interferon gama/genética , Interleucina-8/genética , Pulmão/imunologia , Pulmão/virologia , Proteínas do Nucleocapsídeo/genética , Oligodesoxirribonucleotídeos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Replicação Viral/genética , Replicação Viral/imunologia
14.
Vaccine ; 32(26): 3300-6, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24731813

RESUMO

Previous studies have suggested an important role of the cytokine adjuvant IL-6 in the induction of mucosal immune responses in animals, including mice. Here, we report the in vivo ability of bovine adenovirus (BAdV)-3 expressing bovine (Bo) IL-6, to influence the systemic and mucosal immune responses against bovine herpesvirus (BHV)-1 gDt in calves. To co-express both antigen and cytokine, we first constructed a recombinant BAdV-3 expressing chimeric gDt.BoIL-6 protein (BAV326). Secondly, we constructed another recombinant BAdV-3 simultaneously expressing gDt and BoIL-6 using IRES containing a bicistronic cassette gDt-IRES.IL-6, (BAV327). Recombinant proteins expressed by BAV326 and BAV327 retained antigenicity (gDt) and biological activity (BoIL-6). Intranasal immunization of calves with recombinant BAV326, BAV327 or BAV308 (gDt alone) resulted in demonstrable levels of gDt-specific IgG responses in sera and IgA response in nasal secretions, in all animals. In addition, all calves developed complement-independent neutralizing antibody responses against BHV-1. However, no significant difference could be observed in the induction of systemic or mucosal immune response in animals immunized with recombinant BAV326 or BAV327 co-expressing BoIL-6. Moreover, there was no difference in the protection against BHV-1 challenge particularly in the amount of virus excretion in the nasal cavity in calves immunized with BAV326, BAV327 or BAV308. These data suggest that the BoIL-6 had no modulating effect on the induction of gDt specific mucosal and systemic immune responses in calves.


Assuntos
Doenças dos Bovinos/prevenção & controle , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 1 , Imunidade nas Mucosas , Interleucina-6/imunologia , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Adenoviridae , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Formação de Anticorpos , Bovinos , Doenças dos Bovinos/virologia , Vetores Genéticos , Infecções por Herpesviridae/prevenção & controle , Imunoglobulina A Secretora/imunologia , Imunoglobulina G/sangue , Testes de Neutralização , Proteínas Recombinantes/imunologia
15.
Vaccine ; 32(21): 2420-7, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24662707

RESUMO

Poly[di(sodiumcarboxylatoethylphenoxy)phosphazene] (PCEP) has shown great potential as a vaccine adjuvant, but the mechanisms that mediate its adjuvant activity have not been investigated. Previously, we had reported the potential of PCEP to induce cytokines and chemokines at the site of injection. Hence, we hypothesized that PCEP creates strong immuno-competent environment leading to recruitment of immune cells at the injection site. Intramuscular injection of mice with PCEP induced significant recruitment of neutrophils, macrophages, monocytes, dendritic cells (DCs), and lymphocytes at the site of injection as well as in the draining lymph nodes. Flow cytometric analysis showed that the majority of the recruited immune cells took up and/or were associated with PCEP at the injection site, with lymphocytes taking up PCEP in lesser quantity. Further, confocal analysis revealed intracytoplasmic lysosomal localization of PCEP in recruited immune cells. These observations suggest that recruitment of distinct immune cells to the site of injection site may be an important mechanism by which PCEP potentiates immune responses to antigens.


Assuntos
Adjuvantes Imunológicos/farmacologia , Linfonodos/citologia , Linfócitos/imunologia , Músculo Esquelético/citologia , Células Mieloides/imunologia , Animais , Linfócitos B/imunologia , Células Dendríticas/imunologia , Feminino , Injeções Intramusculares , Linfonodos/imunologia , Macrófagos/imunologia , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Músculo Esquelético/imunologia , Neutrófilos/imunologia , Fenilpropionatos/farmacologia , Polímeros/farmacologia , Linfócitos T/imunologia
16.
PLoS One ; 9(1): e87145, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498032

RESUMO

Peste des petits ruminants (PPR) is a viral disease which primarily affects small ruminants, causing significant economic losses for the livestock industry in developing countries. It is endemic in Saharan and sub-Saharan Africa, the Middle East and the Indian sub-continent. The primary hosts for peste des petits ruminants virus (PPRV) are goats and sheep; however recent models studying the pathology, disease progression and viremia of PPRV have focused primarily on goat models. This study evaluates the tissue tropism and pathogenesis of PPR following experimental infection of sheep and goats using a quantitative time-course study. Upon infection with a virulent strain of PPRV, both sheep and goats developed clinical signs and lesions typical of PPR, although sheep displayed milder clinical disease compared to goats. Tissue tropism of PPRV was evaluated by real-time RT-PCR and immunohistochemistry. Lymph nodes, lymphoid tissue and digestive tract organs were the predominant sites of virus replication. The results presented in this study provide models for the comparative evaluation of PPRV pathogenesis and tissue tropism in both sheep and goats. These models are suitable for the establishment of experimental parameters necessary for the evaluation of vaccines, as well as further studies into PPRV-host interactions.


Assuntos
Peste dos Pequenos Ruminantes/patologia , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/isolamento & purificação , Tropismo/genética , Animais , Progressão da Doença , Doenças das Cabras/patologia , Doenças das Cabras/virologia , Cabras/virologia , Peste dos Pequenos Ruminantes/veterinária , Ovinos/virologia , Doenças dos Ovinos/patologia , Doenças dos Ovinos/virologia , Replicação Viral/genética
17.
Clin Vaccine Immunol ; 21(4): 463-77, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24451331

RESUMO

Bovine herpesvirus 1 (BoHV-1) causes recurrent respiratory and genital infections in cattle and predisposes them to lethal secondary infections. While modified live and killed BoHV-1 vaccines exist, these are not without problems. Development of an effective DNA vaccine for BoHV-1 has the potential to address these issues. As a strategy to enhance DNA vaccine immunity, a plasmid encoding the bovine neutrophil beta-defensin 3 (BNBD3) as a fusion with truncated glycoprotein D (tgD) and a mix of two plasmids encoding BNBD3 and tgD were tested in mice and cattle. In mice, coadministration of BNBD3 on the separate plasmid enhanced the tgD-induced gamma interferon (IFN-γ) response but not the antibody response. BNBD3 fused to tgD did not affect the antibody levels or the number of IFN-γ-secreting cells but increased the induction of tgD-specific cytotoxic T lymphocytes (CTLs). In cattle, the addition of BNBD3 as a fusion construct also modified the immune response. While the IgG and virus-neutralizing antibody levels were not affected, the number of IFN-γ-secreting cells was increased after BoHV-1 challenge, specifically the CD8(+) IFN-γ(+) T cells, including CD8(+) IFN-γ(+) CD25(+) CTLs. While reduced virus shedding, rectal temperature, and weight loss were observed, the level of protection was comparable to that observed in pMASIA-tgD-vaccinated animals. These data show that coadministration of BNBD3 with a protective antigen as a fusion in a DNA vaccine strengthened the Th1 bias and increased cell-mediated immune responses but did not enhance protection from BoHV-1 infection.


Assuntos
Adjuvantes Imunológicos/farmacologia , Doenças dos Bovinos/prevenção & controle , Infecções por Herpesviridae/veterinária , Vacinas de DNA/imunologia , Proteínas Virais/imunologia , beta-Defensinas/farmacologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Temperatura Corporal , Peso Corporal , Bovinos , Infecções por Herpesviridae/prevenção & controle , Imunoglobulina G/sangue , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Subpopulações de Linfócitos T/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Virais/genética , Eliminação de Partículas Virais , beta-Defensinas/genética
18.
Vaccines (Basel) ; 2(3): 500-14, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26344742

RESUMO

The potent adjuvant activity of the novel adjuvant, poly[di(sodiumcarboxylatoethylphenoxy)phosphazene] (PCEP), with various antigens has been reported previously. However, very little is known about its mechanisms of action. We have recently reported that intramuscular injection of PCEP induces NLRP3, an inflammasome receptor gene, and inflammatory cytokines, including IL-1ß and IL-18, in mouse muscle tissue. Caspase-1 is required for the processing of pro-forms of IL-1ß and IL-18 into mature forms and is a critical constituent of the NLRP3 inflammasome. Hence, in the present study, we investigated the role of caspase-1 in the secretion of IL-1ß and IL-18 in PCEP-stimulated splenic dendritic cells (DCs). Caspase inhibitor YVAD-fmk-treated splenic DCs showed significantly reduced IL-1ß and IL-18 secretion in response to PCEP stimulation. Further, PCEP had no effect on the expression of MHC class II or co-stimulatory molecules, CD86 and CD40, suggesting that PCEP does not induce DC maturation. However, PCEP directly activated B-cells to induce significant production of IgM. In addition, PCEP+ovalbumin (OVA) immunized mice showed significantly increased production of antigen-specific IFN-γ by CD4⁺ and CD8⁺ T-cells. We conclude that PCEP activates innate immunity, leading to increased antigen-specific T-cell responses.

19.
Clin Invest Med ; 37(6): E409-13, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25618274

RESUMO

Infectious diseases continue to cause significant morbidity and mortality in both animals and humans. Indeed, every year infectious diseases cost the global economy billions of dollars in losses and are responsible for approximately one-third of all human deaths. These deaths occur from routine infections, hospital acquired infections (approximately 100,000 deaths occur annually in North America due to hospital-acquired infections), occasional pandemics or regional outbreaks. The most recent regional outbreak is Ebola in West Africa. This infection has caused significant challenges for the regional health care community and has had a global impact. The challenge in the control of infectious diseases is not only due to routine infections but also to the continued emergence and re-emergence of infectious diseases. These new threats occur on a regular basis with approximately thirty new emerging or re-emerging diseases recorded in the last thirty years. The majority of these emerging diseases are zoonotic (over 70%) causing even greater challenges to their control in humans and animals.


Assuntos
Doenças Transmissíveis Emergentes/prevenção & controle , Surtos de Doenças , Doença pelo Vírus Ebola/prevenção & controle , Doença Iatrogênica/prevenção & controle , Controle de Infecções/métodos , Vacinação , África Ocidental/epidemiologia , Animais , Doenças Transmissíveis Emergentes/mortalidade , Doença pelo Vírus Ebola/mortalidade , Humanos , Doença Iatrogênica/epidemiologia , América do Norte/epidemiologia , Zoonoses/mortalidade , Zoonoses/prevenção & controle
20.
Vaccine ; 31(31): 3148-55, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23684829

RESUMO

Whooping cough is a respiratory illness most severe in infants and young children. While the introduction of whole-cell (wP) and acellular pertussis (aP) vaccines has greatly reduced the burden of the disease, pertussis remains a problem in neonates and adolescents. New vaccines are needed that can provide early life and long-lasting protection of infants. Vaccination at an early age, however, is problematic due to the interference with maternally derived antibodies (MatAbs) and the bias towards Th2-type responses following vaccination. Here we report the development of a novel vaccine formulation against pertussis that is highly protective in the presence of MatAbs. We co-formulated pertussis toxoid (PTd) and filamentous hemagglutinin (FHA) with cytosine-phosphate-guanosine oligodeoxynucleotides (CpG ODN), cationic innate defense regulator (IDR) peptide and polyphosphazene (PP) into microparticle and soluble vaccine formulations and tested them in murine and porcine models in the presence and absence of passive immunity. Vaccines composed of the new adjuvant formulations induced an earlier onset of immunity, higher anti-pertussis IgG2a and IgA titers, and a balanced Th1/Th2-type responses when compared to immunization with Quadracel(®), one of the commercially available vaccines for pertussis. Most importantly, the vaccines offered protection against challenge infection in the presence of passively transferred MatAbs.


Assuntos
Adesinas Bacterianas/imunologia , Anticorpos Antibacterianos/imunologia , Imunidade Materno-Adquirida , Vacina contra Coqueluche/imunologia , Toxoides/imunologia , Fatores de Virulência de Bordetella/imunologia , Coqueluche/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antibacterianos/sangue , Carga Bacteriana , Feminino , Hemaglutininas/imunologia , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Oligodesoxirribonucleotídeos , Vacina contra Coqueluche/uso terapêutico , Suínos , Equilíbrio Th1-Th2 , Coqueluche/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA