Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 81(24): 13412-23, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913817

RESUMO

A novel genetic vaccine that is based on a Venezuelan equine encephalitis virus (VEE) replicon launched from plasmid DNA is described. The plasmid encodes a VEE replicon under the transcriptional control of the cytomegalovirus immediate-early promoter (VEE DNA). The VEE DNA consistently expressed 3- to 15-fold more green fluorescent protein in vitro than did a conventional DNA vaccine. Furthermore, transfection with the DNA-launched VEE replicon induced apoptosis and type I interferon production. Inoculation of mice with VEE DNA encoding human immunodeficiency virus type 1 gp160 significantly increased humoral responses by several orders of magnitude compared to an equal dose of a conventional DNA vaccine. These increases were also observed at 10- and 100-fold-lower doses of the VEE DNA. Cellular immune responses measured by gamma interferon and interleukin 2 enzyme-linked immunospot assay were significantly higher in mice immunized with the VEE DNA at decreased doses. The immune responses induced by the VEE DNA-encoded antigen, however, were independent of an intact type I interferon signaling pathway. Moreover, the DNA-launched VEE replicon induced an efficient prime to a VEE replicon particle (VRP) boost, increasing humoral and cellular immunity by at least 1 order of magnitude compared to VEE DNA only. Importantly, immunization with VEE DNA, as opposed to VRP, did not induce any anti-VRP neutralizing antibodies. Increased potency of DNA vaccines and reduced vector immunity may ultimately have an impact on the design of vaccination strategies in humans.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Encefalite Equina Venezuelana/genética , Vírus da Encefalite Equina Venezuelana/imunologia , Interferon Tipo I/biossíntese , Interleucina-2/biossíntese , Plasmídeos/genética , Replicon/imunologia , Vacinas de DNA/imunologia , Animais , Apoptose , Linhagem Celular , Chlorocebus aethiops , Vetores Genéticos , Proteína gp160 do Envelope de HIV/genética , Proteína gp160 do Envelope de HIV/imunologia , HIV-1/genética , Humanos , Imunização , Imunoglobulina G/sangue , Células L , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células NIH 3T3 , Regiões Promotoras Genéticas , Replicon/genética , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Células Vero
2.
Virology ; 390(2): 157-62, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19570561

RESUMO

For some patients infection with Crimean Congo hemorrhagic fever virus (CCHFV) causes a severe disease characterized by fever, vascular leakage and coagulopathy. Knowledge of CCHF pathogenesis is limited and today there is no information about the specific target cells of CCHFV. In this study we analyzed the permissiveness of human peripheral blood mononuclear cells (PBMCs) including monocyte-derived dendritic cells (moDCs) to CCHFV infection. Interestingly, we found that moDCs are the most permissive to CCHFV infection and this infection induced cytokine release from moDCs. Furthermore, supernatants from infected moDCs were found to activate human endothelial cells.


Assuntos
Células Dendríticas/virologia , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Doadores de Sangue , Células Cultivadas , Citocinas/metabolismo , Células Endoteliais/imunologia , Humanos , Leucócitos Mononucleares/virologia
3.
Virology ; 392(1): 103-13, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19631955

RESUMO

Dengue viruses (DENV) are the etiological agents of dengue fever (DF) and dengue hemorrhagic fever (DHF). The DENV complex consists of four closely related viruses designated DENV serotypes 1 through 4. Although infection with one serotype induces cross reactive antibody to all 4 serotypes, the long-term protective antibody response is restricted to the serotype responsible for infection. Cross reactive antibodies appear to enhance infection during a second infection with a different serotype. The goal of the present study was to characterize the binding specificity and functional properties of human DENV immune sera. The study focused on domain III of the viral envelope protein (EDIII), as this region has a well characterized epitope that is recognized by strongly neutralizing serotype-specific mouse monoclonal antibodies (Mabs). Our results demonstrate that EDIII-reactive antibodies are present in primary and secondary DENV immune human sera. Human antibodies bound to a serotype specific epitope on EDIII after primary infection and a serotype cross reactive epitope on EDIII after secondary infection. However, EDIII binding antibodies constituted only a small fraction of the total antibody in immune sera binding to DENV. Studies with complete and EDIII antibody depleted human immune sera demonstrated that EDIII binding antibodies play a minor role in DENV neutralization. We propose that human antibodies directed to other epitopes on the virus are primarily responsible for DENV neutralization. Our results have implications for understanding protective immunity following natural DENV infection and for evaluating DENV vaccines.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Dengue/imunologia , Animais , Antígenos Virais/química , Antígenos Virais/genética , Vírus da Dengue/classificação , Vírus da Dengue/genética , Humanos , Técnicas In Vitro , Camundongos , Testes de Neutralização , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
4.
J Clin Microbiol ; 45(11): 3777-80, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17804661

RESUMO

As dengue vaccines enter clinical trials, there is a need for rapid and quantitative assays to measure neutralization. We have developed flow-based neutralization assays which generated results similar to those generated by the established, plaque reduction neutralization test. The flow assays are an improvement, as they use human cells and allow for high-throughput screening.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Citometria de Fluxo/métodos , Testes de Neutralização/métodos , Humanos , Soros Imunes/imunologia , Ensaio de Placa Viral
5.
Intervirology ; 48(4): 255-61, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15920350

RESUMO

OBJECTIVES: The potential risk of accidental infection by hantaviruses in a clinical or research laboratory necessitates special precautionary measures. A biosafety program must address handling and disposal of infectious materials as well as appropriate virus inactivation or depletion procedures to permit necessary further processing of specimens outside the biosafety level 3 laboratory. METHODS: To study the elimination of hantavirus infectivity, the effects of different chemical and physical inactivation and depletion procedures were investigated on Hantaan virus-containing materials. An infectivity assay for hantaviruses was utilised to verify these procedures which are commonly preceding investigations such as ELISA, flow cytometry analysis, Western blot or immunofluorescence assay. RESULTS: Chemical inactivation with methanol, paraformaldehyde, acetone/methanol and detergent-containing lysis buffer as well as physical forces such as UV irradiation and filtration efficiently reduced viral infectivity in infected cells and their supernatants below the detection limit. CONCLUSION: The virus inactivation and depletion methods described herein are suitable to prepare non-infectious samples for further use in immunological, virological and cell-biological assays.


Assuntos
Vírus Hantaan/efeitos dos fármacos , Vírus Hantaan/efeitos da radiação , Inativação de Vírus/efeitos dos fármacos , Inativação de Vírus/efeitos da radiação , Animais , Chlorocebus aethiops , Contenção de Riscos Biológicos , Desinfetantes/química , Desinfetantes/farmacologia , Desinfecção/métodos , Filtração/métodos , Vírus Hantaan/patogenicidade , Febre Hemorrágica com Síndrome Renal/prevenção & controle , Febre Hemorrágica com Síndrome Renal/transmissão , Humanos , Raios Ultravioleta , Células Vero
6.
J Immunol ; 173(5): 3383-91, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15322202

RESUMO

Human dendritic cells (DCs) are essential for the antiviral immune response and represent a strategically important target for immune evasion of viruses, including human CMV (HCMV). Recently, HCMV has been discovered to encode a unique IL-10 homologue (cmvIL-10). In this study we investigated the capacity of cmvIL-10 to shape phenotype, function, and survival of DCs. For comparison we included human IL-10 and another IL-10 homologue encoded by EBV, which does not directly target DCs. Interestingly, cmvIL-10 strongly activated STAT3 in immature DCs despite its low sequence identity with human IL-10. For most molecules cmvIL-10 blocked LPS-induced surface up-regulation, confirming its role as an inhibitor of maturation. However, a small number of molecules on LPS-treated DCs including IDO, a proposed tolerogenic molecule, showed a different behavior and were up-regulated in response to cmvIL-10. Intriguingly, the expression of C-type lectin DC-specific ICAM-grabbing nonintegrin, a receptor for HCMV infection found exclusively on DCs, was also enhanced by cmvIL-10. This phenotypic change was mirrored by the efficiency of HCMV infection. Moreover, DCs stimulated with LPS and simultaneously treated with cmvIL-10 retained the function of immature DCs. Finally, cmvIL-10 increased apoptosis associated with DC maturation by blocking up-regulation of the antiapoptotic long form cellular FLIP. Taken together, these findings show potential mechanisms by which cmvIL-10 could assist HCMV to infect DCs and to impair DC function and survival.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Células Dendríticas/metabolismo , Interleucina-10/metabolismo , Apoptose/imunologia , Apoptose/fisiologia , Moléculas de Adesão Celular/metabolismo , Citomegalovirus/genética , Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/virologia , Humanos , Interleucina-10/genética , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/metabolismo , Fator de Transcrição STAT3 , Transativadores/metabolismo
7.
Virology ; 308(1): 64-73, 2003 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-12706090

RESUMO

Old world hantaviruses, causing hemorrhagic fever with renal syndrome (HFRS), still present a public health problem in Asia and Eastern Europe. The majority of cases has been recorded in China. The aim of our study was to generate human recombinant neutralizing antibodies to a hantavirus by phage display technology. To preserve the structural identity of viral protein, the panning procedure was performed on native Hantaan (HTN) (76-118) virus propagated in Vero-E6 cells. In total, five complete human recombinant IgG antibodies were produced in a baculovirus expression system. All of them were able to completely neutralize HTN, and Seoul (SEO) virus in a plaque reduction neutralization test (PRNT). Three of these antibodies could also completely neutralize Dobrava (DOB) virus but not Puumala (PUU) virus. All antibodies bind to Hantaan virus G2 protein localized in the virus envelope. The sequence areas within the HTN (76-118)-G2 protein detected by five selected antibodies were mapped using peptide scans. Two partial epitopes, 916-KVMATIDSF-924 and 954-LVTKDIDFD-963, were recognized, which presumably are of paramount importance for docking of the virus to host cell receptors. A consensus motif 916-KVXATIXSF-924 could be identified by mutational analysis. The neutralizing antibodies to the most widely distributed hantaviruses causing HFRS might be promising candidates for the development of an agent for prevention and treatment of HFRS in patients.


Assuntos
Anticorpos Antivirais/imunologia , Vírus Hantaan/imunologia , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/genética , Especificidade de Anticorpos , Baculoviridae/metabolismo , Sítios de Ligação de Anticorpos/genética , Chlorocebus aethiops , Sequência Consenso , Mapeamento de Epitopos , Epitopos/genética , Vetores Genéticos , Vírus Hantaan/genética , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Dados de Sequência Molecular , Testes de Neutralização , Biblioteca de Peptídeos , Proteínas Recombinantes/imunologia , Alinhamento de Sequência , Células Vero , Proteínas do Envelope Viral/genética
8.
J Virol ; 76(21): 10724-33, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12368315

RESUMO

Dendritic cells (DCs) play a pivotal role as antigen-presenting cells in the antiviral immune response. Here we show that Hantaan virus (HTNV), which belongs to the Bunyaviridae family (genus Hantavirus) and causes hemorrhagic fever with renal syndrome, productively infects human DCs in vitro. In the course of HTNV infection, DCs did not show any cytopathic effect and viral replication did not induce cell lysis or apoptosis. Furthermore, HTNV did not affect apoptosis-inducing signals that are important for the homeostatic control of mature DCs. In contrast to immunosuppressive viruses, e.g., human cytomegalovirus, HTNV activated immature DCs, resulting in upregulation of major histocompatibility complex (MHC), costimulatory, and adhesion molecules. Intriguingly, strong upregulation of MHC class I molecules and an increased intercellular cell adhesion molecule type 1 expression was also detected on HTNV-infected endothelial cells. In addition, antigen uptake by HTNV-infected DCs was reduced, another characteristic feature of DC maturation. Consistent with these findings, we observed that HTNV-infected DCs stimulated T cells as efficiently as did mature DCs. Finally, infection of DCs with HTNV induced the release of the proinflammatory cytokines tumor necrosis factor alpha and alpha interferon. Taken together, our findings indicate that hantavirus-infected DCs may significantly contribute to hantavirus-associated pathogenesis.


Assuntos
Células Dendríticas/imunologia , Vírus Hantaan/imunologia , Animais , Antígenos CD/biossíntese , Antígeno B7-1/biossíntese , Antígeno B7-2 , Antígenos CD40/biossíntese , Linhagem Celular , Chlorocebus aethiops , Células Dendríticas/virologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Interferon gama/metabolismo , Interleucina-12/metabolismo , Glicoproteínas de Membrana/biossíntese , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Células Vero
9.
J Virol ; 78(12): 6143-50, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15163707

RESUMO

Hantaviruses represent important human pathogens and can induce hemorrhagic fever with renal syndrome (HFRS), which is characterized by endothelial dysfunction. Both pathogenic and nonpathogenic hantaviruses replicate without causing any apparent cytopathic effect, suggesting that immunopathological mechanisms play an important role in pathogenesis. We compared the antiviral responses triggered by Hantaan virus (HTNV), a pathogenic hantavirus associated with HFRS, and Tula virus (TULV), a rather nonpathogenic hantavirus, in human umbilical vein endothelial cells (HUVECs). Both HTNV- and TULV-infected cells showed increased levels of molecules involved in antigen presentation. However, TULV-infected HUVECs upregulated HLA class I molecules more rapidly. Interestingly, HTNV clearly induced the production of beta interferon (IFN-beta), whereas expression of this cytokine was barely detectable in the supernatant or in extracts from TULV-infected HUVECs. Nevertheless, the upregulation of HLA class I on both TULV- and HTNV-infected cells could be blocked by neutralizing anti-IFN-beta antibodies. Most strikingly, the antiviral MxA protein, which interferes with hantavirus replication, was already induced 16 h after infection with TULV. In contrast, HTNV-infected HUVECs showed no expression of MxA until 48 h postinfection. In accordance with the kinetics of MxA expression, TULV replicated only inefficiently in HUVECs, whereas HTNV-infected cells produced high titers of virus particles that decreased after 48 h postinfection. Both hantavirus species, however, could replicate equally well in Vero E6 cells, which lack an IFN-induced MxA response. Thus, delayed induction of antiviral MxA in endothelial cells after infection with HTNV could allow viral dissemination and contribute to the pathogenesis leading to HFRS.


Assuntos
Células Endoteliais/imunologia , Células Endoteliais/virologia , Vírus Hantaan/patogenicidade , Orthohantavírus/patogenicidade , Animais , Chlorocebus aethiops , Proteínas de Ligação ao GTP/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon beta/metabolismo , Proteínas de Resistência a Myxovirus , Veias Umbilicais , Células Vero , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa