Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Adv Virus Res ; 97: 187-243, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28057259

RESUMO

Safety tested Modified Vaccinia virus Ankara (MVA) is licensed as third-generation vaccine against smallpox and serves as a potent vector system for development of new candidate vaccines against infectious diseases and cancer. Historically, MVA was developed by serial tissue culture passage in primary chicken cells of vaccinia virus strain Ankara, and clinically used to avoid the undesirable side effects of conventional smallpox vaccination. Adapted to growth in avian cells MVA lost the ability to replicate in mammalian hosts and lacks many of the genes orthopoxviruses use to conquer their host (cell) environment. As a biologically well-characterized mutant virus, MVA facilitates fundamental research to elucidate the functions of poxvirus host-interaction factors. As extremely safe viral vectors MVA vaccines have been found immunogenic and protective in various preclinical infection models. Multiple recombinant MVA currently undergo clinical testing for vaccination against human immunodeficiency viruses, Mycobacterium tuberculosis or Plasmodium falciparum. The versatility of the MVA vector vaccine platform is readily demonstrated by the swift development of experimental vaccines for immunization against emerging infections such as the Middle East Respiratory Syndrome. Recent advances include promising results from the clinical testing of recombinant MVA-producing antigens of highly pathogenic avian influenza virus H5N1 or Ebola virus. This review summarizes our current knowledge about MVA as a unique strain of vaccinia virus, and discusses the prospects of exploiting this virus as research tool in poxvirus biology or as safe viral vector vaccine to challenge existing and future bottlenecks in vaccinology.


Assuntos
Vetores Genéticos/genética , Vaccinia virus/genética , Vacinas Virais/imunologia , Viroses/prevenção & controle , Animais , Desenho de Fármacos , Vetores Genéticos/história , Vetores Genéticos/metabolismo , História do Século XX , História do Século XXI , Humanos , Vaccinia virus/metabolismo , Vacinas Virais/genética , Vacinas Virais/história , Viroses/virologia
2.
Gene Ther ; 23(10): 753-759, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27416077

RESUMO

Immune responses against multiple epitopes are required for the prevention of hepatitis C virus (HCV) infection, and the progression to phase I trials of candidates may be guided by comparative immunogenicity studies in non-human primates. Four vectors, DNA, SFV, human serotype 5 adenovirus (HuAd5) and Modified Vaccinia Ankara (MVA) poxvirus, all expressing hepatitis C virus Core, E1, E2 and NS3, were combined in three prime-boost regimen, and their ability to elicit immune responses against HCV antigens in rhesus macaques was explored and compared. All combinations induced specific T-cell immune responses, including high IFN-γ production. The group immunized with the SFV+MVA regimen elicited higher E2-specific responses as compared with the two other modalities, while animals receiving HuAd5 injections elicited lower IL-4 responses as compared with those receiving MVA. The IFN-γ responses to NS3 were remarkably similar between groups. Only the adenovirus induced envelope-specific antibody responses, but these failed to show neutralizing activity. Therefore, the two novel regimens failed to induce superior responses as compared with already existing HCV vaccine candidates. Differences were found in response to envelope proteins, but the relevance of these remain uncertain given the surprisingly poor correlation with immunogenicity data in chimpanzees, underlining the difficulty to predict efficacy from immunology studies.


Assuntos
Linfócitos B/imunologia , Epitopos/genética , Hepacivirus/imunologia , Linfócitos T/imunologia , Vacinas contra Hepatite Viral/imunologia , Adenoviridae/genética , Animais , Linhagem Celular , Cricetinae , Epitopos/imunologia , Vetores Genéticos/genética , Imunogenicidade da Vacina , Interferon gama/sangue , Interleucina-4/sangue , Macaca mulatta , Masculino , Vaccinia virus/genética , Vacinas contra Hepatite Viral/genética
3.
Vet Microbiol ; 187: 70-74, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-27066711

RESUMO

Porcine circovirus type 2 (PCV2) is the assumed causative agent of a number of different diseases summarized as porcine circovirus diseases (PCVD). The virus is shed via different se- and excretions of PCV2 infected pigs. Transmission of the virus occurs horizontally and vertically either by oronasal or diaplacental infection. Recent research emphasizes the importance of diaplacental PCV2 infection or the infection in early stages of the piglet's life attributable to excretion of PCV2 by the dams within the suckling period. To estimate the prevalence of intrauterine PCV2 infections under field conditions in Bavaria the PCV2 status of farrowing sows (n=198) and corresponding pre-suckling piglets (n=590) of 20 piglet producing farms was examined. PCV2 viral load and anti-PCV2 antibodies in the serum of the sows and piglets were examined at time of farrowing or before colostrum intake, respectively. PCV2 excretion of the sows via saliva, feces and urine was examined additionally. PCV2 specific antibodies in the serum of the sows were detectable on 11 farms with a mean in herd seroprevalence of 35.5% in these farms. Only 0.65% of all samples collected from 198 sows were positive for PCV2 DNA (serum: 1%; feces: 0.5%; saliva: 0.5%; urine: 0.6%). PCV2 DNA was detectable in sample material from seronegative sows as well as from seropositive sows. In none of the pre-suckling serum samples of the piglets IgG antibodies against PCV2 or PCV2 DNA were present. No correlation between the antibody- and viremia status of the sows and the PCV2 excretion was detectable. In contrast to reports about a high prevalence of viremic pre-suckling piglets in the suckling period in North America, the results of the present study reveal that diaplacental infection with PCV2 is comparatively rare in Southern Germany and infection of piglets within the suckling period seems to be more likely.


Assuntos
Infecções por Circoviridae/veterinária , Circovirus/fisiologia , Doenças dos Suínos/epidemiologia , Animais , Animais Recém-Nascidos/virologia , Anticorpos Antivirais/sangue , Infecções por Circoviridae/epidemiologia , Infecções por Circoviridae/virologia , Feminino , Alemanha/epidemiologia , Prevalência , Suínos , Doenças dos Suínos/virologia , Carga Viral , Eliminação de Partículas Virais
4.
Vet Microbiol ; 176(3-4): 337-43, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25669598

RESUMO

PCV2 can be divided into three different genotypes: PCV2a, PCV2b and PCV2c. Since 2004/2005 PCV2b has become the predominant genotype in the domestic pig population worldwide. In the years 2010 and 2012 PCV2b mutant strains (mPCV2), classified as PCV2b-1C strains, were detected in porcine circovirus diseases (PCVD) affected pigs in China and the United States, respectively. Within one year (April 2013-April 2014) newly emerging mPCV2 strains were isolated in seven German pig farms routinely vaccinating against PCV2. Histopathological, clinical and molecular biological findings including in-situ hybridization (ISH) and real-time PCR indicate PCVD in the affected animals. Characterized isolates from five farms were closely related to the PCV2b-1C reference strain BDH (GenBank no. HM038017), whereas strains from two other farms were only 99.1% and 99.0% identical (based on the nucleotide sequence of the complete genome) to mPCV2 strain BDH, respectively.


Assuntos
Infecções por Circoviridae/veterinária , Circovirus/genética , Doenças Transmissíveis Emergentes/veterinária , Sus scrofa , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/virologia , Animais , Sequência de Bases , Infecções por Circoviridae/epidemiologia , Doenças Transmissíveis Emergentes/virologia , Genótipo , Alemanha/epidemiologia , Hibridização In Situ/veterinária , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Análise de Sequência de DNA/veterinária , Suínos
5.
Acta Clin Belg ; 70(1): 11-5, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25176405

RESUMO

OBJECTIVES: This study aimed to establish acceptable quality control ranges for temocillin disk diffusion tests and Etest(®) minimal inhibitory concentrations. METHODS: According to Clinical and Laboratory Standards Institute (CLSI) guideline, a Tier 2 quality control study was performed and involves seven laboratories. Each of them tested 10 replicates of two quality control strains (Escherichia coli ATCC 25922 and E. coli ATCC 35218) on three different media lots and, for disk diffusion, two disk lots. RESULTS: Proposed zone diameter quality control ranges were 12-25 mm for E. coli ATCC 25922 and 19-28 mm for E. coli ATCC 35218. Proposed Etest quality control ranges were 3-24 mg/l for E. coli ATCC 25922 and 2-6 mg/l E. coli ATCC 35218. CONCLUSION: Based on our results, we would advise the use of E. coli ATCC 35218 as QC strain for temocillin susceptibility testing and Etest because ranges obtained are narrower than with E. coli ATCC 25922 and do not overlap temocillin breakpoint.


Assuntos
Testes de Sensibilidade a Antimicrobianos por Disco-Difusão/normas , Escherichia coli , Penicilinas , Controle de Qualidade , Padrões de Referência
6.
Allergy ; 68(8): 1021-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23909913

RESUMO

BACKGROUND: Modified vaccinia virus Ankara (MVA)-encoding antigens are considered as safe vaccine candidates for various infectious diseases in humans. Here, we investigated the immune-modulating properties of MVA-encoding ovalbumin (MVA-OVA) on the allergen-specific immune response. METHODS: The immune-modulating properties of MVA-OVA were investigated using GM-CSF-differentiated BMDCs from C57BL/6 mice. OVA expression upon MVA-OVA infection of BMDCs was monitored. Activation and maturation markers on viable MVA-OVA-infected mDCs were analyzed by flow cytometry. Secretion of INF-γ, IL-2, and IL-10 was determined in a co-culture of BMDCs infected with wtMVA or MVA-OVA and OVA-specific OT-I CD8(+) and OT-II CD4(+ ) T cells. BALB/c mice were vaccinated with wtMVA, MVA-OVA, or PBS, sensitized to OVA/alum and challenged with a diet containing chicken egg white. OVA-specific IgE, IgG1, and IgG2a and cytokine secretion from mesenteric lymph node (MLN) cells were analyzed. Body weight, body temperature, food uptake, intestinal inflammation, and health condition of mice were monitored. RESULTS: Infection with wtMVA and MVA-OVA induced comparable activation of mDCs. MVA-OVA-infected BMDCs expressed OVA and induced enhanced IFN-γ and IL-2 secretion from OVA-specific CD8(+ ) T cells in comparison with OVA, wtMVA, or OVA plus wtMVA. Prophylactic vaccination with MVA-OVA significantly repressed OVA-specific IgE, whereas OVA-specific IgG2a was induced. MVA-OVA vaccination suppressed TH 2 cytokine production in MLN cells and prevented the onset of allergic symptoms and inflammation in a mouse model of OVA-induced intestinal allergy. CONCLUSION: Modified vaccinia virus Ankara-ovalbumin (MVA-OVA) vaccination induces a strong OVA-specific TH 1- immune response, likely mediated by the induction of IFN-γ and IgG2a. Finally, MVA-based vaccines need to be evaluated for their therapeutic potential in established allergy models.


Assuntos
Alérgenos/imunologia , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/prevenção & controle , Imunoterapia Adotiva/métodos , Mucosa Intestinal/imunologia , Vaccinia virus/imunologia , Vacinas Virais/imunologia , Alérgenos/genética , Alérgenos/uso terapêutico , Animais , Transplante de Medula Óssea/métodos , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/imunologia , Células Dendríticas/transplante , Células Dendríticas/virologia , Modelos Animais de Doenças , Hipersensibilidade Alimentar/genética , Inflamação/imunologia , Inflamação/prevenção & controle , Inflamação/virologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ovalbumina/genética , Ovalbumina/imunologia , Ovalbumina/uso terapêutico , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , Vacínia/genética , Vacínia/imunologia , Vacínia/patologia , Vaccinia virus/genética , Vacinas Virais/genética , Vacinas Virais/uso terapêutico
7.
Vaccine ; 27(45): 6296-9, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19840663

RESUMO

Highly pathogenic avian influenza viruses of the H5N1 subtype are responsible for an increasing number of infections in humans since 2003. More than 60% of the infections is lethal and new infections are reported frequently. In the light of the pandemic threat caused by these events the rapid availability of safe and effective vaccines is desirable. Modified vaccinia virus Ankara (MVA) expressing the HA gene of an influenza A/H5N1 virus is a promising candidate vaccine that induced protective immunity against infection with homologous and heterologous influenza A/H5N1 viruses in mice. We also evaluated the recombinant MVA vector expressing the HA of influenza A/H5N1 virus A/Vietnam/1194/04 (MVA-HA-VN/04) in non-human primates. Cynomolgus macaques were immunized twice and then challenged with influenza virus A/Vietnam/1194/04 (clade 1) or A/Indonesia/5/05 (clade 2.1) to assess the level of protective immunity. Immunization with MVA-HA-VN/04 induced (cross-reactive) antibodies and prevented virus replication in the upper and lower respiratory tract and the development of severe necrotizing bronchointerstitial pneumonia. Therefore MVA-HA-VN/04 is a promising vaccine candidate for the induction of protective immunity against highly pathogenic avian influenza A/H5N1 viruses.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Anticorpos Antivirais/sangue , Feminino , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Vaccinia virus/imunologia
8.
J Infect Dis ; 199(3): 405-13, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19061423

RESUMO

BACKGROUND: Highly pathogenic avian influenza viruses of the H5N1 subtype have been responsible for an increasing number of infections in humans since 2003. More than 60% of infected individuals die, and new infections are reported frequently. In light of the pandemic threat caused by these events, the rapid availability of safe and effective vaccines is desirable. Modified vaccinia virus Ankara (MVA) expressing the hemagglutinin (HA) gene of H5N1 viruses is a promising candidate vaccine that induced protective immunity against infection with homologous and heterologous H5N1 influenza virus in mice. METHODS: In the present study, we evaluated a recombinant MVA vector expressing the HA gene of H5N1 influenza virus A/Vietnam/1194/04 (MVA-HA-VN/04) in nonhuman primates. Cynomolgus macaques were immunized twice and then were challenged with influenza virus A/Vietnam/1194/04 (clade 1) or A/Indonesia/5/05 (clade 2.1) to assess the level of protective immunity. RESULTS: Immunization with MVA-HA-VN/04 induced (cross-reactive) antibodies and prevented virus replication in the upper and lower respiratory tract and the development of severe necrotizing bronchointerstitial pneumonia. CONCLUSION: Therefore, MVA-HA-VN/04 is a promising vaccine candidate for the induction of protective immunity against highly pathogenic H5N1 avian influenza viruses in humans.


Assuntos
Hemaglutininas/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza , Infecções por Orthomyxoviridae/prevenção & controle , Vaccinia virus/genética , Animais , Anticorpos Antivirais/sangue , Encéfalo/virologia , Hemaglutininas/genética , Hemaglutininas/metabolismo , Imuno-Histoquímica , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Recombinação Genética , Sistema Respiratório/virologia , Baço/virologia , Vacinas Sintéticas , Vaccinia virus/classificação , Vaccinia virus/metabolismo
9.
Cell Death Differ ; 13(1): 109-18, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16003387

RESUMO

Infection with viruses often protects the infected cell against external stimuli to apoptosis. Here we explore the balance of apoptosis induction and inhibition for infection with the modified vaccinia virus Ankara (MVA), using two MVA mutants with experimentally introduced deletions. Deletion of the E3L-gene from MVA transformed the virus from an inhibitor to an inducer of apoptosis. Noxa-deficient mouse embryonic fibroblasts (MEF) were resistant to MVA-DeltaE3L-induced apoptosis. When the gene encoding F1L was deleted from MVA, apoptosis resulted that required Bak or Bax. MVA-DeltaF1L-induced apoptosis was blocked by Bcl-2. When expressed in HeLa cells, F1L blocked apoptosis induced by forced expression of the BH3-only proteins, Bim, Puma and Noxa. Finally, biosensor analysis confirmed direct binding of F1L to BH3 domains. These data describe a molecular framework of how a cell responds to MVA infection by undergoing apoptosis, and how the virus blocks apoptosis by interfering with critical steps of its signal transduction.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a RNA/fisiologia , Vaccinia virus/fisiologia , Vaccinia virus/patogenicidade , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Deleção de Genes , Genes Virais , Células HeLa , Humanos , Camundongos , Mitocôndrias/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas de Ligação a RNA/genética , Homologia de Sequência de Aminoácidos , Vaccinia virus/genética , Proteínas Virais/genética , Virulência/genética , Virulência/fisiologia , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
10.
J Gen Virol ; 85(Pt 8): 2407-2419, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15269383

RESUMO

The immunogenicity and protective efficacy of a DNA and recombinant modified vaccinia Ankara (MVA) vaccine administered by two different routes were investigated. DNA expressing HIV-1 IIIB env, gag, RT, rev, tat and nef, and MVA expressing HIV-1 IIIB nef, tat and rev and simian immunodeficiency virus (SIV) macJ5 gag/pol and vaccinia HIV-1 env, were used as immunogens. Four cynomolgus macaques received DNA intramuscularly (i.m.) at month 0 and intrarectally (i.r.) and intra-orally (i.o.) at 2 months, followed by MVA i.m. at 4 months and i.r. and i.o. at 8 months. Another group of four monkeys received the same immunogens but only i.m. Overall, stronger cellular immune responses measured by ELISPOT and T-cell proliferation assay were detected in the group primed i.m. and boosted mucosally. Following homologous intravenous simian-human immunodeficiency virus (SHIV) challenge, one of eight vaccinated animals was completely protected. This monkey, immunized i.m. and i.r.+i.o., exhibited the highest levels of HIV Env, Nef and Tat antibodies, high HIV Tat cytotoxic T-lymphocyte activity and T-lymphocyte proliferative responses to HIV Env. Four weeks post-challenge none of the monkeys immunized i.m. and i.r.+i.o., and only two out of four animals immunized i.m., demonstrated detectable plasma viral RNA levels. In contrast, all eight control animals had demonstrable plasma viral RNA levels 4 weeks post-challenge. Thus, stronger cellular immune responses and reduction of challenge virus burden were demonstrated in animals immunized i.m. as well as mucosally, compared with animals immunized i.m. only. The breadth and magnitude of the induced immune responses correlated with protective efficacy.


Assuntos
Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vacinas de DNA/imunologia , Vaccinia virus/imunologia , Animais , Anticorpos Anti-HIV/sangue , Imunização , Interferon gama/biossíntese , Macaca fascicularis , Vacinas contra a SAIDS/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/administração & dosagem , Carga Viral
11.
Cancer Res ; 61(24): 8647-50, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11751378

RESUMO

Sequence analyses of the transporter associated with antigen processing (TAP) in tumor cell lines with deficient MHC class I surface expression identified a bp deletion at position 1489 near the ATP-binding domain of Tap1, causing a frameshift in one melanoma cell line. The impaired TAP1 protein expression was associated with deficient TAP2 protein expression, peptide binding, translocation, and MHC class I surface expression. Stable TAP1 gene transfer reconstitutes the described defects, whereas lysis by HLA-A2-restricted CTLs was still abrogated. This was attributable to a 2-bp insertion at position 890 in the HLA-A2 gene and was corrected after HLA-A2 cotransfection. This study describes for the first time mutations in two distinct components of the MHC class I antigen processing pathway, suggesting an immune selection against CTLs recognizing both TAP-dependent and -independent T-cell epitopes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , Antígeno HLA-A2/imunologia , Melanoma/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/genética , Apresentação de Antígeno/imunologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/imunologia , Carcinoma de Células Renais/metabolismo , Antígeno HLA-A2/biossíntese , Antígeno HLA-A2/genética , Humanos , Vigilância Imunológica/imunologia , Neoplasias Renais/genética , Neoplasias Renais/imunologia , Neoplasias Renais/metabolismo , Melanoma/genética , Melanoma/metabolismo , Mutação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Citotóxicos/imunologia , Transfecção , Células Tumorais Cultivadas
12.
Vaccine ; 19(25-26): 3526-36, 2001 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-11348720

RESUMO

The immunogenicity of two vector-based vaccines, either given alone or in a prime-boost regimen, was investigated. Cynomolgus macaques were immunised with modified vaccinia virus Ankara (MVA) expressing simian immunodeficiency virus (SIV)macJ5 env, gag-pol, nef, rev, and tat genes (MVA-SIVmac) or primed with a Semliki forest virus (SFV) vaccine expressing the same genes (SFV-SIVmac) and boosted with MVA-SIVmac. Generally, antibody responses, T-cell proliferative responses and cytotoxic T-cell responses remained low or undetectable in vaccinees receiving MVA-SIVmac or SFV-SIVmac alone. In contrast, monkeys who first received SFV-SIVmac twice and then were boosted with MVA-SIVmac showed increased antibody responses as well as high T-cell proliferative responses. Three of these vaccinees had cytotoxic T-lymphocytes directed against three or four of the gene products. No evidence of protection was seen against an intrarectal heterologous SIVsm challenge given 3 months after the last immunisation. The study demonstrates a prime-boost strategy that efficiently induces both humoral and cellular immune responses.


Assuntos
Vacinas contra a SAIDS/administração & dosagem , Vírus da Floresta de Semliki/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vaccinia virus/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/isolamento & purificação , Animais , Anticorpos Antivirais/biossíntese , Especificidade de Anticorpos , Vetores Genéticos , Imunidade Celular , Imunização Secundária , Ativação Linfocitária , Contagem de Linfócitos , Macaca fascicularis , RNA Viral/sangue , Vacinas contra a SAIDS/genética , Vírus da Floresta de Semliki/genética , Linfócitos T/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vaccinia virus/genética
13.
Virology ; 282(2): 329-37, 2001 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-11289815

RESUMO

It is speculated that a virus-encoded superantigen is involved in the pathogenesis of human and simian immunodeficiency virus infections and that the accessory protein Nef might be that superantigen. We are able to show, using a murine superantigen screening system, that Nef does not display features characteristic of a superantigen. Upon transfection into MHC class II expressing antigen-presenting cells, it is expressed, but fails to induce Vbeta-specific expansion of peripheral T lymphocytes, which is a characteristic feature of superantigens in mixed lymphocyte culture. Therefore, we cannot support the hypothesis that Nef is a superantigen. The observations in favor of that hypothesis must be explained by other mechanisms.


Assuntos
Produtos do Gene nef/imunologia , Antígenos HIV/imunologia , HIV/imunologia , Modelos Imunológicos , Vírus da Imunodeficiência Símia/imunologia , Superantígenos/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Divisão Celular , Técnicas de Cocultura , Citometria de Fluxo , Produtos do Gene nef/genética , Produtos do Gene nef/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Ativação Linfocitária , Camundongos , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Produtos do Gene nef do Vírus da Imunodeficiência Humana
14.
Vaccine ; 19(20-22): 2764-8, 2001 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-11282186

RESUMO

Modified vaccinia virus Ankara (MVA) has been used as an experimental vaccine vector against respiratory infections. We have tested the safety and immunogenicity of a recombinant virus expressing the hemagglutinin of measles virus (MVA-MV-H) using the mouse model of measles virus induced encephalitis and the cotton rat model for respiratory infection. MVA-MV-H proved to induce a TH1 response, neutralizing antibodies and conferred protection against both encephalitis and lung infection. The cotton rat is very sensitive to infection with replication competent vaccinia virus. In these animals MVA-MV-H proved to be a very well tolerated vaccine. However, the efficiency in the presence of MV specific maternal antibodies was low (even using a prime-boost strategy) and therefore might have to be improved.


Assuntos
Hemaglutininas Virais/imunologia , Vacina contra Sarampo/imunologia , Sarampo/prevenção & controle , Vacinas Sintéticas/imunologia , Vaccinia virus/imunologia , Animais , Anticorpos Antivirais/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Sigmodontinae , Vacinação , Vaccinia virus/genética
16.
J Med Primatol ; 29(3-4): 268-73, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11085589

RESUMO

A large number of recombinant of viral and bacterial systems have been engineered as vectors to express foreign genes for vaccination and/or gene therapy. A common problem is the immune response to the vector itself. The presence of anti-vector immune responses may preclude sufficient 'priming' or immunogenicity if pre-existing immune responses are present, or they may impair optimal 'boosting' upon repeated immunization or delivery with the same vector. To circumvent this problem we developed a strategy using different chimeric vectors which share only the expression of common specific antigens desired for immunization. This approach not only has the advantage of avoiding increased anti-vector responses, but allows the use of combinations of vectors which could subsequently present the same or related antigen differently to the immune system as well as at alternative sites to induce the optimal type of immunity against the pathogen of interest.


Assuntos
Antígenos Virais/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vacinas Sintéticas/imunologia , Animais , Antígenos Virais/genética , Quimera , Genes env , Genes gag , Genes nef , Genes pol , Genes rev , Genes tat , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle
17.
Int J Cancer ; 88(3): 432-8, 2000 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11054673

RESUMO

The melanosomal protein tyrosinase is considered as a target of specific immunotherapy against melanoma. Two tyrosinase-derived peptides are presented in association with HLA-A2.1 [Wölfel et al., Eur. J. Immunol., 24, 759-764 (1994)]. Peptide 1-9 (MLLAVLYCL) is generated from the putative signal sequence. The internal peptide 369-377 is posttranslationally converted at residue 371, and its presentation is dependent on functional TAP transporters and proteasomes [Mosse et al., J. exp. Med.187, 37-48 (1998)]. Herein, we report on the processing and transport requirements for the signal sequence-derived peptide 1-9 that were studied in parallel to those for peptide 369-377. After infection of TAP-deficient (T2) and TAP-positive (T1) cells with a Modified Vaccinia Ankara construct carrying the human tyrosinase gene (MVA-hTyr), we found that recognition by CTL against peptide 1-9 did not require TAP function as opposed to recognition by CTL against peptide 369-377. When target cells with intact processing and transport functions were infected with MVA-hTyr, lysis by CTL against peptide 1-9 was not impaired by lactacystin, a specific inhibitor for the proteasome, whereas lysis by CTL against peptide 369-377 was completely abrogated. Taken together, peptide 1-9 derived from the signal sequence of tyrosinase is presented in a TAP-independent fashion and does not require proteasomes for processing. Cellular immune responses against this hydrophobic peptide can be monitored with lymphokine spot assays as documented in the case of a patient with metastatic melanoma, in whom we observed a preferential T-cell response against tyrosinase peptide 1-9 subsequent to chemoimmunotherapy. Independence of cytosolic processing and transport pathways and potentially enhanced expression levels make signal sequence-derived peptides and their carrier proteins important candidates for specific immunotherapy.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Apresentação de Antígeno , Cisteína Endopeptidases/fisiologia , Melanoma/imunologia , Monofenol Mono-Oxigenase/imunologia , Complexos Multienzimáticos/fisiologia , Sinais Direcionadores de Proteínas , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Epitopos , Antígeno HLA-A2/fisiologia , Humanos , Complexo de Endopeptidases do Proteassoma , Linfócitos T Citotóxicos/imunologia , Células Tumorais Cultivadas
18.
Biotechniques ; 28(6): 1137-42, 1144-6, 1148, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10868279

RESUMO

Recombinant vaccinia viruses are extremely valuable tools for research in molecular biology and immunology. The extension of vaccinia vector technology to replication-deficient and safety-tested virus strains such as modified vaccinia virus Ankara (MVA) have made this versatile eukaryotic expression system even more attractive for basic and clinical research. Here, we report on easily obtaining recombinant MVA using stringent growth selection on rabbit kidney RK-13 cells. We describe the construction and use of new MVA vector plasmids that carry an expression cassette of the vaccinia virus host range gene, K1L, as a transient selectable marker. These plasmids allow either stable insertion of additional recombinant genes into the MVA genome or precisely targeted mutagenesis of MVA genomic sequences. Repetitive DNA sequences flanking the K1L gene were designed to remove the marker gene from the viral genome by homologous recombination under nonselective growth conditions. The convenience of this new selection technique is demonstrated by isolating MVA recombinants that produce green fluorescent protein and by generating MVA deletion mutants.


Assuntos
Engenharia Genética , Vaccinia virus/genética , Proteínas Virais/genética , Animais , Vetores Genéticos , Coelhos
19.
Cancer ; 88(7): 1650-62, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10738224

RESUMO

BACKGROUND: Papillomavirus is the etiologic agent associated with cervical carcinoma. The papilloma E2 protein is able to regulate negatively the expression of E6 and E7 papilloma oncoproteins. Therefore, a new, highly attenuated vaccinia virus known as modified vaccinia virus Ankara (MVA), which carries the papillomavirus E2 gene, was used for the treatment of tumors associated with human papillomavirus. METHODS: Analysis of expression of the E2 gene from the recombinant vaccinia virus was performed by reverse transcription-polymerase chain reaction of RNA isolated from infected cells. Detection of the E2 protein was done by immunoprecipitation from proteins labeled with [(35)S]-methionine, isolated from infected cells. The therapeutic effect of the MVA E2 recombinant virus over human tumors was tested in nude mice bearing tumors generated by inoculation of HeLa cells. Series of 10 nude mice with tumors of different sizes were injected with MVA, MVA E2, or phosphate-buffered saline. Tumor size was monitored every week to assess growth. RESULTS: The MVA E2 recombinant virus efficiently expressed the E2 protein in BS-C-1 cells. This protein was able to repress, in vivo, the papillomavirus P105 promoter, which controls the expression of the E6 and E7 oncoproteins. In nude mice the MVA E2 virus reduced tumor growth very efficiently. In contrast, tumors continued to grow in mice treated with MVA or PBS. The life expectancy of MVA E2-treated mice was also increased three- to fourfold compared with that of animals that received MVA or PBS. CONCLUSIONS: The growth of human tumors was efficiently inhibited by the MVA E2 recombinant vaccinia virus. The absence of side effects in treated animals suggested that the MVA E2 virus is a safe biologic agent that could in the future be used in humans for the treatment of cervical carcinoma.


Assuntos
Proteínas de Ligação a DNA/genética , Vaccinia virus/genética , Proteínas Virais/genética , Células 3T3 , Animais , Apoptose , Linhagem Celular , Embrião de Galinha , Intervalo Livre de Doença , Terapia Genética , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Nus , Modelos Genéticos , Transplante de Neoplasias , Plasmídeos , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , beta-Galactosidase/metabolismo
20.
Vaccine ; 19(9-10): 1180-7, 2000 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-11137255

RESUMO

Although current influenza vaccines have been shown to reduce influenza-related morbidity and mortality, there is a desire to develop more efficacious products. Vaccines which can induce CD8(+) cytotoxic T cell (CTL) responses in addition to strong antibody responses may be more effective in preventing disease since it has been demonstrated that CTL contribute to protective immunity, even against drift variants of influenza A viruses. The immunogenicity of two types of experimental influenza vaccines, which were based on immune stimulating complexes (ISCOM), were evaluated and compared with a conventional non-adjuvanted inactivated split virion vaccine, after immunization of human volunteers. In this randomized, double blind study, it was shown that the ISCOM vaccines altered the kinetics of the serum antibody response, resulting in more rapid titer rises against the vaccine strains. This accelerated antibody response coincided with enhanced in vitro proliferative T cell responses, which were observed shortly after vaccination. In addition, CTL responses were observed in a higher proportion of the vaccinees receiving an ISCOM vaccine, than in vaccinees receiving the conventional influenza vaccine.


Assuntos
Adjuvantes Imunológicos/farmacologia , Anticorpos Antivirais/biossíntese , ISCOMs/farmacologia , Vacinas contra Influenza/imunologia , Ativação Linfocitária , Linfócitos T Citotóxicos/imunologia , Adolescente , Adulto , Método Duplo-Cego , Humanos , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...