Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Science ; 292(5514): 69-74, 2001 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-11393868

RESUMO

Heterologous prime/boost regimens have the potential for raising high levels of immune responses. Here we report that DNA priming followed by a recombinant modified vaccinia Ankara (rMVA) booster controlled a highly pathogenic immunodeficiency virus challenge in a rhesus macaque model. Both the DNA and rMVA components of the vaccine expressed multiple immunodeficiency virus proteins. Two DNA inoculations at 0 and 8 weeks and a single rMVA booster at 24 weeks effectively controlled an intrarectal challenge administered 7 months after the booster. These findings provide hope that a relatively simple multiprotein DNA/MVA vaccine can help to control the acquired immune deficiency syndrome epidemic.


Assuntos
Vacinas contra a AIDS/imunologia , Síndrome da Imunodeficiência Adquirida/prevenção & controle , Vacinas de DNA/imunologia , Vacinas contra a AIDS/administração & dosagem , Síndrome da Imunodeficiência Adquirida/imunologia , Síndrome da Imunodeficiência Adquirida/virologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Centro Germinativo/imunologia , Anticorpos Anti-HIV/sangue , Anticorpos Anti-HIV/imunologia , HIV-1/genética , HIV-1/imunologia , HIV-1/fisiologia , Imunidade nas Mucosas , Imunização Secundária , Memória Imunológica , Interferon gama/biossíntese , Linfonodos/imunologia , Macaca mulatta , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Linfócitos T/imunologia , Vacinas de DNA/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vaccinia virus/imunologia , Carga Viral
2.
Vaccine ; 19(27): 3700-9, 2001 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-11395204

RESUMO

Modified vaccinia virus Ankara (MVA)-based recombinant viruses have been shown to be potent vaccine candidates for several infectious and neoplastic diseases. Since a major application of these live, replication-deficient vectors would be their use in immunocompromised or potentially immunocompromised individuals, a preclinical safety study was carried out. Macaques were inoculated with high doses of MVA (10(9)) via various routes, after immune-suppression by total-body irradiation, anti-thymocyte globulin treatment, or measles virus (MV) infection. No clinical, haematological or pathological abnormalities related to MVA inoculation were observed during a 13-day follow-up period. The presence of MVA genomes was demonstrated by nested PCR during the course of the experiment in all macaques, but from none of these animals replication competent MVA could be reisolated. These data suggest that MVA can safely be used as a basis for recombinant human vaccines, and that it is also safe for use in immunocompromised individuals.


Assuntos
Terapia de Imunossupressão , Macaca fascicularis/imunologia , Vaccinia virus/imunologia , Vacinas Virais/efeitos adversos , Animais , Anticorpos Antivirais/sangue , DNA Viral/isolamento & purificação , Feminino , Genoma Viral , Imunoglobulina G/sangue , Injeções Intradérmicas/efeitos adversos , Reação em Cadeia da Polimerase , Vaccinia virus/genética , Vaccinia virus/isolamento & purificação
3.
J Virol ; 75(11): 5151-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333896

RESUMO

Since cytotoxic T lymphocytes (CTLs) are critical for controlling human immunodeficiency virus type 1 (HIV-1) replication in infected individuals, candidate HIV-1 vaccines should elicit virus-specific CTL responses. In this report, we study the immune responses elicited in rhesus monkeys by a recombinant poxvirus vaccine and the degree of protection afforded against a pathogenic simian-human immunodeficiency virus SHIV-89.6P challenge. Immunization with recombinant modified vaccinia virus Ankara (MVA) vectors expressing SIVmac239 gag-pol and HIV-1 89.6 env elicited potent Gag-specific CTL responses but no detectable SHIV-specific neutralizing antibody (NAb) responses. Following intravenous SHIV-89.6P challenge, sham-vaccinated monkeys developed low-frequency CTL responses, low-titer NAb responses, rapid loss of CD4+ T lymphocytes, high-setpoint viral RNA levels, and significant clinical disease progression and death in half of the animals by day 168 postchallenge. In contrast, the recombinant MVA-vaccinated monkeys demonstrated high-frequency secondary CTL responses, high-titer secondary SHIV-89.6-specific NAb responses, rapid emergence of SHIV-89.6P-specific NAb responses, partial preservation of CD4+ T lymphocytes, reduced setpoint viral RNA levels, and no evidence of clinical disease or mortality by day 168 postchallenge. There was a statistically significant correlation between levels of vaccine-elicited CTL responses prior to challenge and the control of viremia following challenge. These results demonstrate that immune responses elicited by live recombinant vectors, although unable to provide sterilizing immunity, can control viremia and prevent disease progression following a highly pathogenic AIDS virus challenge.


Assuntos
HIV-1/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Vacinas Sintéticas/administração & dosagem , Vaccinia virus/genética , Animais , Anticorpos Antivirais/análise , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/imunologia , Progressão da Doença , Produtos do Gene env/imunologia , Produtos do Gene gag/imunologia , Produtos do Gene pol/imunologia , HIV-1/genética , Humanos , Macaca mulatta , RNA Viral/sangue , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Linfócitos T Citotóxicos/imunologia , Fatores de Tempo , Vacinas Sintéticas/imunologia , Vaccinia virus/imunologia
4.
J Virol ; 75(5): 2462-7, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11160750

RESUMO

Increasing evidence suggests that the generation of cytotoxic T-lymphocyte (CTL) responses specific for a diversity of viral epitopes will be needed for an effective human immunodeficiency virus type 1 (HIV-1) vaccine. Here, we determine the frequencies of CTL responses specific for the simian immunodeficiency virus Gag p11C and HIV-1 Env p41A epitopes in simian-human immunodeficiency virus (SHIV)-infected and vaccinated rhesus monkeys. The p11C-specific CTL response was high frequency and dominant and the p41A-specific CTL response was low frequency and subdominant in both SHIV-infected monkeys and in monkeys vaccinated with recombinant modified vaccinia virus Ankara vectors expressing these viral antigens. Interestingly, we found that plasmid DNA vaccination led to high-frequency CTL responses specific for both of these epitopes. These data demonstrate that plasmid DNA may be useful in eliciting a broad CTL response against multiple epitopes.


Assuntos
Vacinas contra a AIDS/imunologia , Epitopos de Linfócito T/imunologia , Vacinas contra a SAIDS/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/imunologia , Animais , Produtos do Gene gag/genética , Produtos do Gene gag/imunologia , Proteína gp41 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Humanos , Epitopos Imunodominantes/imunologia , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Vacinação
5.
Curr Protoc Protein Sci ; Chapter 5: Unit5.12, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18429178

RESUMO

This unit describes the maintenance of cell lines used with vaccinia virus, both in monolayer cultures and in suspension. The suspended cell culture is then used in the preparation of vaccinia virus stocks. The preparation of chick embryo fibroblasts (CEF) is also presented for use in the production of the highly attenuated and host range-restricted modified vaccinia virus Ankara (MVA) strain of vaccinia virus. Additionally, support protocols are presented for the titration of standard and MVA vaccinia virus stocks.


Assuntos
Células Cultivadas/virologia , Vaccinia virus/crescimento & desenvolvimento , Animais , Técnicas de Cultura de Células/métodos , Embrião de Galinha , Fibroblastos/citologia , Fibroblastos/virologia
6.
Curr Protoc Protein Sci ; Chapter 5: Unit5.13, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18429179

RESUMO

This unit first describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector to generate a recombinant virus. Methods are also presented for purifying vaccinia virus and for isolating viral DNA, which can be used during transfection. Also presented are selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented.


Assuntos
Recombinação Genética/genética , Vaccinia virus/genética , Animais , DNA Viral/genética , Vetores Genéticos/genética , Humanos , Modelos Genéticos , Vaccinia virus/crescimento & desenvolvimento , Replicação Viral
7.
Curr Protoc Mol Biol ; Chapter 16: Unit16.16, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18265123

RESUMO

This unit describes the maintenance of cell lines used with vaccinia virus, both in monolayer cultures and in suspension. The suspended cell culture is then used in the preparation of vaccinia virus stocks. The preparation of chick embryo fibroblasts (CEF) is also presented for use in the production of the highly attenuated and host range-restricted modified vaccinia virus Ankara (MVA) strain of vaccinia virus. Additionally, support protocols are presented for the titration of standard and MVA vaccinia virus stocks.


Assuntos
Vaccinia virus/crescimento & desenvolvimento , Animais , Técnicas de Cultura de Células , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Fibroblastos , Células HeLa , Humanos , Ensaio de Placa Viral
8.
Curr Protoc Mol Biol ; Chapter 16: Unit16.17, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18265124

RESUMO

This unit first describes how to infect cells with vaccinia virus and then transfect them with a plasmid-transfer vector to generate a recombinant virus. Methods are also presented for purifying vaccinia virus and for isolating viral DNA, which can be used during transfection. Also presented are selection and screening methods used to isolate recombinant viruses and a method for the amplification of recombinant viruses. Finally, a method for live immunostaining that has been used primarily for detection of recombinant modified vaccinia virus Ankara (MVA) is presented.


Assuntos
Vaccinia virus/genética , Animais , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Concanavalina A , DNA Viral/genética , DNA Viral/isolamento & purificação , Vetores Genéticos , Humanos , Recombinação Genética , Transfecção , Vaccinia virus/isolamento & purificação
9.
J Mol Endocrinol ; 24(3): 419-32, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10828835

RESUMO

Insulin-stimulated signaling pathways are activated upon interactions between the intracellular domains of the receptor and its downstream effectors. Insulin receptor substrate proteins (IRS-1, -2, -3 and -4) are the best-studied substrates for the insulin receptor kinase (IRK). We have previously shown that IRS-1 and IRS-2 interact with the juxtamembrane (JM) but not with the carboxyl-terminal (CT) region of the insulin receptor (IR) in vitro. However, the precise role of these IR regions in mediating insulin's bioeffects is still unresolved. In the present work we made use of vaccinia virus as a vector for quantitative expression of the JM and CT domains within the cytoplasm of physiologically insulin-responsive primary rat adipocytes and rat hepatoma Fao cells. We could demonstrate that overexpression of either the JM or the CT domains did not inhibit either insulin binding or insulin-stimulated receptor autophosphorylation. In contrast, metabolic effects such as insulin-induced glucose utilization in adipocytes, and insulin-induced amino acid utilization in Fao hepatoma cells were inhibited (70-80%) in cells overexpressing the JM but not the CT domains of IR. The inhibitory effects of the overexpressed JM domain were accompanied by inhibition of insulin-stimulated IRS-1 phosphorylation, decreased IRS-1-associated PI3K activity, and decreased phosphorylation of the downstream effectors of PI3K, PKB and p70 S6K. Insulin-stimulated thymidine incorporation in Fao cells was also inhibited (40%) upon overexpression of the JM but not the CT region of IR. Our findings suggest that interactions between the JM region of IR and its downstream effectors are obligatory for insulin-stimulated metabolic functions in physiologically relevant insulin responsive cells. They also rule out the possibility that interaction of proteins, including PI3K, with the CT domain can provide an alternative pathway.


Assuntos
Adipócitos/metabolismo , Insulina/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Proteínas Serina-Treonina Quinases , Receptor de Insulina/metabolismo , Adipócitos/enzimologia , Animais , Células Cultivadas , Replicação do DNA , Ativação Enzimática , Vetores Genéticos , Glucose/metabolismo , Proteínas Substratos do Receptor de Insulina , Neoplasias Hepáticas Experimentais/patologia , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Wistar , Receptor de Insulina/química , Proteínas Quinases S6 Ribossômicas/metabolismo , Células Tumorais Cultivadas , Vaccinia virus/genética
10.
J Virol ; 74(9): 4236-43, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10756037

RESUMO

Recombinant modified vaccinia virus Ankara (MVA), encoding the measles virus (MV) fusion (F) and hemagglutinin (H) (MVA-FH) glycoproteins, was evaluated in an MV vaccination-challenge model with macaques. Animals were vaccinated twice in the absence or presence of passively transferred MV-neutralizing macaque antibodies and challenged 1 year later intratracheally with wild-type MV. After the second vaccination with MVA-FH, all the animals developed MV-neutralizing antibodies and MV-specific T-cell responses. Although MVA-FH was slightly less effective in inducing MV-neutralizing antibodies in the absence of passively transferred antibodies than the currently used live attenuated vaccine, it proved to be more effective in the presence of such antibodies. All vaccinated animals were effectively protected from the challenge infection. These data suggest that MVA-FH should be further tested as an alternative to the current vaccine for infants with maternally acquired MV-neutralizing antibodies and for adults with waning vaccine-induced immunity.


Assuntos
Hemaglutininas Virais/imunologia , Vacina contra Sarampo/imunologia , Sarampo/prevenção & controle , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Antivirais/imunologia , Feminino , Expressão Gênica , Vetores Genéticos , Hemaglutininas Virais/genética , Macaca fascicularis , Sarampo/imunologia , Vírus do Sarampo/imunologia , Linfócitos T/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vaccinia virus , Proteínas Virais de Fusão/genética
11.
Vaccine ; 18(5-6): 392-7, 1999 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-10519927

RESUMO

Intranasal and intramuscular immunizations of mice with the highly attenuated MVA strain of vaccinia virus expressing the respiratory syncytial virus (RSV) F or G glycoprotein induced higher RSV antibody titers than those achieved by infection with RSV and greatly restricted the replication of RS challenge virus in both the upper and lower respiratory tracts. In addition, a recombinant MVA expressing both RSV F and G was stable and was as immunogenic as a combination of two single recombinant viruses. The levels of antibodies to RSV F and G, induced by previous intranasal infection with attenuated RSV, were boosted by intramuscular immunization with recombinant MVA. These data support further development of recombinant MVA as a RSV vaccine.


Assuntos
Vírus Defeituosos/genética , Proteína HN , Vírus Sinciciais Respiratórios/imunologia , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Camundongos , Camundongos Endogâmicos BALB C , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral
12.
J Infect Dis ; 179(6): 1345-51, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10228053

RESUMO

Two parainfluenza virus type 3 (PIV3) vaccine candidates-cp45, a live attenuated derivative of the JS wild type (wt), and a replication-defective vaccinia virus recombinant expressing the hemagglutinin-neuraminidase or fusion glycoprotein of human PIV3 (modified vaccinia virus Ankara [MVA]/PIV3 recombinants)-were evaluated in rhesus monkeys to determine whether successful immunization could be achieved in the presence of passively transferred PIV3 antibodies. The cp45 virus, administered intranasally (in) and intratracheally (it) in the presence of high levels of PIV3 antibodies, replicated efficiently in the nasopharynx and protected against challenge with wt human PIV3. The MVA recombinants, administered in, it, and intramuscularly in the absence of passive antibody, conferred protection against replication of PIV3 wt challenge virus, but this was largely abrogated when immunization occurred in the presence of passive antibodies. Because immunization for the prevention of HPIV3 disease must occur in early infancy when maternal antibodies are present, the live attenuated cp45 virus continues to be a promising vaccine candidate for this age group.


Assuntos
Antígenos Virais/uso terapêutico , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/prevenção & controle , Vacinação , Vacinas Virais/uso terapêutico , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/uso terapêutico , Imunização Passiva , Macaca mulatta , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Sistema Respiratório/virologia , Vacinas Sintéticas/uso terapêutico , Replicação Viral
13.
Vaccine ; 17(3): 261-8, 1999 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-9987162

RESUMO

Genes encoding the glycosylated precursor of the membrane (prM) and envelope (E) proteins of a Korean strain of Japanese encephalitis virus (JEV) were inserted into the genome of the host-range restricted, highly attenuated, and safety-tested MVA strain of vaccinia virus. MVA recombinants containing the JEV genes, under strong synthetic or modified H5 vaccinia virus promoters, were isolated. Synthesis of JEV prM and E proteins was detected by immunofluorescence microscopy, flow cytometry, and polyacrylamide gel electrophoresis. Mice inoculated and boosted by various routes with either of the MVA recombinants produced JEV neutralizing antibodies, that had titres comparable with those induced by an inactivated JEV vaccine, as well as haemagglutination-inhibiting antibodies. Mice immunized with 2 x 10(6) infectious units of MVA/JEV recombinants by intramuscular or intraperitoneal routes were completely protected against a 10(5) LD50 JEV challenge at 9 weeks of age.


Assuntos
Encefalite Japonesa/prevenção & controle , Regulação Viral da Expressão Gênica/fisiologia , Imunização , Vaccinia virus/imunologia , Vacinas Virais , Animais , Camundongos , Camundongos Endogâmicos ICR , Recombinação Genética , Especificidade da Espécie , Vacinas Atenuadas , Vaccinia virus/genética , Proteínas do Envelope Viral/genética , Proteínas da Matriz Viral/genética
14.
Virology ; 251(2): 334-42, 1998 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-9837798

RESUMO

The severely attenuated and host range (HR) restricted modified vaccinia virus Ankara (MVA) was derived by >500 passages in chick embryo fibroblasts, during which multiple deletions and mutations occurred. To determine the basis of the HR defect, we prepared cosmids from the parental vaccinia virus Ankara genome and transfected them into nonpermissive monkey BS-C-1 cells that had been infected with MVA. Recombinant viruses that formed macroscopic plaques were detected after transfections with DNA segments that mapped near the left end of the viral genome. Plaque-forming viruses, generated by transfections with four individual cosmids and one pair of minimally overlapping cosmids, were purified, and their HRs were evaluated in BS-C-1 cells, rabbit RK-13 cells, and human HeLa, MRC-5, and A549 cells. The acquisition of the K1L and SPI-1 HR genes and the repair of large deletions were determined by polymerase chain reaction or pulse-field gel electrophoresis of NotI restriction enzyme digests of genomic DNA. The following results indicated the presence of previously unrecognized vaccinia virus HR genes: (1) the major mutations that restrict HR are within the left end of the MVA genome because the phenotypes of some recombinants approached that of the parental virus, (2) acquisition of the K1L gene correlated with the ability of recombinant viruses to propagate in RK-13 cells but did not enhance replication in human or monkey cell lines, (3) acquisition of the SPI-1 gene correlated with virus propagation in A549 cells but not with the extent of virus spread in monkey or other human cell lines, (4) there are at least two impaired HR genes because rescue occurred with nonoverlapping or minimally overlapping cosmids and recombinant viruses with intermediate HRs were isolated, and (5) at least one of the new HR genes did not map within any of the major deletions because the size of the left terminal NotI fragment was not appreciably altered in some recombinant viruses.


Assuntos
Vaccinia virus/genética , Vaccinia virus/fisiologia , Vacinas Virais , Replicação Viral , Animais , Embrião de Galinha , Cosmídeos , DNA Viral/química , Eletroforese em Gel de Campo Pulsado , Marcadores Genéticos , Haplorrinos , Células HeLa , Humanos , Reação em Cadeia da Polimerase , Coelhos , Mapeamento por Restrição , Vaccinia virus/metabolismo
15.
J Virol ; 72(10): 8264-72, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9733870

RESUMO

To improve the safety of recombinant vaccinia virus vaccines, modified vaccinia virus Ankara (MVA) has been employed, because it has a replication defect in most mammalian cells. Here we apply MVA to human immunodeficiency virus type 1 (HIV-1) vaccine development by incorporating the envelope protein gp160 of HIV-1 primary isolate strain 89.6 (MVA 89.6) and use it to induce mucosal cytotoxic-T-lymphocyte (CTL) immunity. In initial studies to define a dominant CTL epitope for HIV-1 89.6 gp160, we mapped the epitope to a sequence, IGPGRAFYAR (from the V3 loop), homologous to that recognized by HIV MN loop-specific CTL and showed that HIV-1 MN-specific CTLs cross-reactively recognize the corresponding epitope from strain 89.6 presented by H-2Dd. Having defined the CTL specificity, we immunized BALB/c mice intrarectally with recombinant MVA 89.6. A single mucosal immunization with MVA 89.6 was able to elicit long-lasting antigen-specific mucosal (Peyer's patch and lamina propria) and systemic (spleen) CTL responses as effective as or more effective than those of a replication-competent vaccinia virus expressing 89.6 gp160. Immunization with MVA 89.6 led to (i) the loading of antigen-presenting cells in vivo, as measured by the ex vivo active presentation of the P18-89.6 peptide to an antigen-specific CTL line, and (ii) the significant production of the proinflammatory cytokines (interleukin-6 and tumor necrosis factor alpha) in the mucosal sites. These results indicate that nonreplicating recombinant MVA may be at least as effective for mucosal immunization as replicating recombinant vaccinia virus.


Assuntos
Vacinas contra a AIDS/administração & dosagem , Proteína gp160 do Envelope de HIV/genética , Imunidade nas Mucosas , Linfócitos T Citotóxicos/imunologia , Vacinas Sintéticas/administração & dosagem , Vaccinia virus/genética , Vacinas contra a AIDS/imunologia , Animais , Linhagem Celular , Reações Cruzadas , Vírus Defeituosos/genética , Feminino , Memória Imunológica , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Reto , Fator de Necrose Tumoral alfa/biossíntese , Vacinas Sintéticas/imunologia
16.
Proc Natl Acad Sci U S A ; 95(17): 10112-6, 1998 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-9707609

RESUMO

The utility of modified vaccinia virus Ankara (MVA) as a vector for eliciting AIDS virus-specific cytotoxic T lymphocytes (CTL) was explored in the simian immunodeficiency virus (SIV)/rhesus monkey model. After two intramuscular immunizations with recombinant MVA-SIVSM gag pol, the monkeys developed a Gag epitope-specific CTL response readily detected in peripheral blood lymphocytes by using a functional killing assay. Moreover, those immunizations also elicited a population of CD8+ T lymphocytes in the peripheral blood that bound a specific major histocompatibility complex class I/peptide tetramer. These Gag epitope-specific CD8+ T lymphocytes also were demonstrated by using both functional and tetramer-binding assays in lymph nodes of the immunized monkeys. These observations suggest that MVA may prove a useful vector for an HIV-1 vaccine. They also suggest that tetramer staining may be a useful technology for monitoring CTL generation in vaccine trials in nonhuman primates and in humans.


Assuntos
Proteínas de Fusão gag-pol/imunologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Linfócitos T Citotóxicos/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Vacinas contra a AIDS/isolamento & purificação , Animais , Sequência de Bases , Primers do DNA/genética , Epitopos , Proteínas de Fusão gag-pol/genética , Vetores Genéticos , Antígenos de Histocompatibilidade Classe I/química , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imunização , Macaca mulatta , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
17.
Vaccine ; 16(13): 1324-30, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9682397

RESUMO

Immunization of rhesus monkeys with modified vaccinia Ankara (MVA) recombinants expressing the haemagglutinin-neuraminidase (HN) or fusion (F) glycoproteins of human parainfluenza virus type 3 (HPIV3) was compared with an intranasally-administered live, attenuated HPIV3 vaccine candidate, the cp45 derivative of the JS strain of wildtype HPIV3. The MVA recombinants, when given parenterally (i.m.) or as a parenteral-local (i.m. and i.t.) combination, induced an antibody response comparable to that of cp45 and protected the upper and lower respiratory tracts of the rhesus monkeys against challenge with wildtype HPIV3. When given by the i.n. route alone, the MVA/PIV3 recombinants induced a serum antibody response that was comparable to that of cp45 and induced resistance in the lower respiratory tract. Despite the ability of the intranasally-administered MVA/PIV3 recombinants to stimulate a good serological response and to protect the lower respiratory tract, they unexpectedly failed to induce a significant level of resistance in the upper respiratory tract. The live, attenuated virus vaccine candidate induced almost complete resistance in both the upper and lower tracts. The data thus identify two vaccine candidates that can protect both the upper and lower respiratory tracts of rhesus monkey, parenterally-administered MVA/PIV3 and intranasally-administered cp45. Further studies with these vaccines in non-human primates and humans should identify the relative merits of these immunogens for use in the very young infant.


Assuntos
Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/prevenção & controle , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/biossíntese , Embrião de Galinha , Proteína HN/imunologia , Injeções Intramusculares , Macaca mulatta , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/normas , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/normas , Vaccinia virus/imunologia , Proteínas Virais de Fusão/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/normas
18.
Vaccine ; 14(15): 1451-8, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8994321

RESUMO

The highly attenuated, replication-deficient, modified vaccinia virus Ankara (MVA) was used to express the fusion (F) and/or hemagglutinin-neuraminidase (HN) glycoproteins of parainfluenza virus 3 (PIV3). Initial recombinant viruses in which the HN gene was regulated by a very strong synthetic earlyllate promoter replicated poorly in permissive chick embryo cells evidently due to toxic levels of the gene product. This result led us to construct and evaluate a modified earlyllate promoter derived from the H5 gene of vaccinia virus. Reporter gene experiments indicated that the enhanced H5 promoter was about five times stronger than the 7.5 promoter used in previous recombinant vaccinia/ PIV3 viruses. Although the overall expression from the modified H5 promoter was less than that of the strong synthetic promoter, early expression, determined in the presence of an inhibitor of DNA replication, was higher. Importantly, recombinant MVA employing the modified H5 promoter to regulate the F or HN gene of PIV3 replicated to high titers in chick cells and expressed functional F or HN proteins as measured by syncytial formation upon dual infection of mammalian cells. Cotton rats inoculated with recombinant MVA expressing F or HN by intramuscular or intranasal routes produced high levels of antibody. The virus expressing HN, however, was the more effective of the two in inducing immunity to PIV3 challenge, reducing PIV3 viral titers in the nasal turbinates by at least 4.7 logs and in the lungs by 3.4 logs, similar to that achieved by immunization with PIV3. These studies support further testing of recombinant MVA/PIV3 viruses as safe and effective candidate vaccines.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/prevenção & controle , Vacinas Sintéticas/administração & dosagem , Vaccinia virus/imunologia , Vacinas Virais/administração & dosagem , Animais , Células HeLa , Humanos , Sigmodontinae , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Vacinas Virais/imunologia , Replicação Viral/genética
19.
Proc Natl Acad Sci U S A ; 93(18): 9788-92, 1996 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-8790409

RESUMO

Human herpesviruses 6 and 7 (HHV-6 and HHV-7) are prevalent lymphotropic viruses that infect more than 80% of children at infancy or during early childhood. Infection ranges from asymptomatic to severe disease. HHV-6B causes exanthem subitum. The virus can be recovered from peripheral blood mononuclear cells during the acute phase of exanthem subitum, but the host remains latently infected throughout life. In immunocompromised patients undergoing kidney, liver, or bone marrow transplantation latent HHV-6B is reactivated, at times causing severe or fatal disease. Here, we describe the establishment of an in vitro system for reactivation of HHV-6B and HHV-7 from latency. HHV-7 is reactivated from latently infected peripheral blood mononuclear cells by T-cell activation. HHV-6B could not be reactivated under similar conditions; however, the latent HHV-6B could be recovered after the cells were infected with HHV-7. Once reactivated, the HHV-6B genomes became prominent and the HHV-7 disappeared. We conclude that HHV-7 can provide a transacting function(s) mediating HHV-6 reactivating from latency. Understanding the activation process is critical for the development of treatments to control the activation of latent viruses so as to avoid these sometimes life threatening infections in transplant recipients.


Assuntos
Herpesvirus Humano 6/crescimento & desenvolvimento , Herpesvirus Humano 7/crescimento & desenvolvimento , Ativação Viral , Eletroforese em Gel de Poliacrilamida , Humanos , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Latência Viral
20.
J Virol ; 70(9): 6418-24, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8709274

RESUMO

Mice immunized with two intragastrically administered doses of a replication-deficient recombinant vaccinia virus containing the hemagglutinin and nucleoprotein genes from H1N1 influenza virus developed serum anti-H1 immunoglobulin G (IgG) antibody that completely protected the lungs from challenge with H1N1. Almost all of the mice given two intragastric doses also developed mucosal anti-H1 IgA antibody, and those with high anti-H1 IgA titers had completely protected noses. Intramuscular injection of the vaccine protected the lungs but not the noses from challenge. We also found that the vaccine enhanced recovery from infection caused by a shifted (H3N2) influenza virus, probably through the induction of nucleoprotein-specific cytotoxic T-lymphocyte activity. A replication-deficient, orally administered, enteric-coated, vaccinia virus-vectored vaccine might safely protect humans against influenza.


Assuntos
Proteína HN/imunologia , Hemaglutininas Virais/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas Sintéticas , Vaccinia virus/imunologia , Administração Oral , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Feminino , Genes Virais , Proteína HN/biossíntese , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas Virais/biossíntese , Imunoglobulina A/biossíntese , Imunoglobulina A/sangue , Vírus da Influenza A/isolamento & purificação , Vacinas contra Influenza/administração & dosagem , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Vacinas Sintéticas/administração & dosagem , Vaccinia virus/fisiologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA