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










Base de dados
Intervalo de ano de publicação
1.
J Pediatric Infect Dis Soc ; 4(4): e143-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26582883

RESUMO

We conducted a phase I clinical trial (clinicaltrials.gov identifier, NCT00641017) of the experimental live-attenuated human parainfluenza virus type 1 (HPIV-1) vaccine rHPIV-1/84/del 170/942A sequentially in 3 groups: adults, HPIV-1-seropositive children, and HPIV-1-seronegative children, the target population for vaccination. rHPIV-1/84/del 170/942A was appropriately restricted in replication in adults and HPIV-1-seropositive children but was overattenuated (ie, insufficiently infectious and immunogenic) for HPIV-1-seronegative children.


Assuntos
Vacinas contra Parainfluenza/uso terapêutico , Infecções por Paramyxoviridae/prevenção & controle , Adulto , Anticorpos Antivirais/sangue , Pré-Escolar , Método Duplo-Cego , Humanos , Lactente , Vírus da Parainfluenza 1 Humana , Infecções por Paramyxoviridae/epidemiologia , Vacinas Atenuadas/uso terapêutico
2.
Blood ; 125(10): 1601-10, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25568348

RESUMO

The treatment of non-Hodgkin lymphomas has benefited enormously from the introduction of monoclonal antibody-based therapies. However, the efficacy of these treatments varies with lymphoma subtypes and typically decreases with subsequent relapses. Here, we report on antigen-armed antibodies (AgAbs) as a potential treatment of B-cell lymphoma. AgAbs include antigens from ubiquitous pathogens, such as Epstein-Barr virus (EBV), that persist in their host and elicit strong lifelong T-cell responses. They act as vectors by introducing antigen directly into tumor cells to induce an antigen-specific CD4(+) T-cell response against these cells. We have fused antibodies targeting human B-cell surface receptors (CD19-22) to immunodominant T-cell antigens from EBV proteins, including EBNA1, EBNA3B, and EBNA3C. Exposure of EBV-transformed B cells and of Burkitt lymphoma cells to AgAbs led to antigen presentation, T-cell recognition, and target cell killing. The efficiency of AgAb action paralleled the abundance of the targeted molecules on lymphoma cells as well as their HLA class II expression levels. AgAbs can also induce activation and proliferation of EBV-specific memory CD4(+) T cells ex vivo. These studies show the potential of AgAbs as an effective therapeutic strategy against B-cell lymphomas.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Anticorpos Antineoplásicos/genética , Apresentação de Antígeno , Linfócitos B/imunologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Epitopos de Linfócito T , Herpesvirus Humano 4/imunologia , Humanos , Memória Imunológica , Ativação Linfocitária , Receptores de Antígenos de Linfócitos B/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Linfócitos T Citotóxicos/imunologia
3.
Virology ; 433(2): 320-8, 2012 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22959894

RESUMO

Human parainfluenza viruses (PIVs) cause acute respiratory illness in children, the elderly, and immunocompromised patients. PIV3 is a common cause of bronchiolitis and pneumonia, whereas PIV1 and 2 are frequent causes of upper respiratory tract illness and croup. To assess how PIV1, 2, and 3 differ with regard to replication and induction of type I interferons, interleukin-6, and relevant chemokines, we infected primary human airway epithelium (HAE) cultures from the same tissue donors and examined replication kinetics and cytokine secretion. PIV1 replicated to high titer yet did not induce cytokine secretion until late in infection, while PIV2 replicated less efficiently but induced an early cytokine peak. PIV3 replicated to high titer but induced a slower rise in cytokine secretion. The T cell chemoattractants CXCL10 and CXCL11 were the most abundant chemokines induced. Differences in replication and cytokine secretion might explain some of the differences in PIV serotype-specific pathogenesis and epidemiology.


Assuntos
Brônquios/imunologia , Brônquios/virologia , Vírus da Parainfluenza 1 Humana/fisiologia , Vírus da Parainfluenza 2 Humana/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Traqueia/imunologia , Traqueia/virologia , Células Cultivadas , Quimiocinas/biossíntese , Citocinas/biossíntese , Citocinas/genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interleucina-6/biossíntese , Interleucina-6/genética , Cinética , Vírus da Parainfluenza 1 Humana/classificação , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 1 Humana/patogenicidade , Vírus da Parainfluenza 2 Humana/classificação , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 2 Humana/patogenicidade , Vírus da Parainfluenza 3 Humana/classificação , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Polimorfismo de Nucleotídeo Único , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Sorotipagem , Especificidade da Espécie , Replicação Viral
4.
Expert Rev Respir Med ; 5(4): 515-26, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21859271

RESUMO

In children under 5 years of age, human parainfluenza viruses (HPIVs) as a group are the second most common etiology of acute respiratory illness leading to hospitalization, surpassed only by respiratory syncytial virus but ahead of influenza viruses. Using reverse genetics systems for HPIV serotypes 1, 2 and 3 (HPIV1, 2 and 3), several live-attenuated HPIVs have been generated and evaluated as intranasal vaccines in adults and in children. Two vaccines against HPIV3 were found to be well tolerated, infectious and immunogenic in Phase I trials in HPIV3-seronegative infants and children and should progress to proof-of-concept trials. Vaccines against HPIV1 and HPIV2 are less advanced and have just entered pediatric trials.


Assuntos
Desenho de Fármacos , Vacinas contra Parainfluenza/administração & dosagem , Infecções por Respirovirus/prevenção & controle , Respirovirus/imunologia , Doença Aguda , Administração Intranasal , Aerossóis , Pré-Escolar , Humanos , Lactente , Vacinas contra Parainfluenza/química , Vacinas contra Parainfluenza/genética , Respirovirus/genética , Infecções por Respirovirus/epidemiologia , Infecções por Respirovirus/imunologia , Resultado do Tratamento , Vacinas Atenuadas/administração & dosagem
5.
Vaccine ; 28(3): 767-79, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19857454

RESUMO

A novel recombinant human parainfluenza virus type 1 (rHPIV1), rHPIV1-C+P, in which the overlapping open reading frames of the C and P genes were separated in order to introduce mutations into the C gene without affecting P, was generated. Infectious rHPIV1-C+P was readily recovered and replicated as efficiently as HPIV1 wild type (wt) in vitro and in African green monkeys (AGMs). rHPIV1-C+P expressed increased levels of C protein and, surprisingly, activated the type I IFN and apoptosis responses more strongly than HPIV1 wt. rHPIV1-C+P provided a useful backbone for recovering an attenuated P/C gene mutation (Delta 84-85), which was previously unrecoverable, likely due to detrimental effects of the deletion on the P protein. rHPIV1-C(Delta 84-85)+P and an additional mutant, rHPIV1-C(Delta 169-170)+P, were found to replicate to similar titers in vitro and to activate the type I IFN and apoptosis responses to a similar degree as rHPIV1-C+P. rHPIV1-C(Delta 84-85)+P was found to be highly attenuated in AGMs, and all viruses were immunogenic and effective in protecting AGMs against challenge with HPIV1 wt. rHPIV1-C(Delta 84-85)+P will be investigated as a potential live-attenuated vaccine candidate for HPIV1.


Assuntos
Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 1 Humana/patogenicidade , Fosfoproteínas/genética , Deleção de Sequência , Proteínas Virais/genética , Animais , Anticorpos Antivirais/sangue , Apoptose , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Humanos , Interferon Tipo I/biossíntese , Dados de Sequência Molecular , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 1 Humana/genética , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Carga Viral , Virulência , Replicação Viral
6.
Herpesviridae ; 1(1): 6, 2010 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-21429237

RESUMO

Genetic mutant organisms pervade all areas of Biology. Early on, herpesviruses (HV) were found to be amenable to genetic analysis using homologous recombination techniques in eukaryotic cells. More recently, HV genomes cloned onto a bacterial artificial chromosome (BAC) have become available. HV BACs can be easily modified in E.coli and reintroduced in eukaryotic cells to produce infectious viruses. Mutants derived from HV BACs have been used both to understand the functions of all types of genetic elements present on the virus genome, but also to generate mutants with potentially medically relevant properties such as preventative vaccines. Here we retrace the development of the BAC technology applied to the Epstein-Barr virus (EBV) and review the strategies available for the construction of mutants. We expand on the appropriate controls required for proper use of the EBV BACs, and on the technical hurdles researchers face in working with these recombinants. We then discuss how further technological developments might successfully overcome these difficulties. Finally, we catalog the EBV BAC mutants that are currently available and illustrate their contributions to the field using a few representative examples.

7.
J Virol ; 83(4): 1892-910, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19052086

RESUMO

Human parainfluenza virus type 1 (HPIV1) is an important respiratory pathogen in children and the most common cause of viral croup. We performed a microarray-based analysis of gene expression kinetics to examine how wild-type (wt) HPIV1 infection altered gene expression in human respiratory epithelial cells and what role beta interferon played in this response. We similarly evaluated HPIV1-P(C-), a highly attenuated and apoptosis-inducing virus that does not express any of the four C proteins, and HPIV1-C(F170S), a less attenuated mutant that contains a single point mutation in C and, like wt HPIV1, does not efficiently induce apoptosis, to examine the role of the C proteins in controlling host gene expression. We also used these data to investigate whether the phenotypic differences between the two C mutants could be explained at the transcriptional level. Mutation or deletion of the C proteins of HPIV1 permitted the activation of over 2,000 cellular genes that otherwise would be repressed by HPIV1 infection. Thus, the C proteins profoundly suppress the response of human respiratory cells to HPIV1 infection. Cellular pathways targeted by the HPIV1 C proteins were identified and their transcriptional control was analyzed using bioinformatics. Transcription factor binding sites for IRF and NF-kappaB were overrepresented in some of the C protein-targeted pathways, but other pathways were dominated by less-known factors, such as forkhead transcription factor FOXD1. Surprisingly, the host responses to the P(C-) and C(F170S) mutants were very similar, and only subtle differences in the expression kinetics of caspase 3 and TRAIL receptor 2 were observed. Thus, changes in host cell transcription did not reflect the striking phenotypic differences observed between these two viruses.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Parainfluenza 1 Humana/fisiologia , Transcrição Gênica , Proteínas Virais/metabolismo , Linhagem Celular , Células Epiteliais/virologia , Deleção de Genes , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Vírus da Parainfluenza 1 Humana/genética , Mutação Puntual , Mucosa Respiratória/virologia , Proteínas Virais/genética
8.
J Virol ; 82(18): 8965-77, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18614629

RESUMO

Recombinant human parainfluenza virus type 1 (rHPIV1) was modified to create rHPIV1-P(C-), a virus in which expression of the C proteins (C', C, Y1, and Y2) was silenced without affecting the amino acid sequence of the P protein. Infectious rHPIV1-P(C-) was readily recovered from cDNA, indicating that the four C proteins were not essential for virus replication. Early during infection in vitro, rHPIV1-P(C-) replicated as efficiently as wild-type (wt) HPIV1, but its titer subsequently decreased coincident with the onset of an extensive cytopathic effect not observed with wt rHPIV1. rHPIV1-P(C-) infection, but not wt rHPIV1 infection, induced caspase 3 activation and nuclear fragmentation in LLC-MK2 cells, identifying the HPIV1 C proteins as inhibitors of apoptosis. In contrast to wt rHPIV1, rHPIV1-P(C-) and rHPIV1-C(F170S), a mutant encoding an F170S substitution in C, induced interferon (IFN) and did not inhibit IFN signaling in vitro. However, only rHPIV1-P(C-) induced apoptosis. Thus, the anti-IFN and antiapoptosis activities of HPIV1 were separable: both activities are disabled in rHPIV1-P(C-), whereas only the anti-IFN activity is disabled in rHPIV1-C(F170S). In African green monkeys (AGMs), rHPIV1-P(C-) was considerably more attenuated than rHPIV1-C(F170S), suggesting that disabling the anti-IFN and antiapoptotic activities of HPIV1 had additive effects on attenuation in vivo. Although rHPIV1-P(C-) protected against challenge with wt HPIV1, its highly restricted replication in AGMs and in primary human airway epithelial cell cultures suggests that it might be overattenuated for use as a vaccine. Thus, the C proteins of HPIV1 are nonessential but have anti-IFN and antiapoptosis activities required for virulence in primates.


Assuntos
Apoptose/efeitos dos fármacos , Interferons/antagonistas & inibidores , Vírus da Parainfluenza 1 Humana/fisiologia , Vírus da Parainfluenza 1 Humana/patogenicidade , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Animais , Sequência de Bases , Caspase 3/metabolismo , Linhagem Celular , Chlorocebus aethiops , Ativação Enzimática , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 1 Humana/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Recombinação Genética , Células Vero , Proteínas Virais/genética
9.
J Virol ; 82(16): 8059-70, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18524813

RESUMO

Human parainfluenza virus type 1 (HPIV1) is a significant cause of pediatric respiratory disease in the upper and lower airways. An in vitro model of human ciliated airway epithelium (HAE), a useful tool for studying respiratory virus-host interactions, was used in this study to show that HPIV1 selectively infects ciliated cells within the HAE and that progeny virus is released from the apical surface with little apparent gross cytopathology. In HAE, type I interferon (IFN) is induced following infection with an HPIV1 mutant expressing defective C proteins with an F170S amino acid substitution, rHPIV1-C(F170S), but not following infection with wild-type HPIV1. IFN induction coincided with a 100- to 1,000-fold reduction in virus titer, supporting the hypothesis that the HPIV1 C proteins are critical for the inhibition of the innate immune response. Two recently characterized live attenuated HPIV1 vaccine candidates expressing mutant C proteins were also evaluated in HAE. The vaccine candidates, rHPIV1-C(R84G/Delta170)HN(T553A)L(Y942A) and rHPIV1-C(R84G/Delta170)HN(T553A)L(Delta1710-11), which contain temperature-sensitive (ts) attenuating (att) and non-ts att mutations, were highly restricted in growth in HAE at permissive (32 degrees C) and restrictive (37 degrees C) temperatures. The viruses grew slightly better at 37 degrees C than at 32 degrees C, and rHPIV1-C(R84G/Delta170)HN(T553A)L(Y942A) was less attenuated than rHPIV1-C(R84G/Delta170)HN(T553A)L(Delta1710-11). The level of replication in HAE correlated with that previously observed for African green monkeys, suggesting that the HAE model has potential as a tool for the preclinical evaluation of HPIV1 vaccines, although how these in vitro data will correlate with vaccine virus replication in seronegative human subjects remains to be seen.


Assuntos
Brônquios/virologia , Cílios/virologia , Interferons/metabolismo , Mutação , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 1 Humana/metabolismo , Traqueia/virologia , Brônquios/metabolismo , Humanos , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Microscopia Confocal , Modelos Biológicos , Fenótipo , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Temperatura , Traqueia/metabolismo
10.
Virol J ; 4: 67, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17605811

RESUMO

BACKGROUND: Two recombinant, live attenuated human parainfluenza virus type 1 (rHPIV1) mutant viruses have been developed, using a reverse genetics system, for evaluation as potential intranasal vaccine candidates. These rHPIV1 vaccine candidates have two non-temperature sensitive (non-ts) attenuating (att) mutations primarily in the P/C gene, namely CR84GHNT553A (two point mutations used together as a set) and CDelta170 (a short deletion mutation), and two ts att mutations in the L gene, namely LY942A (a point mutation), and LDelta1710-11 (a short deletion), the last of which has not been previously described. The latter three mutations were specifically designed for increased genetic and phenotypic stability. These mutations were evaluated on the HPIV1 backbone, both individually and in combination, for attenuation, immunogenicity, and protective efficacy in African green monkeys (AGMs). RESULTS: The rHPIV1 mutant bearing the novel LDelta1710-11 mutation was highly ts and attenuated in AGMs and was immunogenic and efficacious against HPIV1 wt challenge. The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates were highly ts, with shut-off temperatures of 38 degrees C and 35 degrees C, respectively, and were highly attenuated in AGMs. Immunization with rHPIV1-CR84G/Delta170HNT553ALY942A protected against HPIV1 wt challenge in both the upper and lower respiratory tracts. In contrast, rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 was not protective in AGMs due to over-attenuation, but it is expected to replicate more efficiently and be more immunogenic in the natural human host. CONCLUSION: The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates are clearly highly attenuated in AGMs and clinical trials are planned to address safety and immunogenicity in humans.


Assuntos
Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 1 Humana/imunologia , Vacinas de DNA/imunologia , Proteínas Virais/genética , Administração Intranasal , Animais , Sítios de Ligação Microbiológicos/genética , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Humanos , Dados de Sequência Molecular , Mutação , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 1 Humana/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/imunologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/prevenção & controle , Infecções por Respirovirus/virologia , Resultado do Tratamento , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Células Vero , Proteínas Virais/imunologia , Replicação Viral
11.
Vaccine ; 25(10): 1856-67, 2007 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-17240000

RESUMO

Vaccines are urgently needed to elicit immunity to different influenza virus strains. DNA vaccines can elicit partial protective immunity, however their efficacy requires improvement. We assessed the capacity of individual type I IFN multigene family members as subtype transgenes to abrogate influenza virus replication in a vaccination/challenge mouse model. Differences in antiviral efficacy were found among the subtypes with IFNA5 and IFNA6 being most effective, while IFNA1 was the least effective in reducing lung virus replication. Mice vaccinated with combinatorial HA/IFNA6 or NP/IFNA6 showed reduced lung viral titres, clinical score, body weight loss, and pulmonary tissue damage compared to IFNA6, HA, or NP viral vaccination alone. In addition, IFNA6 increased IgG2a titres with upregulation of IFN-gamma response in the respiratory tract. We conclude that IFN-alpha 6 has antiviral and immunomodulatory effects, which improve efficacy of DNA vaccines for enhanced control of influenza.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Interferon-alfa/imunologia , Nucleoproteínas/imunologia , Proteínas de Ligação a RNA/imunologia , Vacinas de DNA/imunologia , Proteínas do Core Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Peso Corporal , Modelos Animais de Doenças , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Imunoglobulina G/sangue , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/fisiologia , Vacinas contra Influenza/genética , Interferon-alfa/classificação , Interferon-alfa/genética , Interferon gama/biossíntese , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo , Nucleoproteínas/genética , Infecções por Orthomyxoviridae/prevenção & controle , Proteínas de Ligação a RNA/genética , Vacinas de DNA/genética , Proteínas do Core Viral/genética
12.
Virology ; 352(1): 61-73, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16750233

RESUMO

Recombinant human parainfluenza virus type 1 (HPIV1) and mutants containing point and deletion (Delta) mutations in the P/C gene (r-CDelta10-15HNT553A, r-CR84G, r-CF170S and r-CDelta170), which have previously been evaluated as HPIV1 vaccine candidates, were evaluated for their effect on the type I interferon (IFN) response in vitro. HPIV1 wt infection inhibited the IFN response by inhibiting IFN regulatory factor-3 (IRF-3) activation and IFN production in A549 cells and IFN signaling in Vero cells. In contrast, r-CR84G, r-CF170S and r-CDelta170 were defective for inhibition of IRF-3 activation and IFN production and r-CF170S and r-CDelta170 did not inhibit IFN signaling. Thus, HPIV1 antagonizes the IFN response at both the level of induction and signaling, and antagonism at both levels was disrupted by mutations in the P/C gene. Because CF170S affects C and not P, the anti-IFN function can be attributed to the C proteins. These data, in the context of previous in vivo studies, suggest that the loss of antagonism of the IFN response at both the level of induction and signaling, observed with the P/C mutants, r-CF170S and r-CDelta170, was necessary for significant attenuation in African green monkeys (AGMs).


Assuntos
Interferon Tipo I/metabolismo , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 1 Humana/patogenicidade , Fosfoproteínas/genética , Vacinas Atenuadas/imunologia , Proteínas Virais/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Humanos , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 1 Humana/imunologia , Fosfoproteínas/imunologia , Mutação Puntual , Transdução de Sinais , Vacinas Atenuadas/genética , Células Vero , Proteínas Virais/imunologia
13.
Vaccine ; 24(14): 2674-84, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16364511

RESUMO

The P/C gene of human parainfluenza virus type 1 (HPIV1) encodes a nested set of related accessory C proteins, C'/C/Y1/Y2, which have been shown in other paramyxoviruses to have a role in evasion of the type I interferon (IFN) response following virus infection. We previously demonstrated that a set of two amino acid substitutions, CR84G/HNT553A, and a separate amino acid substitution, CF170S, are independently attenuating for HPIV1 in African green monkeys (AGMs). However, in each case the attenuation (att) phenotype is vulnerable to reversion by a single nucleotide change back to wild type. Using reverse genetics, recombinant HPIV1 (rHPIV1) vaccine candidates were generated that were designed for increased genetic and phenotypic stability by: (i) creating a two-amino acid deletion and substitution at the site of the CF170S mutation, yielding CDelta170; (ii) introducing a six amino acid deletion in the N-terminal region of C, CDelta10-15; and (iii) combining these stable deletion mutations with the att CR84G/HNT553A mutation. The resulting rHPIV1 vaccine candidates were evaluated for attenuation in hamsters and AGMs and for immunogenicity and protective efficacy in AGMs. The CDelta10-15 mutation was attenuating in hamsters but not in AGMs, and likely will be of limited value for an HPIV1 vaccine. Conversely, the CR84G/HNT553A mutation set was attenuating in AGMs but not in hamsters. Thus, these two mutations demonstrated reciprocal host range phenotypes involving different regions of C. The CDelta170 mutation conferred a significant level of attenuation in hamsters and AGMs that closely resembled that of CF170S and will be of particular utility for vaccine development because it involves a deletion of six nucleotides rendering it highly refractory to reversion. The combination of the CR84G/HNT553A mutation set and the CDelta170 deletion mutation yielded a virus, rCR84G/Delta170 HNT553A, that exhibited a satisfactory level of attenuation in hamsters and AGMs and was immunogenic and highly protective against HPIV1 wt challenge. This virus will be evaluated clinically as a live intranasal HPIV1 vaccine, one that can be further attenuated as necessary by the introduction of additional stabilized att mutations previously developed in the L protein.


Assuntos
Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 1 Humana/imunologia , Vacinas Atenuadas/imunologia , Animais , Linhagem Celular , Cricetinae , Humanos , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 1 Humana/patogenicidade , Mutação Puntual , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética
14.
Methods Mol Med ; 116: 207-19, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16000864

RESUMO

Delivery of type I interferon (IFN) subtypes by intramuscular inoculation of mice with a recombinant mammalian expression vector encoding IFN stimulates the immune response. Such immunomodulation drives towards a Th1-like response. The degree of stimulation of the immune response was influenced by several parameters of the naked deoxyribonucleic acid (DNA) vaccination protocol. Pretreatment of mice with bupivacaine increased transgene expression in situ. The specific subtype gene of type I IFN, the DNA concentration, the combined use of two or more subtypes, and the timing of the DNA immunisations were all found to influence the level of efficacy of IFN gene therapy in a mouse model for cytomegalovirus (CMV) infection and disease. In addition, adjuvant therapy, using type I IFN genes, for DNA virus vaccination (CMV glycoprotein B) enhanced viral-specific immunity and reduced the severity of myocarditis in mice. Thus, type I IFN gene therapy has potent adjuvant properties when delivered as DNA and can be used to regulate virus infection and disease via pleiotropic actions in the stimulation of immune responses.


Assuntos
Infecções por Citomegalovirus , Técnicas de Transferência de Genes , Terapia Genética/métodos , Interferons/uso terapêutico , Isoformas de Proteínas/uso terapêutico , Anestésicos Locais/metabolismo , Animais , Bupivacaína/metabolismo , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/terapia , Humanos , Interferons/genética , Interferons/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/patologia , Miocardite/virologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transgenes , Vacinas de DNA
15.
Vaccine ; 23(38): 4631-46, 2005 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-15951066

RESUMO

A set of recombinant, live attenuated human parainfluenza virus type 1 (rHPIV1) vaccine candidates was evaluated for attenuation, immunogenicity, and protective efficacy in African green monkeys (AGMs). Temperature sensitive (ts) and non-ts attenuating (att) mutations in the P/C and L genes were introduced individually or in various combinations into rHPIV1, including the C(R84G) and HN(T553A) mutations identified in the present work and the C(F170S), L(Y942A), and L(L992C) mutations identified previously. The rHPIV1 vaccine candidates exhibited a spectrum of attenuation in AGMs. One genetically and phenotypically stable vaccine candidate, rC(R84G/F170S)L(Y942A/L992C), was attenuated and efficacious in AGMs and is a promising live attenuated intranasal HPIV1 vaccine candidate suitable for clinical evaluation.


Assuntos
Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 1 Humana/imunologia , Vacinas Atenuadas/imunologia , Animais , Anticorpos Antivirais/sangue , Modelos Animais de Doenças , Humanos , Macaca mulatta , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 1 Humana/genética , Vírus da Parainfluenza 1 Humana/patogenicidade , Mutação Puntual , Infecções por Respirovirus/prevenção & controle , Temperatura , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Proteínas Virais/genética , Proteínas Virais/imunologia
16.
Immunol Cell Biol ; 82(2): 119-26, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15061762

RESUMO

Gene therapy using DNA encoding type I IFN subtypes IFNA6, IFNA9 and IFNB suppresses murine cytomegalovirus (MCMV)-myocarditis, a predominantly cell-mediated disease in BALB/c mice. CD8(+) T cells are the principal cell type within the inflamed myocardium. As such, we investigated the effects of IFN subtype treatment on this T-cell subset and other cell types in the cardiac infiltrate. In the acute phase of disease, IFNA6 and IFNA9 treatments significantly reduced the number of CD8(+) T cells within the foci of cellular infiltration in the heart. During the chronic phase, which is primarily autoimmune in nature, IFNB treatment significantly reduced CD8(+) T cells. B-cell and neutrophil numbers in the cardiac infiltrate were also reduced following IFNB immunotherapy. Although early inflammatory responses are important for resolution of virus infection, high numbers of lymphocytes persisting in the myocardium may lead to exacerbation of disease. Our data suggests that type I IFN DNA therapy regulates cardiac cellular infiltration. Thus, treatment with IFN-beta administered prophylactically to high-risk patients in acquiring CMV infection may reduce the development of chronic autoimmune myocarditis.


Assuntos
Doenças Autoimunes/terapia , Terapia Genética , Interferon-alfa/genética , Interferon beta/genética , Miocardite/terapia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , DNA/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Masculino , Camundongos , Miocardite/imunologia , Miocardite/metabolismo
17.
Biol Proced Online ; 5: 43-52, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12734557

RESUMO

Type I interferon (IFN) gene therapy modulates the immune response leading to inflammatory heart disease following cytomegalovirus (CMV) infection in a murine model of post-viral myocarditis. Efficacy of different immunisation protocols for the IFN constructs was influenced by the dose of DNA, subtype choice, combination use, pre-medication, and timing of DNA administration. Optimal efficacy was found with bupivacaine treatment prior to DNA inoculation of 200mg IFN DNA 14 days prior to virus challenge. Maximal antiviral and antimyocarditic effects were achieved with this vaccination schedule. Furthermore, inoculation of synergistic IFN subtypes demonstrated enhanced efficacy when delivered either alone or with CMV gB DNA vaccination in the CMV model. Thus naked DNA delivery of IFN provides an avenue of immunotherapy for regulating herpesvirus-induced diseases.

18.
Blood ; 101(7): 2727-35, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12446459

RESUMO

Type I interferons (IFNs), pleiotropic cytokines with antiviral, antiproliferative, apoptotic, and immunoregulatory functions, are efficacious in the treatment of malignancies, viral infections, and autoimmune diseases. Binding of these cytokines to their cognate receptor leads to activation of the Jak-signal transducers and activators of transcription (STAT) signaling pathway and altered gene expression. This signal pathway has been intensely studied using human IFN-alpha 2 and IFN-beta. However, there are over 14 human IFN-alpha subtypes and over 10 murine IFN-alpha subtypes, with a single IFN-beta subtype in both species. J2E cells are immortalized at the proerythroblast stage of development and produce a rapid and fatal erythroleukemia in vivo. These cells retain the ability to respond to erythropoietin in vitro by proliferating, differentiating, and remaining viable in the absence of serum. Here, we show that J2E cells are also functionally regulated differentially by IFN subtype treatment in vitro. A novel finding was the selective activation of STAT and mitogen-activated protein kinase (MAPK) molecules by different subtypes binding the IFN receptor. These findings indicate distinct effects for individual type I IFN subtypes, which are able to differentially activate members of the STAT and MAPK family. Finally, we investigated the efficacy of IFN naked DNA therapy in treating J2E-induced erythroleukemia in athymic nude mice. IFN subtypes differentially regulated the onset of erythroleukemia with delayed onset and increased survival, possibly via a reduction in cell viability, and enhanced antiproliferative and apoptotic effects observed for IFNA6 and IFNA9 treatment, respectively. Moreover, these data highlight the necessity to choose the best IFN subtype in disease treatment.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interferon Tipo I/uso terapêutico , Leucemia Eritroblástica Aguda/terapia , Proteínas do Leite , Transativadores/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , DNA/administração & dosagem , DNA/uso terapêutico , Proteínas de Ligação a DNA/efeitos dos fármacos , Terapia Genética , Interferon Tipo I/genética , Interferon Tipo I/farmacologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/farmacologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Transdução de Sinais/efeitos dos fármacos , Taxa de Sobrevida , Transativadores/efeitos dos fármacos , Resultado do Tratamento , Células Tumorais Cultivadas
19.
Immunol Cell Biol ; 80(5): 425-35, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12225378

RESUMO

Delivery of type I IFN transgenes by naked DNA immunization can protect against cytomegalovirus infection and myocarditis. Here, we investigate IFN transgene expression, antiviral efficacy, and immunomodulation of myocarditis using various treatment regimes in a mouse CMV model. In vivo expression of the IFN transgene was observed in the sera for 35 days post-DNA inoculation. Prophylactic IFN-A6 and IFN-B DNA treatment for 14 days prior to murine cytomegalovirus (MCMV) infection was more efficacious in significantly reducing viral titres, than 2 days prior to or 2 days post-virus infection. Similarly, IFN-A6 DNA treatment commencing 14 days prior to virus infection was superior in suppressing both acute and chronic myocarditis. Furthermore, reduction of autoantibody titres was more pronounced when IFN was administered 14 days prior to viral infection. Combinational IFN gene therapy was assessed for synergy between IFN subtypes. Combination treatment with either IFN-A6/A9 or IFN-A6/B greatly reduced spleen viral titres while IFN-A6/B and IFN-A9/B reduced virus replication in the liver. Only IFN-A6/A9 and IFN-A9/B reduced acute viral myocarditis, whereas IFNA6/B treatment was most efficacious for autoimmune chronic myocarditis. Finally, treatment with IFN-A6 DNA 2 weeks post-MCMV infection proved effective at inhibiting the development of chronic autoimmune myocarditis. These findings suggest that immunomodulation of both antiviral and autoimmune responses by IFN DNA immunization may be an avenue for improved viral immunotherapy.


Assuntos
Doenças Autoimunes/prevenção & controle , Infecções por Citomegalovirus/prevenção & controle , Imunoterapia Ativa , Interferon Tipo I/genética , Interferon beta/genética , Muromegalovirus , Miocardite/prevenção & controle , Vacinas de DNA/uso terapêutico , Doença Aguda , Animais , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Doenças Autoimunes/terapia , Doenças Autoimunes/virologia , Células COS , Chlorocebus aethiops , Doença Crônica , Citocinas/biossíntese , Infecções por Citomegalovirus/terapia , DNA Complementar/genética , Modelos Animais de Doenças , Esquemas de Imunização , Injeções Intramusculares , Interferon Tipo I/fisiologia , Interferon-alfa , Interferon beta/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/fisiologia , Músculo Esquelético/fisiologia , Miocardite/terapia , Miocardite/virologia , Regeneração , Organismos Livres de Patógenos Específicos , Baço/virologia , Transgenes , Vacinas de DNA/imunologia , Replicação Viral
20.
Immunology ; 106(3): 428-37, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12100732

RESUMO

Type I interferons (IFNs) are produced early in response to viral infection and modulate adaptive immunity. Previously we demonstrated localized protection against murine cytomegalovirus (MCMV) infection in IFN DNA-inoculated mice. Here we examine the effect of seven IFN subtypes (IFNA1, A2, A4, A5, A6, A9 and B), administered by DNA inoculation, on systemic MCMV infection and myocarditis. IFN transgene expression altered the pathogenesis of MCMV infection with regard to virus titre and myocarditis. IFNA6 treatment reduced MCMV replication whilst IFNA5 and A2 enhanced virus replication. IFNA6, A9, and B treatment inhibited acute myocarditis. A T helper type 1-like, antibody and cytokine, response correlated with decreased virus titre and myocarditis. In addition, IFNA6 was able to reduce chronic cardiac inflammation. This research into the effectiveness of seven type I IFNs, using DNA gene therapy, highlights the need for correct subtype usage in the treatment of disease. We demonstrate effective subtypes for treatment in both the acute and chronic phases of MCMV infection and the resultant development of myocarditis.


Assuntos
Terapia Genética/métodos , Infecções por Herpesviridae/terapia , Interferon Tipo I/genética , Muromegalovirus/imunologia , Miocardite/prevenção & controle , Doença Aguda , Animais , Doença Crônica , Citocinas/biossíntese , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Imunoglobulina G/biossíntese , Interferon Tipo I/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/fisiologia , Miocardite/imunologia , Miocardite/virologia , Miosinas/imunologia , Transgenes/imunologia , Carga Viral , Replicação Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...