Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Pediatric Infect Dis Soc ; 12(3): 173-176, 2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-36594442

RESUMO

We conducted a phase I clinical trial of the live-attenuated recombinant human parainfluenza virus type 2 (HPIV2) vaccine candidate rHPIV2-15C/948L/∆1724 sequentially in adults, HPIV2-seropositive children, and HPIV2-seronegative children, the target population for vaccination. rHPIV2-15C/948L/∆1724 was appropriately restricted in replication in adults and HPIV2-seropositive children but was overattenuated for HPIV2-seronegative children.


Assuntos
Vírus da Parainfluenza 2 Humana , Vacinas Sintéticas , Adulto , Criança , Humanos , Anticorpos Antivirais , Vírus da Parainfluenza 1 Humana , Vírus da Parainfluenza 3 Humana , Vacinas Atenuadas
2.
J Virol ; 90(21): 10022-10038, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27581977

RESUMO

Human respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are major pediatric respiratory pathogens that lack vaccines. A chimeric bovine/human PIV3 (rB/HPIV3) virus expressing the unmodified, wild-type (wt) RSV fusion (F) protein from an added gene was previously evaluated in seronegative children as a bivalent intranasal RSV/HPIV3 vaccine, and it was well tolerated but insufficiently immunogenic for RSV F. We recently showed that rB/HPIV3 expressing a partially stabilized prefusion form (pre-F) of RSV F efficiently induced "high-quality" RSV-neutralizing antibodies, defined as antibodies that neutralize RSV in vitro without added complement (B. Liang et al., J Virol 89:9499-9510, 2015, doi:10.1128/JVI.01373-15). In the present study, we modified RSV F by replacing its cytoplasmic tail (CT) domain or its CT and transmembrane (TM) domains (TMCT) with counterparts from BPIV3 F, with or without pre-F stabilization. This resulted in RSV F being packaged in the rB/HPIV3 particle with an efficiency similar to that of RSV particles. Enhanced packaging was substantially attenuating in hamsters (10- to 100-fold) and rhesus monkeys (100- to 1,000-fold). Nonetheless, TMCT-directed packaging substantially increased the titers of high-quality RSV-neutralizing serum antibodies in hamsters. In rhesus monkeys, a strongly additive immunogenic effect of packaging and pre-F stabilization was observed, as demonstrated by 8- and 30-fold increases of RSV-neutralizing serum antibody titers in the presence and absence of added complement, respectively, compared to pre-F stabilization alone. Analysis of vaccine-induced F-specific antibodies by binding assays indicated that packaging conferred substantial stabilization of RSV F in the pre-F conformation. This provides an improved version of this well-tolerated RSV/HPIV3 vaccine candidate, with potently improved immunogenicity, which can be returned to clinical trials. IMPORTANCE: Human respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are major viral agents of acute pediatric bronchiolitis and pneumonia worldwide that lack vaccines. A bivalent intranasal RSV/HPIV3 vaccine candidate consisting of a chimeric bovine/human PIV3 (rB/HPIV3) strain expressing the RSV fusion (F) protein was previously shown to be well tolerated by seronegative children but was insufficiently immunogenic for RSV F. In the present study, the RSV F protein was engineered to be packaged efficiently into vaccine virus particles. This resulted in a significantly enhanced quantity and quality of RSV-neutralizing antibodies in hamsters and nonhuman primates. In nonhuman primates, this effect was strongly additive to the previously described stabilization of the prefusion conformation of the F protein. The improved immunogenicity of RSV F by packaging appeared to involve prefusion stabilization. These findings provide a potently more immunogenic version of this well-tolerated vaccine candidate and should be applicable to other vectored vaccines.


Assuntos
Anticorpos Neutralizantes/genética , Vetores Genéticos/genética , Vírus da Parainfluenza 3 Bovina/genética , Vírus da Parainfluenza 3 Humana/genética , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/genética , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Capsídeo/metabolismo , Bovinos , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Humanos , Macaca mulatta , Vírus da Parainfluenza 3 Bovina/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/imunologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/virologia , Células Vero , Proteínas Virais de Fusão/imunologia , Replicação Viral/genética
3.
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
4.
J Virol ; 89(18): 9499-510, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26157122

RESUMO

UNLABELLED: Respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are the first and second leading viral agents of severe respiratory tract disease in infants and young children worldwide. Vaccines are not available, and an RSV vaccine is particularly needed. A live attenuated chimeric recombinant bovine/human PIV3 (rB/HPIV3) vector expressing the RSV fusion (F) glycoprotein from an added gene has been under development as a bivalent vaccine against RSV and HPIV3. Previous clinical evaluation of this vaccine candidate suggested that increased genetic stability and immunogenicity of the RSV F insert were needed. This was investigated in the present study. RSV F expression was enhanced 5-fold by codon optimization and by modifying the amino acid sequence to be identical to that of an early passage of the original clinical isolate. This conferred a hypofusogenic phenotype that presumably reflects the original isolate. We then compared vectors expressing stabilized prefusion and postfusion versions of RSV F. In a hamster model, prefusion F induced increased quantity and quality of RSV-neutralizing serum antibodies and increased protection against wild-type (wt) RSV challenge. In contrast, a vector expressing the postfusion F was less immunogenic and protective. The genetic stability of the RSV F insert was high and was not affected by enhanced expression or the prefusion or postfusion conformation of RSV F. These studies provide an improved version of the previously well-tolerated rB/HPIV3-RSV F vaccine candidate that induces a superior RSV-neutralizing serum antibody response. IMPORTANCE: Respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (HPIV3) are two major causes of pediatric pneumonia and bronchiolitis. The rB/HPIV3 vector expressing RSV F protein is a candidate bivalent live vaccine against HPIV3 and RSV. Previous clinical evaluation indicated the need to increase the immunogenicity and genetic stability of the RSV F insert. Here, we increased RSV F expression by codon optimization and by modifying the RSV F amino acid sequence to conform to that of an early passage of the original isolate. This resulted in a hypofusogenic phenotype, which likely represents the original phenotype before adaptation to cell culture. We also included stabilized versions of prefusion and postfusion RSV F protein. Prefusion RSV F induced a larger quantity and higher quality of RSV-neutralizing serum antibodies and was highly protective. This provides an improved candidate for further clinical evaluation.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Expressão Gênica , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Chlorocebus aethiops , Cricetinae , Humanos , Vírus da Parainfluenza 3 Humana/genética , Proteínas Recombinantes , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/metabolismo , Células Vero , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/genética , Vacinas Virais/genética , Vacinas Virais/metabolismo
5.
J Virol ; 89(6): 3318-31, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25589643

RESUMO

UNLABELLED: Human parainfluenza virus type 3 (HPIV3), a paramyxovirus, is a major viral cause of severe lower respiratory tract disease in infants and children. The gene-end (GE) transcription signal of the HPIV3 matrix (M) protein gene is identical to those of the nucleoprotein and phosphoprotein genes except that it contains an apparent 8-nucleotide insert. This was associated with an increased synthesis of a readthrough transcript of the M gene and the downstream fusion (F) protein gene. We hypothesized that this insert may function to downregulate the expression of F protein by interfering with termination/reinitiation at the M-F gene junction, thus promoting the production of M-F readthrough mRNA at the expense of monocistronic F mRNA. To test this hypothesis, two similar recombinant HPIV3 viruses from which this insert in the M-GE signal was removed were generated. The M-GE mutants exhibited a reduction in M-F readthrough mRNA and an increase in monocistronic F mRNA. This resulted in a substantial increase in F protein synthesis in infected cells as well as enhanced incorporation of F protein into virions. The efficiency of mutant virus replication was similar to that of wild-type (wt) HPIV3 both in vitro and in vivo. However, the F-protein-specific serum antibody response in hamsters was increased for the mutants compared to wt HPIV3. This study identifies a previously undescribed viral mechanism for attenuating the host adaptive immune response. Repairing the M-GE signal should provide a means to increase the antibody response to a live attenuated HPIV3 vaccine without affecting viral replication and attenuation. IMPORTANCE: The HPIV3 M-GE signal was previously shown to contain an apparent 8-nucleotide insert that was associated with increased synthesis of a readthrough mRNA of the M gene and the downstream F gene. However, whether this had any significant effect on the synthesis of monocistronic F mRNA or F protein, virus replication, virion morphogenesis, and immunogenicity was unknown. Here, we show that the removal of this insert shifts F gene transcription from readthrough M-F mRNA to monocistronic F mRNA. This resulted in a substantial increase in the amount of F protein expressed in the cell and packaged in the virus particle. This did not affect virus replication but increased the F-specific antibody response in hamsters. Thus, in wild-type HPIV3, the aberrant M-GE signal operates a previously undescribed mechanism that reduces the expression of a major neutralization and protective antigen, resulting in reduced immunogenicity. This has implications for the design of live attenuated HPIV3 vaccines; specifically, the antibody response against F can be elevated by "repairing" the M-GE signal to achieve higher-level F antigen expression, with no effect on attenuation.


Assuntos
Anticorpos Antivirais/imunologia , Regulação Viral da Expressão Gênica , Vírus da Parainfluenza 3 Humana/genética , Infecções por Respirovirus/imunologia , Transcrição Gênica , Proteínas Virais de Fusão/genética , Proteínas da Matriz Viral/genética , Animais , Sequência de Bases , Cricetinae , Regulação para Baixo , Humanos , Mesocricetus , Dados de Sequência Molecular , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/virologia , Proteínas Virais de Fusão/imunologia , Proteínas da Matriz Viral/imunologia
6.
Curr Pharm Des ; 20(42): 6522-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25341929

RESUMO

As the number of novel drugs that have entered the market in oncology has slowed in recent years, there has been a dramatic shift towards new therapeutic approaches. The majority of cancer patients die from metastasis formation, which has prompted the pharmaceutical industry to begin to investigate a new class of agents: anti-metastatics. This review provides an overview of the targets, mechanisms of action, and drug substances currently in the pharma pipeline to inhibit tumor cell migration and metastasis formation.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/patologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Movimento Celular/efeitos dos fármacos , Humanos , Neoplasias/metabolismo
7.
J Virol ; 88(8): 4237-50, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24478424

RESUMO

UNLABELLED: A recombinant chimeric bovine/human parainfluenza type 3 virus (rB/HPIV3) vector expressing the respiratory syncytial virus (RSV) fusion F glycoprotein previously exhibited disappointing levels of RSV F immunogenicity and genetic stability in children (D. Bernstein et al., Pediatr. Infect. Dis. J. 31:109-114, 2012; C.-F. Yang et al., Vaccine 31:2822-2827, 2013). To investigate parameters that might affect vaccine performance and stability, we constructed and characterized rB/HPIV3 viruses expressing RSV F from the first (pre-N), second (N-P), third (P-M), and sixth (HN-L) genome positions. There was a 30- to 69-fold gradient in RSV F expression from the first to the sixth position. The inserts moderately attenuated vector replication in vitro and in the upper and lower respiratory tracts of hamsters: this was not influenced by the level of RSV F expression and syncytium formation. Surprisingly, inserts in the second, third, and sixth positions conferred increased temperature sensitivity: this was greatest for the third position and was the most attenuating in vivo. Each rB/HPIV3 vector induced a high titer of neutralizing antibodies in hamsters against RSV and HPIV3. Protection against RSV challenge was greater for position 2 than for position 6. Evaluation of insert stability suggested that RSV F is under selective pressure to be silenced during vector replication in vivo, but this was not exacerbated by a high level of RSV F expression and generally involved a small percentage of recovered vector. Vector passaged in vitro accumulated mutations in the HN open reading frame, causing a dramatic increase in plaque size that may have implications for vaccine production and immunogenicity. IMPORTANCE: The research findings presented here will be instrumental for improving the design of a bivalent pediatric vaccine for respiratory syncytial virus and parainfluenza virus type 3, two major causes of severe respiratory tract infection in infants and young children. Moreover, this knowledge has general application to the development and clinical evaluation of other mononegavirus vectors and vaccines.


Assuntos
Vírus da Parainfluenza 3 Bovina/genética , Vírus da Parainfluenza 3 Humana/genética , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Anticorpos Antivirais/imunologia , Cricetinae , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Mesocricetus , Vírus da Parainfluenza 3 Bovina/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Engenharia de Proteínas , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/química , Vacinas contra Vírus Sincicial Respiratório/genética , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/imunologia , Vírus Sinciciais Respiratórios/genética , Proteínas Virais de Fusão/administração & dosagem , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Replicação Viral
8.
Vaccine ; 31(48): 5706-12, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24103895

RESUMO

BACKGROUND: Human parainfluenza virus type 3 (HPIV3) is a common cause of upper and lower respiratory tract illness in infants and young children. Live-attenuated cold-adapted HPIV3 vaccines have been evaluated in infants but a suitable interval for administration of a second dose of vaccine has not been defined. METHODS: HPIV3-seronegative children between the ages of 6 and 36 months were randomized 2:1 in a blinded study to receive two doses of 105 TCID50 (50% tissue culture infectious dose) of live-attenuated, recombinant cold-passaged human PIV3 vaccine (rHPIV3cp45) or placebo 6 months apart. Serum antibody levels were assessed prior to and approximately 4-6 weeks after each dose. Vaccine virus infectivity, defined as detection of vaccine-HPIV3 in nasal wash and/or a≥4-fold rise in serum antibody titer, and reactogenicity were assessed on days 3, 7, and 14 following immunization. RESULTS: Forty HPIV3-seronegative children (median age 13 months; range 6-35 months) were enrolled; 27 (68%) received vaccine and 13 (32%) received placebo. Infectivity was detected in 25 (96%) of 26 evaluable vaccinees following doses 1 and 9 of 26 subject (35%) following dose 2. Among those who shed virus, the median duration of viral shedding was 12 days (range 6-15 days) after dose 1 and 6 days (range 3-8 days) after dose 2, with a mean peak log10 viral titer of 3.4 PFU/mL (SD: 1.0) after dose 1 compared to 1.5 PFU/mL (SD: 0.92) after dose 2. Overall, reactogenicity was mild, with no difference in rates of fever and upper respiratory infection symptoms between vaccine and placebo groups. CONCLUSION: rHPIV3cp45 was immunogenic and well-tolerated in seronegative young children. A second dose administered 6 months after the initial dose was restricted in those previously infected with vaccine virus; however, the second dose boosted antibody responses and induced antibody responses in two previously uninfected children.


Assuntos
Vacinas contra Parainfluenza/efeitos adversos , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/prevenção & controle , Vacinação/efeitos adversos , Vacinação/métodos , Anticorpos Antivirais/sangue , Pré-Escolar , Método Duplo-Cego , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Humanos , Lactente , Masculino , Cavidade Nasal/virologia , Vacinas contra Parainfluenza/administração & dosagem , Vacinas contra Parainfluenza/genética , Vírus da Parainfluenza 3 Humana/genética , Placebos/administração & dosagem , Infecções por Respirovirus/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
9.
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
10.
Curr Opin Virol ; 2(3): 294-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22709516

RESUMO

Human parainfluenza viruses (HPIVs) are a common cause of acute respiratory illness throughout life. Infants, children, and the immunocompromised are the most likely to develop severe disease. HPIV1 and HPIV2 are best known to cause croup while HPIV3 is a common cause of bronchiolitis and pneumonia. HPIVs replicate productively in respiratory epithelial cells and do not spread systemically unless the host is severely immunocompromised. Molecular studies have delineated how HPIVs evade and block cellular innate immune responses to permit efficient replication, local spread, and host-to-host transmission. Studies using ex vivo human airway epithelium have focused on virus tropism, cellular pathology and the epithelial inflammatory response, elucidating how events early in infection shape the adaptive immune response and disease outcome.


Assuntos
Bronquiolite Viral/patologia , Crupe/patologia , Infecções por Paramyxoviridae/patologia , Infecções por Paramyxoviridae/virologia , Pneumonia Viral/patologia , Respirovirus/patogenicidade , Bronquiolite Viral/imunologia , Bronquiolite Viral/virologia , Pré-Escolar , Crupe/imunologia , Crupe/virologia , Humanos , Evasão da Resposta Imune , Hospedeiro Imunocomprometido , Lactente , Infecções por Paramyxoviridae/imunologia , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Respirovirus/imunologia
11.
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
12.
J Virol ; 85(8): 4007-19, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21289116

RESUMO

Human parainfluenza virus type 2 (HPIV-2), an important pediatric respiratory pathogen, encodes a V protein that inhibits type I interferon (IFN) induction and signaling. Using reverse genetics, we attempted the recovery of a panel of V mutant viruses that individually contained one of six cysteine-to-serine (residues 193, 197, 209, 211, 214, and 218) substitutions, one of two paired charge-to-alanine (R175A/R176A and R205A/K206A) substitutions, or a histidine-to-phenylalanine (H174F) substitution. This mutagenesis was performed using a cDNA-derived HPIV-2 virus that expressed the V and P coding sequences from separate mRNAs. Of the cysteine substitutions, only C193S, C214S, and C218S yielded viable virus, and only the C214S mutant replicated well enough for further analysis. The H174F, R175A/R176A, and R205A/K206A mutants were viable and replicated well. The H174F and R205A/K206A mutants did not differ from the wild-type (WT) V in their ability to physically interact with MDA5, a cytoplasmic sensor of nonself RNA that induces type I IFN. Like WT HPIV-2, these mutants inhibited IFN-ß induction and replicated efficiently in African green monkeys (AGMs). In contrast, the C214S and R175A/R176A mutants did not bind MDA5 efficiently, did not inhibit interferon regulatory factor 3 (IRF3) dimerization or IFN-ß induction, and were attenuated in AGMs. These findings indicate that V binding to MDA5 is important for HPIV-2 virulence in nonhuman primates and that some V protein residues involved in MDA5 binding are not essential for efficient HPIV-2 growth in vitro. Using a transient expression system, 20 additional mutant V proteins were screened for MDA5 binding, and the region spanning residues 175 to 180 was found to be essential for this activity.


Assuntos
RNA Helicases DEAD-box/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Parainfluenza 2 Humana/patogenicidade , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Replicação Viral , Substituição de Aminoácidos/genética , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Humanos , Macaca mulatta , Viabilidade Microbiana , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Vírus da Parainfluenza 2 Humana/genética , Ligação Proteica , Proteínas Virais/genética , Fatores de Virulência/genética
13.
Immunol Cell Biol ; 89(2): 173-82, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20603636

RESUMO

MxA is an antiviral protein induced by interferon (IFN)-α/ß that is known to inhibit the replication of many RNA viruses. In these experiments, the 76-kDa MxA protein expressed in IFN-α-treated cells was shown to have antiviral activity against herpes simplex virus-1 (HSV-1), a human DNA virus. However, MxA was expressed as a 56-kDa protein in HSV-1-infected cells in the absence of IFN-α. This previously unrecognized MxA isoform was produced from an alternatively spliced MxA transcript that had a deletion of Exons 14-16 and a frame shift altering the C-terminus. The variant MxA (varMxA) isoform was associated with HSV-1 regulatory proteins and virions in nuclear replication compartments. varMxA expression enhanced HSV-1 infection as shown by a reduction in infectious virus titers from cells in which MxA had been inhibited by RNA interference and by an increase in HSV-1 titers when the 56-kDa varMxA was expressed constitutively. Thus, the human MxA gene encodes two MxA isoforms, which are expressed differentially depending on whether the stimulus is IFN-α or HSV-1. These findings show that alternative splicing of cellular mRNA can result in expression of a novel isoform of a host defense gene that supports instead of restricting viral infection.


Assuntos
Proteínas de Ligação ao GTP/genética , Herpesvirus Humano 1/fisiologia , Replicação Viral/fisiologia , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/metabolismo , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/ultraestrutura , Humanos , Interferon-alfa/farmacologia , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/virologia , Dados de Sequência Molecular , Proteínas de Resistência a Myxovirus , Biossíntese de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos , Vírion/efeitos dos fármacos , Vírion/fisiologia , Replicação Viral/efeitos dos fármacos
14.
Virology ; 406(1): 65-79, 2010 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-20667570

RESUMO

The HPIV2 V protein inhibits type I interferon (IFN) induction and signaling. To manipulate the V protein, whose coding sequence overlaps that of the polymerase-associated phosphoprotein (P), without altering the P protein, we generated an HPIV2 virus in which P and V are expressed from separate genes (rHPIV2-P+V). rHPIV2-P+V replicated like HPIV2-WT in vitro and in non-human primates. HPIV2-P+V was modified by introducing two separate mutations into the V protein to create rHPIV2-L101E/L102E and rHPIV2-Delta122-127. In contrast to HPIV2-WT, both mutant viruses were unable to degrade STAT2, leaving virus-infected cells susceptible to IFN. Neither mutant, nor HPIV2-WT, induced significant amounts of IFN-beta in infected cells. Surprisingly, neither rHPIV2-L101E/L102E nor rHPIV2-Delta122-127 was attenuated in two species of non-human primates. This indicates that loss of HPIV2's ability to inhibit IFN signaling is insufficient to attenuate virus replication in vivo as long as IFN induction is still inhibited.


Assuntos
Interferons/fisiologia , Vírus da Parainfluenza 2 Humana/genética , Primatas/virologia , Proteínas Virais/genética , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , DNA Viral/genética , Proteínas de Ligação a DNA/metabolismo , Genes Virais , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferon Tipo I/farmacologia , Macaca mulatta , Mutação , Fases de Leitura Aberta , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 2 Humana/patogenicidade , Vírus da Parainfluenza 2 Humana/fisiologia , Fosfoproteínas/genética , Primatas/imunologia , Proteínas Recombinantes , Transdução de Sinais , Especificidade da Espécie , Células Vero , Proteínas Virais/imunologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética , Replicação Viral/imunologia
15.
Vaccine ; 28(15): 2788-98, 2010 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20139039

RESUMO

Human parainfluenza viruses (HPIVs) are common causes of severe pediatric respiratory viral disease. We characterized wild-type HPIV2 infection in an in vitro model of human airway epithelium (HAE) and found that the virus replicates to high titer, sheds apically, targets ciliated cells, and induces minimal cytopathology. Replication of an experimental, live attenuated HPIV2 vaccine strain, containing both temperature sensitive (ts) and non-ts attenuating mutations, was restricted >30-fold compared to rHPIV2-WT in HAE at 32 degrees C and exhibited little productive replication at 37 degrees C. This restriction paralleled attenuation in the upper and lower respiratory tract of African green monkeys, supporting the HAE model as an appropriate and convenient system for characterizing HPIV2 vaccine candidates.


Assuntos
Vírus da Parainfluenza 2 Humana/crescimento & desenvolvimento , Mucosa Respiratória/virologia , Vacinas Virais , Animais , Células Cultivadas , Chlorocebus aethiops , Humanos , Técnicas de Cultura de Órgãos , Vírus da Parainfluenza 2 Humana/patogenicidade , Infecções por Rubulavirus/patologia , Infecções por Rubulavirus/virologia , Vacinas Atenuadas , Virulência
16.
Virology ; 397(2): 285-98, 2010 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19969320

RESUMO

In wild-type human parainfluenza virus type 2 (WT HPIV2), one gene (the P/V gene) encodes both the polymerase-associated phosphoprotein (P) and the accessory V protein. We generated a HPIV2 virus (rHPIV2-V(ko)) in which the P/V gene encodes only the P protein to examine the role of V in replication in vivo and as a potential live attenuated virus vaccine. Preventing expression of V protein severely impaired virus recovery from cDNA and growth in vitro, particularly in IFN-competent cells. rHPIV2-V(ko), unlike WT HPIV2, strongly induced IFN-beta and permitted IFN signaling, leading to establishment of a robust antiviral state. rHPIV2-V(ko) infection induced extensive syncytia and cytopathicity that was due to both apoptosis and necrosis. Replication of rHPIV2-V(ko) was highly restricted in the respiratory tract of African green monkeys and in differentiated primary human airway epithelial (HAE) cultures, suggesting that V protein is essential for efficient replication of HPIV2 in organized epithelial cells and that rHPIV2-V(ko) is over-attenuated for use as a live attenuated vaccine.


Assuntos
Interferons/imunologia , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 2 Humana/fisiologia , Transdução de Sinais , Proteínas Virais/imunologia , Proteínas Virais/fisiologia , Replicação Viral , Animais , Células Cultivadas , Chlorocebus aethiops , Regulação para Baixo , Técnicas de Inativação de Genes , Humanos , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , Vírus da Parainfluenza 2 Humana/genética , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Proteínas Virais/genética
17.
Vaccine ; 25(34): 6409-22, 2007 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-17658669

RESUMO

Previously, we identified several attenuating mutations in the L polymerase protein of human parainfluenza virus type 2 (HPIV2) and genetically stabilized those mutations using reverse genetics [Nolan SM, Surman S, Amaro-Carambot E, Collins PL, Murphy BR, Skiadopoulos MH. Live-attenuated intranasal parainfluenza virus type 2 vaccine candidates developed by reverse genetics containing L polymerase protein mutations imported from heterologous paramyxoviruses. Vaccine 2005;39(23):4765-74]. Here we describe the discovery of an attenuating mutation at nucleotide 15 (15(T-->C)) in the 3' genomic promoter that was also present in the previously characterized mutants. We evaluated the properties of this promoter mutation alone and in various combinations with the L polymerase mutations. Amino acid substitutions at L protein positions 460 (460A or 460P) or 948 (948L), or deletion of amino acids 1724 and 1725 (Delta1724), each conferred a temperature sensitivity (ts) phenotype whereas the 15(T-->C) mutation did not. The 460A and 948L mutations each contributed to restricted replication in the lower respiratory tract of African green monkeys, but the Delta1724 mutation increased attenuation only in certain combinations with other mutations. We constructed two highly attenuated viruses, rV94(15C)/460A/948L and rV94(15C)/948L/Delta1724, that were immunogenic and protective against challenge with wild-type HPIV2 in African green monkeys and, therefore, appear to be suitable for evaluation in humans.


Assuntos
Mutação , Vacinas contra Parainfluenza/imunologia , Vírus da Parainfluenza 2 Humana/imunologia , Regiões Promotoras Genéticas , Vacinas Sintéticas/imunologia , Proteínas Virais/genética , Animais , Linhagem Celular , Cricetinae , Humanos , Macaca mulatta , Mesocricetus , Vírus da Parainfluenza 2 Humana/genética , Vírus da Parainfluenza 2 Humana/fisiologia , Sistema Respiratório/virologia , Temperatura , Vacinas Atenuadas/imunologia , Replicação Viral
18.
J Virol ; 80(23): 11806-16, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16971426

RESUMO

Several functions have been attributed to the serine/threonine protein kinase encoded by open reading frame 66 (ORF66) of varicella-zoster virus (VZV), including modulation of the apoptosis and interferon pathways, down-regulation of major histocompatibility complex class I cell surface expression, and regulation of IE62 localization. The amino acid sequence of the ORF66 protein contains a recognizable conserved kinase domain. Point mutations were introduced into conserved protein kinase motifs to evaluate their importance to ORF66 protein functions. Two substitution mutants were generated, including a G102A substitution, which blocked autophosphorylation and altered IE62 localization, and an S250P substitution, which had no effect on either autophosphorylation or IE62 localization. Both kinase domain mutants grew to titers equivalent to recombinant parent Oka (pOka) in vitro. pOka66G102A had slightly reduced growth in skin, which was comparable to the reduction observed when ORF66 translation was prevented by stop codon insertions in pOka66S. In contrast, infection of T-cell xenografts with pOka66G102A was associated with a significant decrease in infectious virus production equivalent to the impaired T-cell tropism found with pOka66S infection of T-cell xenografts in vivo. Disrupting kinase activity with the G102A mutation did not alter IE62 cytoplasmic localization in VZV-infected T cells, suggesting that decreased T-cell tropism is due to other ORF66 protein functions. The G102A mutation reduced the antiapoptotic effects of VZV infection of T cells. These experiments indicate that the T-cell tropism of VZV depends upon intact ORF66 protein kinase function.


Assuntos
Herpes Zoster/virologia , Herpesvirus Humano 3/fisiologia , Proteínas Quinases/fisiologia , Linfócitos T/virologia , Animais , Linhagem Celular , Herpesvirus Humano 3/patogenicidade , Humanos , Camundongos , Camundongos SCID , Fases de Leitura Aberta/fisiologia , Virulência , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA