Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Front Immunol ; 10: 646, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30984200

RESUMO

Adjuvanted whole inactivated virus (WIV) influenza vaccines show promise as broadly protective influenza vaccine candidates. Using WIV as basis we assessed the relative efficacy of different adjuvants by carrying out a head-to-head comparison of the liposome-based adjuvants CAF01 and CAF09 and the protein-based adjuvants CTA1-DD and CTA1-3M2e-DD and evaluated whether one or more of the adjuvants could induce broadly protective immunity. Mice were immunized with WIV prepared from A/Puerto Rico/8/34 (H1N1) virus intramuscularly with or without CAF01 or intranasally with or without CAF09, CTA1-DD, or CTA1-3M2e-DD, followed by challenge with homologous, heterologous or heterosubtypic virus. In general, intranasal immunizations were significantly more effective than intramuscular immunizations in inducing virus-specific serum-IgG, mucosal-IgA, and splenic IFNγ-producing CD4 T cells. Intranasal immunizations with adjuvanted vaccines afforded strong cross-protection with milder clinical symptoms and better control of virus load in lungs. Mechanistic studies indicated that non-neutralizing IgG antibodies and CD4 T cells were responsible for the improved cross-protection while IgA antibodies were dispensable. The role of CD4 T cells was particularly pronounced for CTA1-3M2e-DD adjuvanted vaccine as evidenced by CD4 T cell-dependent reduction of lung virus titers and clinical symptoms. Thus, intranasally administered WIV in combination with effective mucosal adjuvants appears to be a promising broadly protective influenza vaccine candidate.


Assuntos
Adjuvantes Imunológicos , Proteção Cruzada , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza , Infecções por Orthomyxoviridae/prevenção & controle , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/farmacologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Feminino , Imunoglobulina G/imunologia , Vacinas contra Influenza/química , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Vacinas de Produtos Inativados/química , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/farmacologia
2.
PLoS One ; 12(8): e0183496, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28829831

RESUMO

BACKGROUND: Although human T-lymphotropic virus (HTLV) is transmitted via the same routes as human immunodeficiency virus (HIV), its worldwide seroprevalence differs drastically because HTLV is transmitted mainly via infected cells rather than free virus. The sharing of needles and other equipment places people who inject drugs (PWID) at particularly high-risk for such blood-borne diseases. METHODS: To validate the methodology used to process and analyze the dried blood spots (DBS) utilized in the study, dried serum spots (DSS) with dilutions of sera from known HTLV infected individuals were analyzed by ELISA and Western blot. DBS collected between 2011 and 2015 from 2,077 PWID in eight German cities recruited by respondent-driven sampling were tested for HTLV-specific antibodies. RESULTS: The validation demonstrated that the use of DSS allowed identification of samples with even low titers of HTLV-specific antibodies, although a confirmatory Western blot with an additional venous blood sample would often be required. Despite numerous HIV and HCV positive individuals being identified within the study population, none tested positive for HTLV. CONCLUSION: While the HIV and HCV prevalences in German PWID are comparable to those in other European countries, the very low prevalence of HTLV reflects the situation in the general population.


Assuntos
Infecções por Deltaretrovirus/sangue , Abuso de Substâncias por Via Intravenosa/complicações , Anticorpos Antideltaretrovirus/sangue , Infecções por Deltaretrovirus/complicações , Ensaio de Imunoadsorção Enzimática , Alemanha/epidemiologia , Humanos , Estudos Soroepidemiológicos
3.
BMC Infect Dis ; 17(1): 241, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28376743

RESUMO

BACKGROUND: Current influenza vaccines, based on antibodies against surface antigens, are unable to provide protection against newly emerging virus strains which differ from the vaccine strains. Therefore the population has to be re-vaccinated annually. It is thus important to develop vaccines which induce protective immunity to a broad spectrum of influenza viruses. This trial is designed to evaluate the immunogenicity and safety of FLU-v, a vaccine composed of four synthetic peptides with conserved epitopes from influenza A and B strains expected to elicit both cell mediated immunity (CMI) and humoral immunity providing protection against a broad spectrum of influenza viruses. METHODS: In a single-center, randomized, double-blind and placebo-controlled phase IIb trial, 222 healthy volunteers aged 18-60 years will be randomized (2:2:1:1) to receive two injections of a suspension of 500 µg FLU-v in saline (arm 1), one dose of emulsified 500 µg FLU-v in Montanide ISA-51 and water for injection (WFI) followed by one saline dose (arm 2), two saline doses (arm 3), or one dose of Montanide ISA-51 and WFI emulsion followed by one saline dose (arm 4). All injections will be given subcutaneously. Primary endpoints are safety and FLU-v induced CMI, evaluated by cytokine production by antigen specific T cell populations (flow-cytometry and ELISA). Secondary outcomes are measurements of antibody responses (ELISA and multiplex), whereas exploratory outcomes include clinical efficacy and additional CMI assays (ELISpot) to show cross-reactivity. DISCUSSION: Broadly protective influenza vaccines able to provide protection against multiple strains of influenza are urgently needed. FLU-v is a promising vaccine which has shown to trigger the cell-mediated immune response. The dosages and formulations tested in this current trial are also estimated to induce antibody response. Therefore, both cellular and humoral immune responses will be evaluated. TRIAL REGISTRATION: EudraCT number 2015-001932-38 ; retrospectively registered clinicaltrials.gov NCT02962908 (November 7th 2016).


Assuntos
Imunogenicidade da Vacina , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Adolescente , Adulto , Anticorpos Antivirais/imunologia , Método Duplo-Cego , ELISPOT , Epitopos , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Vacinas contra Influenza/administração & dosagem , Masculino , Manitol/análogos & derivados , Pessoa de Meia-Idade , Ácidos Oleicos , Orthomyxoviridae/imunologia , Vacinas Sintéticas/imunologia , Adulto Jovem
4.
Medicine (Baltimore) ; 96(11): e6339, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28296763

RESUMO

INTRODUCTION: Influenza is a major respiratory viral infection of humans with high mortality and morbidity rates and profound economic impact. Although influenza vaccines are generally updated yearly to match the viruses expected in the coming season, genetic mutation and reassortment can result in unexpected novel strains. Therefore, it is important to develop universal vaccines inducing protective immunity to such strains before they appear. This clinical trial is designed to evaluate the safety and immunogenicity of Multimeric-001 (M-001), which contains conserved epitopes of influenza A and B. M-001 is able to induce both humoral and cellular immunity and provides broad strain coverage. METHODS: In a multicenter, randomized, double-blind, and controlled phase IIb trial, 222 healthy volunteers aged 18 to 60 years will be randomized into 3 groups (1:1:1) to receive either 2 intramuscular injections of 0.5 mg M-001 (arm 1), 1.0 mg M-001 (arm 2), or saline (arm 3-placebo), before receiving an investigational (whole virus, inactivated, aluminum phosphate gel [AlPO4]-adjuvanted) prepandemic influenza vaccine (H5N1). Primary outcomes are safety and cellular immune responses (cell-mediated immunity [CMI]) induced by M-001, evaluated by multiparametric flow cytometry of intracellular cytokines. The secondary outcome is the serum hemagglutination inhibition (HAI) titer toward the H5N1 vaccine strain. Additionally, exploratory outcomes include evaluation of CMI by quantitative reverse transcription polymerase chain reaction of cytokine mRNA, HAI titers toward H5-drifted strains, serum single radial hemolysis titers toward the H5N1 study vaccine, and the association between CMI markers and antibody response. DISCUSSION: There is a need for influenza vaccines that give the population a broader protection against multiple strains of influenza virus. M-001 might be such vaccine which will be tested in this current trial as a standalone vaccine and as a pandemic primer. Both cellular and humoral immune responses will be evaluated. TRIAL REGISTRATION: EudraCT number: 2015-001979-46.


Assuntos
Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Adolescente , Adulto , Citocinas/metabolismo , Método Duplo-Cego , Feminino , Citometria de Fluxo , Testes de Inibição da Hemaglutinação , Humanos , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Vírus da Influenza A/imunologia , Vírus da Influenza B/imunologia , Vacinas contra Influenza/efeitos adversos , Masculino , Pessoa de Meia-Idade , Projetos de Pesquisa , Adulto Jovem
5.
J Virol Methods ; 234: 186-92, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27142113

RESUMO

The detection or quantification of retroviruses is often achieved using an antigen-capture ELISA (AC-ELISA) that targets the Gag capsid (CA) protein. We report here the development of an AC-ELISA specific for the p27-CA protein of HERV-K(HML-2). A monoclonal p27-specific antibody is used for capture and a polyclonal anti-p27-CA immune serum generated in rabbits serves for detection. The assay was shown to be specific for HERV-K(HML-2), showing no evidence of cross reactivity with the human retroviruses HIV-1, HIV-2 and HTLV-1 or with XMRV (as a model non-human gammaretrovirus). Using purified recombinant antigen, the limit of detection was shown to be 130pg/ml. The AC-ELISA can be used to quantify HERV-K(HML-2) expression in teratocarcinoma cell lines and to normalize HERV particles generated by transfecting HEK 293T cells with full-length molecular clones. This novel AC-ELISA also proved useful in studies of virus regulation, for example in demonstrating that HERV-K(HML-2) expression is dramatically enhanced by overexpression of Staufen-1, a binding partner of the HERV-K(HML-2) Rec protein. This specific and sensitive HERV-K(HML-2) AC-ELISA will be a useful tool for investigating many aspects of endogenous retroviruses, from basic research to the role they may play in human diseases or as a surrogate marker for particular diseases.


Assuntos
Proteínas do Capsídeo/análise , Retrovirus Endógenos/isolamento & purificação , Ensaio de Imunoadsorção Enzimática/métodos , Proteínas do Capsídeo/química , Proteínas do Capsídeo/imunologia , Retrovirus Endógenos/imunologia , Produtos do Gene gag/análise , Produtos do Gene gag/imunologia , Células HEK293 , Humanos , Limite de Detecção , RNA Viral , Sensibilidade e Especificidade , Proteínas do Envelope Viral/análise , Proteínas do Envelope Viral/imunologia
6.
Virology ; 489: 1-11, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26685794

RESUMO

Although safety concerns preclude the use of live attenuated HIV vaccines in humans, they provide a useful system for identifying the elusive correlates of protective immunity in the SIV/macaque animal model. However, a number of pieces of evidence suggest that protection may result from prior occupancy of susceptible target cells by the vaccine virus rather than the immune response. To address this, we developed a Nef-deletion variant of an RT-SHIV whose active replication could be shut off by treatment with RT-inhibitors. Groups of macaques were inoculated with the ∆Nef-RT-SHIV and immune responses allowed to develop before antiretroviral treatment and subsequent challenge with wild-type SIVmac239. Vaccinated animals either resisted infection fully or significantly controlled the subsequent viremia. However, there was no difference between animals undergoing replication of the vaccine virus and those without. This strongly suggests that competition for available target cells does not play a role in protection.


Assuntos
Vacinas contra a AIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/fisiologia , Replicação Viral , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Animais , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Linfócitos T/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Viremia/imunologia , Viremia/virologia
7.
J Clin Microbiol ; 52(8): 2977-83, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24920768

RESUMO

An increase in the proportion of ambiguous base calls in HIV-1 pol population sequences during the course of infection has been demonstrated in different study populations, and sequence ambiguity thresholds to classify infections as recent or nonrecent have been suggested. The aim of our study was to evaluate sequence ambiguities as a candidate biomarker for use in an HIV-1 incidence assay using samples from antiretroviral treatment-naive seroconverters with known durations of infection (German HIV-1 Seroconverter Study). We used 2,203 HIV-1 pol population sequences derived from 1,334 seroconverters to assess the sequence ambiguity method (SAM). We then compared the serological incidence BED capture enzyme immunoassay (BED-CEIA) with the SAM for a subset of 723 samples from 495 seroconverters and evaluated a multianalyte algorithm that includes BED-CEIA results, SAM results, viral loads, and CD4 cell counts for 453 samples from 325 seroconverters. We observed a significant increase in the proportion of sequence ambiguities with the duration of infection. A sequence ambiguity threshold of 0.5% best identified recent infections with 76.7% accuracy. The mean duration of recency was determined to be 208 (95% confidence interval, 196 to 221) days. In the subset analysis, BED-CEIA achieved a significantly higher accuracy than the SAM (84.6 versus 75.5%, P < 0.001) and results were concordant for 64.2% (464/723) of the samples. Also, the multianalyte algorithm did not show better accuracy than the BED-CEIA (83.4 versus 84.3%, P = 0.786). In conclusion, the SAM and the multianalyte algorithm including SAM were inferior to the BED-CEIA, and the proportion of sequence ambiguities is therefore not a preferable biomarker for HIV-1 incidence testing.


Assuntos
Marcadores Genéticos , Variação Genética , Infecções por HIV/diagnóstico , Infecções por HIV/virologia , HIV-1/genética , Produtos do Gene pol do Vírus da Imunodeficiência Humana/genética , Adulto , Contagem de Linfócito CD4 , Estudos de Coortes , Feminino , Genótipo , HIV-1/classificação , HIV-1/isolamento & purificação , Humanos , Masculino , Estudos Prospectivos , Carga Viral
8.
PLoS Pathog ; 10(5): e1004110, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24809349

RESUMO

Infection with influenza virus can result in massive pulmonary infiltration and potentially fatal immunopathology. Understanding the endogenous mechanisms that control immunopathology could provide a key to novel adjunct therapies for this disease. Here we show that the cytokine IL-27 plays a crucial role in protection from exaggerated inflammation during influenza virus infection. Using Il-27ra-/- mice, IL-27 was found to limit immunopathology, neutrophil accumulation, and dampened TH1 or TH17 responses via IL-10-dependent and -independent pathways. Accordingly, the absence of IL-27 signals resulted in a more severe disease course and in diminished survival without impacting viral loads. Consistent with the delayed expression of endogenous Il-27p28 during influenza, systemic treatment with recombinant IL-27 starting at the peak of virus load resulted in a major amelioration of lung pathology, strongly reduced leukocyte infiltration and improved survival without affecting viral clearance. In contrast, early application of IL-27 impaired virus clearance and worsened disease. These findings demonstrate the importance of IL-27 for the physiological control of immunopathology and the potential value of well-timed IL-27 application to treat life-threatening inflammation during lung infection.


Assuntos
Imunidade Inata , Vírus da Influenza A/imunologia , Interleucinas/fisiologia , Infecções por Orthomyxoviridae/imunologia , Infecções Respiratórias/imunologia , Animais , Células Cultivadas , Embrião de Galinha , Citoproteção/genética , Citoproteção/imunologia , Imunidade Inata/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/genética , Receptores de Citocinas/genética , Receptores de Interleucina , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Fatores de Tempo
9.
Virology ; 448: 65-73, 2014 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-24314637

RESUMO

Preclinical evaluation in a small animal model would help the development of gene therapies and vaccines based on foamy virus vectors. The establishment of persistent, non-pathogenic infection with the prototype foamy virus in mice and rabbits has been described previously. To extend this spectrum of available animal models, hamsters were inoculated with infectious cell supernatant or bioballistically with a foamy virus plasmid. In addition, a novel foamy virus from a rhesus macaque was isolated and characterised genetically. Hamsters and mice were infected with this new SFVmac isolate to evaluate whether hamsters are also susceptible to infection. Both hamsters and mice developed humoral responses to either virus subtype. Virus integration and replication in different animal tissues were analysed by PCR and co-cultivation. The results strongly indicate establishment of a persistent infection in hamsters. These studies provide a further small animal model for studying FV-based vectors in addition to the established models.


Assuntos
Cricetinae , Modelos Animais , Doenças dos Primatas/virologia , Infecções por Retroviridae/veterinária , Vírus Espumoso dos Símios/fisiologia , Replicação Viral , Animais , Anticorpos Antivirais/imunologia , Cricetinae/imunologia , Cricetinae/virologia , Macaca mulatta , Mesocricetus , Camundongos , Doenças dos Primatas/imunologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Vírus Espumoso dos Símios/genética , Vírus Espumoso dos Símios/isolamento & purificação
10.
Virology ; 422(2): 402-12, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22153299

RESUMO

Mouse monoclonal antibodies with varying specificities against the Gag capsid of simian and human immunodeficiency virus (SIV/HIV) were generated by immunizing mice with whole inactivated SIVagmTYO-1. Monoclonal antibody AG3.0 showed the broadest reactivity recognizing the Gag capsid protein (p24-27) and Gag precursors p38, p55, and p150 of HIV-1, HIV-2, SIVmac, and SIVagm. Using overlapping peptides, the AG3.0 epitope was mapped in capsid to a sequence (SPRTLNA) conserved among HIV-1, HIV-2, SIVrcm, SIVsm/mac, and SIVagm related viruses. Because of its broad cross-reactivity, AG3.0 was used to develop an antigen capture assay with a lower detection limit of 100 pg/ml HIV-1 Gag p24. Interestingly, AG3.0 was found to have a faster binding on/off rate for SIVagmVer and SIVmac Gag than for SIVagmSab Gag, possibly due to differences outside the SPRTLNA motif. In addition, the ribonucleic acid (RNA) coding for AG3.0 was sequenced to facilitate the development of humanized monoclonal antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas do Capsídeo/imunologia , HIV-1/imunologia , HIV-2/imunologia , Vírus da Imunodeficiência Símia/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/metabolismo , Linfócitos T CD4-Positivos , Linhagem Celular , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Humanos , Camundongos , Dados de Sequência Molecular
11.
J Virol Methods ; 174(1-2): 22-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21419167

RESUMO

Pairs of recombinant MVA (Modified Vaccinia Ankara) and FPV (Fowlpox Virus) expressing the same transgene are reasonable candidates for prime/boost regimens, because cross-reacting immune responses between the two vectors, both non-replicative in mammalian hosts, are very limited. The acceptor virus FPD-Red, a derivative of FPV, carrying a red fluorescent protein gene flanked by the homology regions of MVA deletion III, was constructed. The same MVA Transfer Plasmid Green, designed to insert transgenes into the MVA deletion III locus, can therefore be used to transfer transgenes into both acceptor viruses MVA-Red and FPD-Red with the described recently Red-to-Green gene swapping method. Cells infected by either recombinant virus can be sorted differentially by a simple and reliable FACS-based purification protocol. The procedure is carried out in primary chick embryo fibroblasts grown in serum-free media and was applied to the production of three rMVA/rFPV pairs expressing the H5N1 avian influenza antigens M1, M2 and NP. The viral genes were human codon-optimized and expressed at high levels in both chick and mammalian cells. Both single-step and multiple-step growth analyses showed no significant differences in growth due to the transgenes in either rMVA or rFPV derivatives.


Assuntos
Portadores de Fármacos , Vírus da Varíola das Aves Domésticas/genética , Vetores Genéticos , Vaccinia virus/genética , Vacinas Virais/genética , Animais , Linhagem Celular , Galinhas , Terapia Genética/métodos , Humanos , Virus da Influenza A Subtipo H5N1/genética , Transdução Genética , Transgenes , Vacinas Sintéticas/genética , Proteínas Virais/genética
12.
Vaccine ; 29(8): 1690-9, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21195079

RESUMO

The recent H1N1 influenza pandemic and the inevitable delay between identification of the virus and production of the specific vaccine have highlighted the urgent need for new generation influenza vaccines that can preemptively induce broad immunity to different strains of the virus. In this study we have produced AAV-based vectors expressing the A/Mexico/4603/2009 (H1N1) hemagglutinin (HA), nucleocapsid (NP) and the matrix protein M1 and have evaluated their ability to induce specific immune response and protect mice against homologous and heterologous challenge. Each of the vaccine vectors elicited potent cellular and humoral immune responses in mice. Although immunization with AAV-M1 did not improve survival after challenge with the homologous strain, immunization with the AAV-H1 and AAV-NP vectors resulted in survival of all mice, as did inoculation with a combination of all three vectors. Furthermore, trivalent vaccination also conferred partial protection against challenge with the highly heterologous and virulent A/PR/8/34 strain of H1N1 influenza.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Feminino , Células HEK293 , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunidade Celular , Imunidade Humoral , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Proteínas do Nucleocapsídeo , Infecções por Orthomyxoviridae/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Transfecção , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Ensaio de Placa Viral
13.
Virology ; 399(1): 87-97, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20097401

RESUMO

SIVagm does not induce disease in its African green monkey (AGM) host. In comparison, the hybrid simian-human immunodeficiency virus SHIV89.6P that carries the HIV env gene induces disease in rhesus macaques more rapidly than the SIVmac parent virus. To address the possibility that this enhancement of disease by HIV env would also occur when present in SIVagm, a full-length SIVagm/89.6Penv chimeric lentivirus genome (termed SHIV-MP) was constructed. SHIV-MP replicated similarly to SIVagm in simian peripheral blood mononuclear cells (PBMCs). In inoculated AGMs, rhesus macaques and pig-tailed (PT) macaques the absolute number of CD4(+) T lymphocytes remained at normal levels. The peak levels of productively infected cells in SHIV-MP-infected monkeys ranged from 10(1) to 10(2) per 10(6) PBMCs, while in SIVagm infected macaques the levels were 10-100-fold higher. The env gene of SHIV89.6P therefore appears insufficient to confer acute pathogenicity to a non-pathogenic primate lentivirus due to poor in vivo replication.


Assuntos
Chlorocebus aethiops/virologia , Genes env/fisiologia , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Linhagem Celular , Chlorocebus aethiops/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Genes env/genética , HIV-1/fisiologia , Humanos , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/fisiologia , Carga Viral , Replicação Viral/genética , Replicação Viral/fisiologia
14.
J Gen Virol ; 90(Pt 10): 2513-2518, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19587137

RESUMO

As a prelude to primate studies, the immunogenicity of wild-type and codon-optimized versions of simian immunodeficiency virus (SIV)agm Gag DNA, with and without co-administered granulocyte-macrophage colony-stimulating factor (GM-CSF) DNA, was directly compared in two strains of mice. Gag-specific T cells in the splenocytes of BALB/c and C57BL/6 mice immunized by gene gun were quantified by ELISpot using panels of overlapping synthetic peptides (15mers) spanning the entire capsid proteins of SIVagm, SIVmac and human immunodeficiency virus type 1. Specific antibodies were measured by ELISA. Codon optimization was shown to significantly increase the immune response to the DNA immunogens, reducing the amount of DNA necessary to induce cellular and antibody responses by one and two orders of magnitude, respectively. Co-administration of murine GM-CSF DNA was necessary for the induction of high level T- and B-cell responses. Finally, it was possible to identify both known and novel T-cell epitopes in the Gag proteins of the three viruses.


Assuntos
DNA Viral/imunologia , Produtos do Gene env/imunologia , Proteínas Oncogênicas de Retroviridae/imunologia , Vacinas de DNA/imunologia , Proteínas Virais de Fusão/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Sequência de Aminoácidos , Animais , Produtos do Gene env/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas de Retroviridae/química , Vírus da Imunodeficiência Símia , Baço/citologia , Proteínas Virais de Fusão/química , Replicação Viral , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química
15.
Retrovirology ; 6: 60, 2009 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-19545395

RESUMO

BACKGROUND: We investigated the interplay between complement and antibodies upon priming with single-cycle replicating viral vectors (SCIV) encoding SIV antigens combined with Adeno5-SIV or SCIV pseudotyped with murine leukemia virus envelope boosting strategies. The vaccine was applied via spray-immunization to the tonsils of rhesus macaques and compared with systemic regimens. RESULTS: Independent of the application regimen or route, viral loads were significantly reduced after challenge with SIVmac239 (p < 0.03) compared to controls. Considerable amounts of neutralizing antibodies were induced in systemic immunized monkeys. Most of the sera harvested during peak viremia exhibited a trend with an inverse correlation between complement C3-deposition on viral particles and plasma viral load within the different vaccination groups. In contrast, the amount of the observed complement-mediated lysis did not correlate with the reduction of SIV titres. CONCLUSION: The heterologous prime-boost strategy with replication-deficient viral vectors administered exclusively via the tonsils did not induce any neutralizing antibodies before challenge. However, after challenge, comparable SIV-specific humoral immune responses were observed in all vaccinated animals. Immunization with single cycle immunodeficiency viruses mounts humoral immune responses comparable to live-attenuated immunodeficiency virus vaccines.


Assuntos
Anticorpos Antivirais/imunologia , Proteínas do Sistema Complemento/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Adenovírus Humanos/genética , Aerossóis , Animais , Anticorpos Antivirais/sangue , Vetores Genéticos , Vírus da Leucemia Murina/genética , Macaca mulatta , Testes de Neutralização , Tonsila Palatina/virologia , Vírus da Imunodeficiência Símia/genética , Carga Viral
16.
J Virol ; 82(24): 12464-71, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18842729

RESUMO

Prion diseases have a significant inflammatory component. Glia activation, which is associated with increased production of cytokines and chemokines, may play an important role in disease development. Among the chemokines upregulated highly and early upregulated during scrapie infections are ligands of CXCR3. To gain more insight into the role of CXCR3 in a prion model, CXCR3-deficient (CXCR3(-/-)) mice were infected intracerebrally with scrapie strain 139A and characterized in comparison to similarly infected wild-type controls. CXCR3(-/-) mice showed significantly prolonged survival times of up to 30 days on average. Surprisingly, however, they displayed accelerated accumulation of misfolded proteinase K-resistant prion protein PrP(Sc) and 20 times higher infectious prion titers than wild-type mice at the asymptomatic stage of the disease, indicating that these PrP isoforms may not be critical determinants of survival times. As demonstrated by immunohistochemistry, Western blotting, and gene expression analysis, CXCR3-deficient animals develop an excessive astrocytosis. However, microglia activation is reduced. Quantitative analysis of gliosis-associated gene expression alterations demonstrated reduced mRNA levels for a number of proinflammatory factors in CXCR3(-/-) compared to wild-type mice, indicating a weaker inflammatory response in the knockout mice. Taken together, this murine prion model identifies CXCR3 as disease-modifying host factor and indicates that inflammatory glial responses may act in concert with PrP(Sc) in disease development. Moreover, the results indicate that targeting CXCR3 for treatment of prion infections could prolong survival times, but the results also raise the concern that impairment of microglial migration by ablation or inhibition of CXCR3 could result in increased accumulation of misfolded PrP(Sc).


Assuntos
Doenças Priônicas/metabolismo , Receptores CXCR3/metabolismo , Animais , Biomarcadores/metabolismo , Regulação da Expressão Gênica , Gliose/genética , Gliose/metabolismo , Gliose/patologia , Camundongos , Camundongos Knockout , Microglia/metabolismo , Doenças Priônicas/genética , Doenças Priônicas/patologia , Receptores CXCR3/deficiência , Receptores CXCR3/genética , Taxa de Sobrevida , Fatores de Tempo
17.
J Med Virol ; 80(2): 192-200, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18098144

RESUMO

A human CD4-positive T cell line from a donor homozygous negative for the chemokine receptor CCR5 was established, characterized, and used for determining the coreceptor usage of human immunodeficiency virus type 1 (HIV-1) isolates. Clones of this IL-2 dependent human T-cell lymphotropic virus type 1 (HTLV-I) immortalized cell line, named IsnoR5 clones 1 and 2, are susceptible to infection by HIV-1 isolates that use CXCR4 as a coreceptor but resistant to infection by CCR5 tropic HIV-1 viruses. HIV-1 isolates whose replication is inhibited in IsnoR5 cells in the presence of the bicyclam AMD 3100, a CXCR4 specific inhibitor, utilize a coreceptor distinct from CCR5 and CXCR4. Using a panel of primary HIV-1 isolates we have shown that a single T cell line is sufficient to discriminate between use of CCR5, CXCR4 or an alternative coreceptor. As IsnoR5 clone 1 cells revealed the existence of even minor populations of CXCR4-using virus variants, they could be useful for the early identification of changes in coreceptor usage in HIV infected individuals facilitating the timely introduction of appropriate clinical treatments.


Assuntos
Linfócitos T CD4-Positivos/virologia , HIV-1/classificação , Receptores CCR5/deficiência , Receptores de HIV/deficiência , Linhagem Celular , Transformação Celular Viral , Infecções por HIV/virologia , HIV-1/crescimento & desenvolvimento , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos
18.
J Virol ; 81(23): 13180-90, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17898066

RESUMO

The development of needle-free vaccines is one of the recently defined "grand challenges in global health" (H. Varmus, R. Klausner, R. Klausner, R. Zerhouni, T. Acharya, A. S. Daar, and P. A. Singer, Science 302:398-399, 2003). To explore whether a natural pathway to the inductive site of the mucosa-associated lymphatic tissue could be exploited for atraumatic immunization purposes, replication-deficient viral vector vaccines were sprayed directly onto the tonsils of rhesus macaques. Tonsillar immunization with viral vector vaccines encoding simian immunodeficiency virus (SIV) antigens induced cellular and humoral immune responses. Viral RNA levels after a stringent SIV challenge were reduced, providing a level of protection similar to that observed after systemic immunization with the same vaccines. Thus, atraumatic oral spray immunization with replication-deficient vectors can overcome the epithelial barrier, deliver the vaccine antigen to the mucosa-associated lymphatic tissue, and avoid induction of tolerance, providing a novel approach to circumvent acceptability problems of syringe and needle vaccines for children and in developing countries.


Assuntos
Administração Oral , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/imunologia , Animais , Macaca mulatta , Tonsila Palatina/imunologia , RNA Viral/sangue , Vacinas contra a SAIDS/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Carga Viral
19.
Viral Immunol ; 19(3): 448-57, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16987063

RESUMO

As part of a European multicenter study designed to determine the optimal combination and order of a mixed-modality vaccine against acquired immunodeficiency syndrome, rhesus monkeys received a combination of three different vectors, all expressing the same Simian Immunodeficiency Virus (SIV) genes followed by mucosal challenge with highly pathogenic SIV. In the study reported here, animals were primed with DNA followed by one booster immunization with Semliki Forest Virus (SFV) and two immunizations with modified Vaccinia Ankara (MVA). To address the relevance of mucosal immunization, we compared systemic versus a combination of systemic and mucosal antigen application. Although all vaccinees became infected after intrarectal challenge with SIV, most (six of eight) were protected from profound loss of CD4+ cells. In addition, vaccinees showed lower viral loads than did controls (p < 0.05). Overall, these protective effects were more pronounced in those animals whose schedule included immunization via the mucosa. In summary, the vaccine regimen used here achieved one important criterion of efficacy: the suppression of disease development as indicated by conservation of CD4+ cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vacinas contra a SAIDS/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Administração Retal , Animais , Anticorpos Antivirais/sangue , Vetores Genéticos , Imunização , Imunização Secundária , Macaca mulatta , Vacinas contra a SAIDS/genética , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/patogenicidade , Vacinas de DNA , Vaccinia virus/genética , Vaccinia virus/imunologia
20.
J Biotechnol ; 124(3): 615-25, 2006 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-16563543

RESUMO

Retroviral vectors have yet not been tested for their potential as vaccines despite their frequent utilization in gene therapy allowing for highly efficient gene transfer into a number of cell types and their suitability for large-scale production in biotechnology. To investigate MLV-based vectors suitability for inducing immune response against HIV-1-antigens, we generated a MLV(HIV-1) pseudotype vector enabling CD4-specific transduction of HIV-1 genes env, vpu, tat and rev originating from the pathogenic SHIV-89.6P. Functional expression of the lentiviral genes in packaging cells, human and rhesus CD4+ target cells was demonstrated by various assays. Following highly efficient ex vivo transduction, up to 3.4x10(7) autologous, transfer vector-positive rhesus peripheral blood mononuclear cells (rhPBMCs) were re-inoculated into a rhesus macaque. Five weeks after the initial inoculation HIV-1 Env-specific antibodies were detected using ELISA. ELIspot-assay revealed the induction of a HIV-1 Rev and Env-specific CTL-response 7.5 weeks after immunization. Thus, these novel MLV(HIV-1) vectors facilitate efficient transduction and subsequent expression of HIV-1-genes in CD4-positive host cells. Induction of both humoral and cellular HIV-1-specific immune responses in vivo confirmed their potential as an effective HIV-1 vaccine to be further studied in SHIV/rhesus macaque model of lentivirus infection.


Assuntos
Vacinas contra a AIDS/administração & dosagem , Formação de Anticorpos/imunologia , HIV-1/genética , HIV-1/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Retroviridae/genética , Transfecção/métodos , Vacinas contra a AIDS/genética , Animais , Clonagem Molecular/métodos , Vetores Genéticos/genética , Macaca mulatta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA