Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Virol ; 88(4): 2056-70, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24307589

RESUMO

Infectious clone technologies allow the rational design of live attenuated viral vaccines with the possibility of vaccine-driven coexpression of immunomodulatory molecules for additional vaccine safety and efficacy. The latter could lead to novel strategies for vaccine protection against infectious diseases where traditional approaches have failed. Here we show for the flavivirus Murray Valley encephalitis virus (MVEV) that incorporation of the internal ribosome entry site (IRES) of Encephalomyocarditis virus between the capsid and prM genes strongly attenuated virulence and that the resulting bicistronic virus was both genetically stable and potently immunogenic. Furthermore, the novel bicistronic genome organization facilitated the generation of a recombinant virus carrying an beta interferon (IFN-ß) gene. Given the importance of IFNs in limiting virus dissemination and in efficient induction of memory B and T cell antiviral immunity, we hypothesized that coexpression of the cytokine with the live vaccine might further increase virulence attenuation without loss of immunogenicity. We found that bicistronic mouse IFN-ß coexpressing MVEV yielded high virus and IFN titers in cultured cells that do not respond to the coexpressed IFN. However, in IFN response-sufficient cell cultures and mice, the virus produced a self-limiting infection. Nevertheless, the attenuated virus triggered robust innate and adaptive immune responses evidenced by the induced expression of Mx proteins (used as a sensitive biomarker for measuring the type I IFN response) and the generation of neutralizing antibodies, respectively. IMPORTANCE The family Flaviviridae includes a number of important human pathogens, such as Dengue virus, Yellow fever virus, Japanese encephalitis virus, West Nile virus, and Hepatitis C virus. Flaviviruses infect large numbers of individuals on all continents. For example, as many as 100 million people are infected annually with Dengue virus, and 150 million people suffer a chronic infection with Hepatitis C virus. However, protective vaccines against dengue and hepatitis C are still missing, and improved vaccines against other flaviviral diseases are needed. The present study investigated the effects of a redesigned flaviviral genome and the coexpression of an antiviral protein (interferon) on virus replication, pathogenicity, and immunogenicity. Our findings may aid in the rational design of a new class of well-tolerated and safe vaccines.


Assuntos
Clonagem Molecular/métodos , Vírus da Encefalite do Vale de Murray/genética , Vírus da Encefalomiocardite/genética , Imunidade Celular/imunologia , Ribossomos/genética , Vacinas Sintéticas/genética , Vacinas Virais/biossíntese , Animais , Anticorpos Neutralizantes/imunologia , Chlorocebus aethiops , Primers do DNA/genética , Vírus da Encefalite do Vale de Murray/patogenicidade , Engenharia Genética/métodos , Imuno-Histoquímica , Interferon beta/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Resistência a Myxovirus/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vacinas Sintéticas/virologia , Células Vero , Vacinas Virais/genética
2.
Virol J ; 12: 144, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26377679

RESUMO

BACKGROUND: Our understanding of the proteolytic processing events at the NS1-2A junction in the flavivirus polyprotein has not markedly progressed since the early work conducted on dengue virus (DENV). This work identified an octapeptide sequence located immediately upstream of the cleavage site thought to be important in substrate recognition by an as yet unknown, endoplasmic reticulum-resident host protease. Of the eight amino acid recognition sequence, the highly conserved residues at positions P1, P3, P5, P7 and P8 (with respect to N-terminus of NS2A) are particularly sensitive to amino acid substitutions in terms of DENV NS1-NS2A cleavage efficiency; however, the role of the octapeptide in efficient NS1 and NS2A production of other flaviviruses has not been experimentally addressed. METHODS AND RESULTS: Using site-directed mutagenesis at the NS1-2A cleavage site of Murray Valley encephalitis virus (MVEV), we confirmed the dominant role of conserved octapeptide residues for efficient NS1-2A cleavage, while changes at variable and the P1' residues were mostly tolerated. However, digressions from the consensus cleavage motif derived from studies on DENV were also found. Thus, comparison of the impact on cleavage of mutations at the NS1-2A junction of MVEV and DENV showed virus-specific differences at both conserved and variable residues. CONCLUSION: We show, with subgenomic expression and infectious clone-derived mutants of MVEV that conserved residues in the flavivirus octapeptide motif can be replaced with a different amino acid without markedly reducing cleavage efficiency of NS1 and NS2A.


Assuntos
Vírus da Encefalite do Vale de Murray/fisiologia , Poliproteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Virais/metabolismo , Análise Mutacional de DNA , Vírus da Dengue/fisiologia , Vírus da Encefalite do Vale de Murray/genética , Mutagênese Sítio-Dirigida , Poliproteínas/genética , Proteínas Virais/genética
3.
Eur J Immunol ; 43(7): 1789-98, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23568450

RESUMO

Japanese encephalitis, caused by infection with the neurotropic flavivirus, Japanese encephalitis virus (JEV), is among the most important viral encephalitides in Asia. While previous studies established an essential role of Ab and type I IFN, it is still unclear if the cell-mediated immune responses, through their direct antiviral effector functions, contribute to protection against the fatal disease. We report here that mice defective in both the granule exocytosis and death receptor pathways of cytotoxicity display increased susceptibility to JEV. The two cell contact-dependent cytotoxic effector mechanisms act redundantly within the CNS to reduce disease severity. We also demonstrate that IFN-γ is critical in recovery from primary infection with JEV by a mechanism involving suppression of virus growth in the CNS, and that T cells are the main source of the cytokine that promotes viral clearance from the brain. Finally, we show by in vivo depletion of NK cells that this innate immune cell population is dispensable for control of JEV infection in the periphery and in the CNS. Accordingly, cell contact-dependent cytolytic and IFN-γ-dependent noncytolytic clearance of virus mediated by T cells trafficking into the CNS help in recovery from lethal infection in a mouse model of Japanese encephalitis.


Assuntos
Citotoxicidade Imunológica/imunologia , Encefalite Japonesa/imunologia , Interferon gama/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Modelos Animais de Doenças , Imunidade Celular/imunologia , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
J Virol ; 87(8): 4395-402, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23388724

RESUMO

JE-ADVAX is a new, delta inulin-adjuvanted, Japanese encephalitis (JE) candidate vaccine with a strong safety profile and potent immunogenicity that confers efficient immune protection not only against JE virus but also against related neurotropic flaviviruses such as West Nile virus. In this study, we investigated the immunological mechanism of protection by JE-ADVAX vaccine using knockout mice deficient in B cells or CD8(+) T cells and poor persistence of neutralizing antibody or by adoptive transfer of immune splenocyte subpopulations. We show that memory B cells induced by JE-ADVAX provide long-lived protection against JE even in the absence of detectable pre-exposure serum neutralizing antibodies and without the requirement of CD8(+) T cells. Upon virus encounter, these vaccine-induced memory B cells were rapidly triggered to produce neutralizing antibodies that then protected immunized mice from morbidity and mortality. The findings suggest that the extent of the B-cell memory compartment might be a better immunological correlate for clinical efficacy of JE vaccines than the currently recommended measure of serum neutralizing antibody. This may explain the paradox where JE protection is observed in some subjects even in the absence of detectable serum neutralizing antibody. Our investigation also established the suitability of a novel flavivirus challenge model (ß(2)-microglobulin-knockout mice) for studies of the role of B-cell memory responses in vaccine protection.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Encefalite Japonesa/prevenção & controle , Inulina/análogos & derivados , Vacinas contra Encefalite Japonesa/imunologia , Animais , Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Encefalite Japonesa/imunologia , Inulina/administração & dosagem , Vacinas contra Encefalite Japonesa/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
J Virol ; 87(18): 10324-33, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23864620

RESUMO

West Nile virus (WNV), currently the cause of a serious U.S. epidemic, is a mosquito-borne flavivirus and member of the Japanese encephalitis (JE) serocomplex. There is currently no approved human WNV vaccine, and treatment options remain limited, resulting in significant mortality and morbidity from human infection. Given the availability of approved human JE vaccines, this study asked whether the JE-ADVAX vaccine, which contains an inactivated cell culture JE virus antigen formulated with Advax delta inulin adjuvant, could provide heterologous protection against WNV infection in wild-type and ß2-microglobulin-deficient (ß2m(-/-)) murine models. Mice immunized twice or even once with JE-ADVAX were protected against lethal WNV challenge even when mice had low or absent serum cross-neutralizing WNV titers prior to challenge. Similarly, ß2m(-/-) mice immunized with JE-ADVAX were protected against lethal WNV challenge in the absence of CD8(+) T cells and prechallenge WNV antibody titers. Protection against WNV could be adoptively transferred to naive mice by memory B cells from JE-ADVAX-immunized animals. Hence, in addition to increasing serum cross-neutralizing antibody titers, JE-ADVAX induced a memory B-cell population able to provide heterologous protection against WNV challenge. Heterologous protection was reduced when JE vaccine antigen was administered alone without Advax, confirming the importance of the adjuvant to induction of cross-protective immunity. In the absence of an approved human WNV vaccine, JE-ADVAX could provide an alternative approach for control of a major human WNV epidemic.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos B/imunologia , Proteção Cruzada , Inulina/análogos & derivados , Vacinas contra Encefalite Japonesa/imunologia , Febre do Nilo Ocidental/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Animais , Modelos Animais de Doenças , Feminino , Memória Imunológica , Inulina/administração & dosagem , Vacinas contra Encefalite Japonesa/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sobrevida , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Febre do Nilo Ocidental/imunologia
6.
Virol J ; 11: 60, 2014 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-24678844

RESUMO

BACKGROUND: The RGD motif in the mosquito-borne flaviviruses envelope protein domain III (EDIII) FG loop was shown to bind negatively charged cellular molecules and mediate virus entry in mammals. However, its importance in virus entry in the mosquito has not yet been defined. The sequences of RGD motifs are conserved in JEV-serocomplex members primarily transmitted by Culex mosquitoes but absent from members of the DENV serocomplex, which utilize Aedes mosquitoes as vectors. Interestingly, the RGD sequence is present in the attenuated 17D strain of yellow fever virus as a result of the T380R mutation in the EDIII of Asibi strain following extensive in vitro passage in mice and chicken embryos and was found to contribute to the more rapid clearance in mice challenged with 17D. However, viral infectivity and dissemination in mosquitoes had not been evaluated for this mutant. FINDINGS: The study utilized the reverse genetics system of YFV and Ae. aegypti RexD WE mosquitoes to assess the impact of a T380R mutation in YFV Asibi and 17D/Asibi M-E chimera. The T380R mutation led to higher infection rates but similar dissemination rates when introduced into the YFV Asibi strain and 17D/Asibi M-E chimera. CONCLUSIONS: While the increase of the positive charge in EDIII may reduce the virulence of YFV in mice, this mutation favored the establishment of the viral infection in Ae. aegypti. However, such gain in viral infectivity did not increase dissemination in infected mosquitoes.


Assuntos
Mutação de Sentido Incorreto , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Vírus da Febre Amarela/fisiologia , Aedes , Animais , Análise Mutacional de DNA , Camundongos , Mutagênese , Genética Reversa , Vírus da Febre Amarela/genética
7.
J Virol ; 85(11): 5446-55, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21450826

RESUMO

The immunological correlates for recovery from primary Japanese encephalitis virus (JEV) infection in humans and experimental animals remain poorly defined. To investigate the relative importance of the adaptive immune responses, we have established a mouse model for Japanese encephalitis in which a low-dose virus inoculum was administered into the footpads of adult C57BL/6 mice. In this model, ~60% of the mice developed a fatal encephalitis and a virus burden in the central nervous system (CNS). Using mice lacking B cells (µMT(-/-) mice) and immune B cell transfer to wild-type mice, we show a critically important role for humoral immunity in preventing virus spread to the CNS. T cell help played an essential part in the maintenance of an effective antibody response necessary to combat the infection, since mice lacking major histocompatibility complex class II showed truncated IgM and blunted IgG responses and uniformly high lethality. JEV infection resulted in extensive CD8(+) T cell activation, judged by upregulation of surface markers CD69 and CD25 and cytokine production after stimulation with a JEV NS4B protein-derived H-2D(b)-binding peptide and trafficking of virus-immune CD8(+) T cells into the CNS. However, no significant effect of CD8(+) T cells on the survival phenotype was found, which was corroborated in knockout mice lacking key effector molecules (Fas receptor, perforin, or granzymes) of cytolytic pathways triggered by T lymphocytes. Accordingly, CD8(+) T cells are mostly dispensable for recovery from infection with JEV. This finding highlights the conflicting role that CD8(+) T cells play in the pathogenesis of JEV and closely related encephalitic flaviviruses such as West Nile virus.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Encefalite Japonesa/imunologia , Animais , Sistema Nervoso Central/virologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite Japonesa/mortalidade , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças dos Roedores/imunologia , Doenças dos Roedores/mortalidade , Análise de Sobrevida
8.
J Virol ; 85(21): 11170-82, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21849445

RESUMO

Ectromelia virus (ECTV) is a natural pathogen of mice that causes mousepox, and many of its genes have been implicated in the modulation of host immune responses. Serine protease inhibitor 2 (SPI-2) is one of these putative ECTV host response modifier proteins. SPI-2 is conserved across orthopoxviruses, but results defining its mechanism of action and in vivo function are lacking or contradictory. We studied the role of SPI-2 in mousepox by deleting the SPI-2 gene or its serine protease inhibitor reactive site. We found that SPI-2 does not affect viral replication or cell-intrinsic apoptosis pathways, since mutant viruses replicate in vitro as efficiently as wild-type virus. However, in the absence of SPI-2 protein, ECTV is attenuated in mousepox-susceptible mice, resulting in lower viral loads in the liver, decreased spleen pathology, and substantially improved host survival. This attenuation correlates with more effective immune responses in the absence of SPI-2, including an earlier serum gamma interferon (IFN-γ) response, raised serum interleukin 18 (IL-18), increased numbers of granzyme B(+) CD8(+) T cells, and, most notably, increased numbers and activation of NK cells. Both virus attenuation and the improved immune responses associated with SPI-2 deletion from ECTV are lost when mice are depleted of NK cells. Consequently, SPI-2 renders mousepox lethal in susceptible strains by preventing protective NK cell defenses.


Assuntos
Vírus da Ectromelia/patogenicidade , Ectromelia Infecciosa/mortalidade , Interações Hospedeiro-Patógeno , Células Matadoras Naturais/imunologia , Serpinas/metabolismo , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Vírus da Ectromelia/genética , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/virologia , Deleção de Genes , Interferon gama/metabolismo , Interleucina-18/metabolismo , Fígado/virologia , Subpopulações de Linfócitos/química , Subpopulações de Linfócitos/imunologia , Camundongos , Serpinas/genética , Baço/patologia , Análise de Sobrevida , Carga Viral , Proteínas Virais/genética , Replicação Viral
9.
J Gen Virol ; 92(Pt 10): 2286-2296, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21733886

RESUMO

Murray Valley encephalitis virus (MVEV) is a mosquito-borne flavivirus endemic to Australia and Papua New Guinea. Most strains of MVEV cause potentially fatal cases of encephalitis in humans and horses, and have been shown to be highly neuroinvasive in weanling mice. In contrast, the naturally occurring subtype Alfuy virus (ALFV) has never been associated with human disease, nor is it neuroinvasive in weanling mice, even at high doses. To identify viral factors associated with ALFV attenuation, a chimeric infectious clone was constructed containing the structural genes premembrane (prM) and envelope (E) of ALFV swapped into the MVEV genome. The resulting virus (vMVEV/ALFVstr) was no longer neuroinvasive in mice, suggesting that motifs within prM-E of ALFV confer attenuation. To define these motifs further, mutants were constructed by targeting divergent sequences between the MVEV and ALFV E proteins that are known markers of virulence in other encephalitic flaviviruses. MVEV mutants containing a unique ALFV sequence in the flexible hinge region (residues 273-277) or lacking the conserved glycosylation site at position 154 were significantly less neuroinvasive in mice than wild-type MVEV, as determined by delayed time to death or increased LD(50). Conversely, when the corresponding MVEV sequences were inserted into the vMVEV/ALFVstr chimera, the mutant containing the MVEV hinge sequence was more neuroinvasive than the parental chimera, though not to the same level as wild-type MVEV. These results identify the hinge region and E protein glycosylation as motifs that contribute to the attenuation of ALFV.


Assuntos
Flavivirus/genética , Flavivirus/patogenicidade , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Animais , Modelos Animais de Doenças , Encefalite Viral/mortalidade , Encefalite Viral/patologia , Encefalite Viral/virologia , Infecções por Flavivirus/mortalidade , Infecções por Flavivirus/patologia , Infecções por Flavivirus/virologia , Glicosilação , Dose Letal Mediana , Camundongos , Recombinação Genética , Doenças dos Roedores/mortalidade , Doenças dos Roedores/patologia , Doenças dos Roedores/virologia , Análise de Sobrevida , Virulência
10.
J Virol ; 84(10): 5171-80, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20219924

RESUMO

The E protein of most flaviviruses is modified by Asn-linked glycosylation at residue 153/154 and in the case of the four dengue virus (DENV) serotypes by a second glycan at residue 67. However, the absence of E protein glycosylation among numerous natural isolates of different flaviviruses suggests that the glycan, per se, is not critically important in the virus life cycle. Consistent with this notion, we show that ablation of both glycans from the DENV-2 E protein reduces but does not prevent growth of the variant in mammalian and mosquito cells. We found a pronounced and opposing effect of glycan ablation on two stages of the virus growth cycle: infectivity and release. Loss of either of the two DENV E protein glycans markedly enhanced infectivity of variants for mosquito cells at the expense of efficient virion release. The variants also displayed reduced release in mammalian cells, which was more prominent for viruses lacking the Asn 67-linked glycan than for those lacking the Asn 153-linked glycan, without a marked change in infectivity. Mutations, which compensated for the defect in virus morphogenesis associated with ablation of the Asn 67-linked glycan in mammalian cells but interestingly not in mosquito cells, were identified at the glycosylation acceptor motif and a second site in E protein domain II. The dueling influences of infectivity and release on virus growth affected by the glycans may explain the plasticity in E protein glycosylation among the flaviviruses.


Assuntos
Vírus da Dengue/fisiologia , Vírus da Dengue/patogenicidade , Proteínas do Envelope Viral/fisiologia , Fatores de Virulência/fisiologia , Liberação de Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Culicidae , Mutação de Sentido Incorreto , Supressão Genética , Proteínas do Envelope Viral/genética , Virulência
11.
J Virol ; 84(9): 4212-21, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164231

RESUMO

We previously demonstrated that a single dose of nonadjuvanted intranasal gamma-irradiated influenza A virus can provide robust protection in mice against both homologous and heterosubtypic challenges, including challenge with an H5N1 avian virus strain. We investigated the mechanism behind the observed cross-protection to define which arms of the adaptive immune response are involved in mediating this protection. Studies with gene knockout mice showed the cross-protective immunity to be mediated mainly by T cells and to be dependent on the cytolytic effector molecule perforin. Adoptive transfer of memory T cells from immunized mice, but not of memory B cells, protected naïve recipients against lethal heterosubtypic influenza virus challenge. Furthermore, gamma-irradiated influenza viruses induced cross-reactive Tc-cell responses but not cross-neutralizing or cross-protective antibodies. In addition, histological analysis showed reduced lung inflammation in vaccinated mice compared to that in unvaccinated controls following heterosubtypic challenge. This reduced inflammation was associated with enhanced early recruitment of T cells, both CD4(+) and CD8(+), and with early influenza virus-specific cytotoxic T-cell responses. Therefore, cross-protective immunity induced by vaccination with gamma-irradiated influenza A virus is mediated mainly by Tc-cell responses.


Assuntos
Proteção Cruzada , Raios gama , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Linfócitos T Citotóxicos/imunologia , Transferência Adotiva , Animais , Anticorpos Antivirais/sangue , Peso Corporal , Vírus da Influenza A Subtipo H1N1/efeitos da radiação , Vírus da Influenza A Subtipo H3N2/efeitos da radiação , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Análise de Sobrevida , Vacinas de Produtos Inativados/imunologia
12.
J Cell Biol ; 174(4): 509-19, 2006 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-16893972

RESUMO

Aspergillus fumigatus infections cause high levels of morbidity and mortality in immunocompromised patients. Gliotoxin (GT), a secondary metabolite, is cytotoxic for mammalian cells, but the molecular basis and biological relevance of this toxicity remain speculative. We show that GT induces apoptotic cell death by activating the proapoptotic Bcl-2 family member Bak, but not Bax, to elicit the generation of reactive oxygen species, the mitochondrial release of apoptogenic factors, and caspase-3 activation. Activation of Bak by GT is direct, as GT triggers in vitro a dose-dependent release of cytochrome c from purified mitochondria isolated from wild-type and Bax- but not Bak-deficient cells. Resistance to A. fumigatus of mice lacking Bak compared to wild-type mice demonstrates the in vivo relevance of this GT-induced apoptotic pathway involving Bak and suggests a correlation between GT production and virulence. The elucidation of the molecular basis opens new strategies for the development of therapeutic regimens to combat A. fumigatus and related fungal infections.


Assuntos
Apoptose/fisiologia , Aspergilose/metabolismo , Aspergillus fumigatus/metabolismo , Gliotoxina/toxicidade , Imunidade Inata/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Aspergilose/imunologia , Aspergilose/fisiopatologia , Aspergillus fumigatus/patogenicidade , Caspase 3 , Caspases/metabolismo , Linhagem Celular Transformada , Citocromos c/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fibroblastos/química , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Virulência , Proteína Killer-Antagonista Homóloga a bcl-2/genética
13.
J Gen Virol ; 91(Pt 6): 1450-60, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20147516

RESUMO

We have recently shown that intranasal (i.n.) administration of gamma-irradiated A/PR/8 [A/Puerto Rico/8/34 (H1N1)] protects mice against lethal avian influenza A/Vietnam/1203/2004 (H5N1) and other heterosubtypic influenza A infections. Here, we used gamma-irradiated, formalin- and UV-inactivated A/PC [A/Port Chalmers/1/73 (H3N2)] virus preparations and compared their ability to induce both homologous and heterosubtypic protective immunity. Our data show that, in contrast to i.n. vaccination with formalin- or UV-inactivated virus, or the present commercially available trivalent influenza vaccine, a single dose of gamma-ray-inactivated A/PC (gamma-A/PC) conferred significant protection in mice against both homologous and heterosubtypic virus challenges. A multiple immunization regime was required for formalin-inactivated virus preparations to induce protective immunity against a homotypic virus challenge, but did not induce influenza A strain cross-protective immunity. The highly immunogenic gamma-A/PC, but not formalin- or UV-inactivated A/PC, nor the currently available subvirion vaccine, elicited cytotoxic T-cell responses that are most likely responsible for the cross-protective and long-lasting immunity against highly lethal influenza A infections in mice. Finally, freeze-drying of gamma-A/PC did not affect the ability to induce cross-protective immunity.


Assuntos
Proteção Cruzada , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Inativação de Vírus , Animais , Peso Corporal , Feminino , Formaldeído/toxicidade , Raios gama , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Vírus da Influenza A Subtipo H3N2/efeitos da radiação , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Raios Ultravioleta , Vacinas de Produtos Inativados/imunologia , Carga Viral
14.
J Gen Virol ; 91(Pt 6): 1407-17, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20130134

RESUMO

Advax is a polysaccharide-based adjuvant that potently stimulates vaccine immunogenicity without the increased reactogenicity seen with other adjuvants. This study investigated the immunogenicity of a novel Advax-adjuvanted Vero cell culture candidate vaccine against Japanese encephalitis virus (JEV) in mice and horses. The results showed that, in mice, a two-immunization, low-dose (50 ng JEV antigen) regimen with adjuvanted vaccine produced solid neutralizing immunity comparable to that elicited with live ChimeriVax-JE immunization and superior to that elicited with tenfold higher doses of a traditional non-adjuvanted JEV vaccine (JE-VAX; Biken Institute) or a newly approved alum-adjuvanted vaccine (Jespect; Novartis). Mice vaccinated with the Advax-adjuvanted, but not the unadjuvanted vaccine, were protected against live JEV challenge. Equine immunizations against JEV with Advax-formulated vaccine similarly showed enhanced vaccine immunogenicity, confirming that the adjuvant effects of Advax are not restricted to rodent models. Advax-adjuvanted JEV vaccine elicited a balanced T-helper 1 (Th1)/Th2 immune response against JEV with protective levels of cross-neutralizing antibody against other viruses belonging to the JEV serocomplex, including Murray Valley encephalitis virus (MVEV). The adjuvanted JEV vaccine was well tolerated with minimal reactogenicity and no systemic toxicity in immunized animals. The cessation of manufacture of traditional mouse brain-derived unadjuvanted JEV vaccine in Japan has resulted in a JEV vaccine shortage internationally. There is also an ongoing lack of human vaccines against other JEV serocomplex flaviviruses, such as MVEV, making this adjuvanted, cell culture-grown JEV vaccine a promising candidate to address both needs with one vaccine.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Flavivirus/imunologia , Inulina/análogos & derivados , Vacinas contra Encefalite Japonesa/imunologia , Animais , Chlorocebus aethiops , Reações Cruzadas , Encefalite Japonesa/prevenção & controle , Feminino , Cavalos , Inulina/administração & dosagem , Vacinas contra Encefalite Japonesa/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Análise de Sobrevida , Células Th1/imunologia , Células Th2/imunologia , Vacinação/métodos , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Células Vero
15.
J Virol ; 83(6): 2436-45, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19109382

RESUMO

The Japanese encephalitis virus (JEV) serocomplex, which also includes Murray Valley encephalitis virus (MVEV), is a group of antigenically closely related, mosquito-borne flaviviruses that are responsible for severe encephalitic disease in humans. While vaccines against the prominent members of this serocomplex are available or under development, it is unlikely that they will be produced specifically against those viruses which cause less-frequent disease, such as MVEV. Here we have evaluated the cross-protective values of an inactivated JEV vaccine (JE-VAX) and a live chimeric JEV vaccine (ChimeriVax-JE) against MVEV in two mouse models of flaviviral encephalitis. We show that (i) a three-dose vaccination schedule with JE-VAX provides cross-protective immunity, albeit only partial in the more severe challenge model; (ii) a single dose of ChimeriVax-JE gives complete protection in both challenge models; (iii) the cross-protective immunity elicited with ChimeriVax-JE is durable (>or=5 months) and broad (also giving protection against West Nile virus); (iv) humoral and cellular immunities elicited with ChimeriVax-JE contribute to protection against lethal challenge with MVEV; (v) ChimeriVax-JE remains fully attenuated in immunodeficient mice lacking type I and type II interferon responses; and (vi) immunization with JE-VAX, but not ChimeriVax-JE, can prime heterologous infection enhancement in recipients of vaccination on a low-dose schedule, designed to mimic vaccine failure or waning of vaccine-induced immunity. Our results suggest that the live chimeric JEV vaccine will protect against other viruses belonging to the JEV serocomplex, consistent with the observation of cross-protection following live virus infections.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/imunologia , Vírus da Encefalite do Vale de Murray/imunologia , Encefalite por Arbovirus/prevenção & controle , Animais , Anticorpos Antivirais/sangue , Reações Cruzadas , Feminino , Humanos , Imunização Secundária , Memória Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sobrevida , Fatores de Tempo , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia
16.
J Virol ; 82(12): 6024-33, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400851

RESUMO

The yellow fever virus (YFV) 17D strain is one of the most effective live vaccines for human use, but the in vivo mechanisms for virulence attenuation of the vaccine and the corresponding molecular determinants remain elusive. The vaccine differs phenotypically from wild-type YFV by the loss of viscerotropism, despite replicative fitness in cell culture, and genetically by 20 amino acid changes predominantly located in the envelope (E) protein. We show that three residues in E protein domain III inhibit spread of 17D in extraneural tissues and attenuate virulence in type I/II interferon-deficient mice. One of these residues (Arg380) is a dominant glycosaminoglycan-binding determinant, which mainly accounts for more rapid in vivo clearance of 17D from the bloodstream in comparison to 17D-derived variants with wild-type-like E protein. While other mutations will account for loss of neurotropism and phenotypic stability, the described impact of E protein domain III changes on virus dissemination and virulence is the first rational explanation for the safety of the 17D vaccine in humans.


Assuntos
Glicosaminoglicanos/metabolismo , Proteínas do Envelope Viral/genética , Vacina contra Febre Amarela , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/imunologia , Vírus da Febre Amarela/patogenicidade , Aedes/citologia , Animais , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Camundongos , Camundongos Knockout , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Virulência/genética , Vírus da Febre Amarela/metabolismo
17.
Virology ; 506: 1-6, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28282567

RESUMO

The genetic basis for a dramatically increased virus susceptibility phenotype of MHC-II knockout mice acquired during routine maintenance of the mouse strain was determined. Segregation of the susceptibility allele from the defective MHC-II locus combined with sequence capture and sequencing showed that a Y37L substitution in STAT1 accounted for high flavivirus susceptibility of a newly derived mouse strain, designated Tuara. Interestingly, the mutation in STAT1 gene gave only partial inactivation of the type I interferon antiviral pathway. Accordingly, merely a relatively small impairment of interferon α/ß signalling is sufficient to overcome the ability of the host to control the infection.


Assuntos
Infecções por Flaviviridae/virologia , Flaviviridae/fisiologia , Fator de Transcrição STAT1/imunologia , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Flaviviridae/genética , Infecções por Flaviviridae/genética , Infecções por Flaviviridae/imunologia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT1/química , Fator de Transcrição STAT1/genética
18.
Virus Res ; 228: 61-65, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27865865

RESUMO

Mousepox is caused by the orthopoxvirus ectromelia virus (ECTV), and is thought to be transmitted via skin abrasions. We studied the ECTV virulence factor N1 following subcutaneous infection of mousepox-susceptible BALB/c mice. In this model, ECTV lacking N1L gene was attenuated more than 1000-fold compared with wild-type virus and replication was profoundly reduced as early as four days after infection. However, in contrast to data from an intranasal model, N1 protein was not required for virus dissemination. Further, neither T cell nor cytokine responses were enhanced in the absence of N1. Together with the early timing of reduced virus titres, this suggests that in a cutaneous model, N1 exerts its function at the level of infected cells or in the inhibition of the very earliest effectors of innate immunity.


Assuntos
Vírus da Ectromelia/fisiologia , Ectromelia Infecciosa/virologia , Proteínas Virais/genética , Animais , Interações Hospedeiro-Patógeno , Camundongos , Carga Viral , Proteínas Virais/metabolismo , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Replicação Viral
19.
Antiviral Res ; 69(1): 31-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16309754

RESUMO

Many viruses, including flaviviruses, display affinity for cell surface heparan sulfate (HS) proteoglycans with biological relevance in virus attachment/entry. This raises the possibility of the application of HS mimetics in antiviral therapy. We have evaluated the antiviral effect of the sulfated polysaccharides, suramin, pentosan polysulfate (PPS) and PI-88, which are currently approved or in trial for clinical use, against dengue virus (DEN) and the encephalitic flaviviruses, Japanese encephalitis virus, West Nile virus, and Murray Valley encephalitis virus. A flow cytometry-based method for the measurement of inhibition of virus infectivity was developed, which showed the in vitro antiviral activity of the three compounds, albeit with differences in efficiency which were virus-dependent. The 50% effective concentration (EC(50)) values for DEN inhibition were in the order: PPS

Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Dengue/tratamento farmacológico , Vírus da Encefalite Japonesa (Subgrupo)/efeitos dos fármacos , Heparitina Sulfato/farmacologia , Oligossacarídeos/farmacologia , Animais , Antivirais/química , Antivirais/uso terapêutico , Linhagem Celular , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Encefalite por Arbovirus/tratamento farmacológico , Feminino , Infecções por Flavivirus/tratamento farmacológico , Heparitina Sulfato/uso terapêutico , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligossacarídeos/química , Oligossacarídeos/uso terapêutico , Poliéster Sulfúrico de Pentosana/química , Poliéster Sulfúrico de Pentosana/farmacologia , Poliéster Sulfúrico de Pentosana/uso terapêutico , Suramina/química , Suramina/farmacologia , Suramina/uso terapêutico , Resultado do Tratamento
20.
Expert Rev Vaccines ; 11(2): 177-87, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22309667

RESUMO

Serological cross-reactivity providing cross-protective immunity between antigenically related viruses is a cornerstone of vaccination. It was the immunological basis for the first human vaccine against smallpox introduced more than 200 years ago, and continues to underpin modern vaccine development as has recently been shown for human papillomavirus vaccines, which confer cross-protection against other oncogenic papillomavirus types not present in the vaccine. Here, we review the feasibility of cross-protective vaccination against an antigenic group of clinically important viruses belonging to the Japanese encephalitis serocomplex in the Flaviviridae family. We will discuss evidence suggesting that 'new generation' flavivirus vaccines may provide effective cross-protective immunity against heterologous Japanese encephalitis serocomplex viruses, and appraise potential risks associated with cross-reactive vaccine immunity. The review will also focus on the structural and mechanistic basis for cross-protective immunity among this group of flaviviruses, which is predominantly mediated by antibodies against a single viral surface protein.


Assuntos
Anticorpos Antivirais/imunologia , Proteção Cruzada/imunologia , Vírus da Encefalite Japonesa (Subgrupo)/classificação , Infecções por Flavivirus/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Cricetinae , Vírus da Encefalite Japonesa (Subgrupo)/imunologia , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/virologia , Humanos , Camundongos , Modelos Moleculares , Vacinação , Proteínas do Envelope Viral/química , Vacinas Virais/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA