Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Infect Genet Evol ; 96: 105096, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34601094

RESUMO

Human metapneumovirus (HMPV), an unsegmented negative-strand RNA virus, is the second most detected respiratory pathogen and one of the leading causes of respiratory illness in infants and immunodeficient individuals. HMPV infection of permissive cells in culture triggers a transient IFN response, which is efficiently suppressed later in infection. We report that two structural glycoproteins of the virus - namely G (Glycoprotein) and SH (Small Hydrophobic) - suppress the type I interferon (IFN) response in cell culture. This is manifested by inhibition of diverse steps of IFN induction and response, such as phosphorylation and nuclear translocation of IFN regulatory factor-3 and -7 (IRF3, IRF7), major transcription factors of the IFN gene. Furthermore, HMPV suppresses the cellular response to IFN by inhibiting the phosphorylation of STAT1 (Signal Transducer and Activator of Transcription 1), required for the induction of IFN-stimulated genes that act as antivirals. Site-directed mutagenesis revealed an important role of critical cysteine (Cys) residues in the Cys-rich carboxy terminal region of the SH protein in IFN suppression, whereas for G, the ectodomain plays a role. These results shed light on the mechanism of IFN suppression by HMPV, and may also offer avenues for new antiviral approaches in the future.


Assuntos
Glicoproteínas/metabolismo , Imunidade Inata , Infecções por Paramyxoviridae/imunologia , Proteínas Virais/metabolismo , Células A549 , Humanos , Metapneumovirus/fisiologia , Infecções por Paramyxoviridae/virologia
2.
Front Immunol ; 10: 2411, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31681300

RESUMO

DNA vaccines have great potential to control infectious disease, particularly those caused by intracellular organisms. They are inexpensive to produce and can be quickly modified to combat emerging infectious threats, but often fail to generate a strong immunologic response limiting enthusiasm for their use in humans and animals. To improve the immunogenic response, we developed a DNA vaccine in which the F protein ectodomain of Respiratory Syncytial Virus (RSV-F) was covalently linked to specific antigens of interest. The presence of the RSV-F ectodomain allowed secretion of the translated fusion product out of the originally transfected cells followed by its active binding to adjacent cells. This allowed the targeting of a greater number of cells than those originally transfected, enhancing both humoral and cytotoxic immune responses against the expressed antigen(s). We developed an engrafted mouse model that used antigen-expressing tumor cells to assess the in vivo cytotoxic immune response to specific antigens. We then used this model to demonstrate that a DNA vaccine in which the RSV-F ectodomain is fused to two antigens expressed by Burkholderia pseudomallei, the intracellular gram-negative organism that causes melioidosis, generated a stronger cytotoxic response than a DNA vaccine that lacked the RSV-F sequence while still generating a robust humoral response.


Assuntos
Proteínas de Bactérias , Burkholderia pseudomallei , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sinciciais Respiratórios , Vacinas de DNA , Proteínas Virais de Fusão , Fatores de Virulência , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Domínios Proteicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia
3.
Hum Vaccin Immunother ; 11(12): 2904-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26307915

RESUMO

Respiratory Syncytial Virus is a leading cause of bronchiolitis and pneumonia in infants, the elderly and individuals with compromised immune systems. Despite decades of research, there is currently no available vaccine for RSV. Our group has previously demonstrated that intranasal immunization of mice with RSV inactivated by and adjuvanted with W805EC nanoemulsion elicits robust humoral and cellular immune responses, resulting in protection against RSV infection. This protection was achieved without the induction of airway hyper-reactivity or a Th2-skewed immune response. The cotton rat Sigmodon hispidus has been used for years as an excellent small animal model of RSV disease. Thus, we extended these rodent studies to the more permissive cotton rat model. Intranasal immunization of the nanoemulsion-adjuvanted RSV vaccines induced high antibody titers and a robust Th1-skewed cellular response. Importantly, vaccination provided sterilizing cross-protective immunity against a heterologous RSV challenge and did not induce marked or severe histological effects or eosinophilia in the lung after viral challenge. Overall, these data demonstrate that nanoemulsion-formulated whole RSV vaccines are both safe and effective for immunization in multiple animal models.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Emulsões/uso terapêutico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico , Sigmodontinae/imunologia , Vacinas de Produtos Inativados/imunologia , Administração Intranasal , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Bronquiolite/imunologia , Bronquiolite/prevenção & controle , Bronquiolite/virologia , Proteção Cruzada/imunologia , Feminino , Pulmão/patologia , Pulmão/virologia , Pneumonia Viral/imunologia , Pneumonia Viral/prevenção & controle , Pneumonia Viral/virologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Sigmodontinae/virologia , Células Th1/imunologia , Vacinação , Proteínas Virais de Fusão/imunologia , Carga Viral/imunologia
4.
BMC Res Notes ; 8: 379, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26306622

RESUMO

BACKGROUND: Transient production of gamma-retroviruses, including self-inactivating (SIN) retroviruses, is a common method for rapidly generating virus capable of gene delivery. Stable (continuous) production of virus is preferable to transient production for clinical and biotechnology purposes, however, because it allows for significant quantities of a uniform virus to be generated over a prolonged period of time, thus allowing for longitudinal functional studies and quality analysis. Unfortunately, stable production of SIN retroviruses is difficult to achieve. RESULTS: We describe a novel method to rapidly and cost-effectively create packaging cells capable of continuously producing self-inactivating gamma-retroviruses. We imbedded the SIN proviral construct into a minimal piggyBac transposon vector and then integrated the hybrid vector into packaging cells that already stably expressed the viral gag-pro-pol and envelope genes. Cells that stably produced self-inactivating gamma-retroviruses could be identified (and purified) as early as 3 weeks after initial transfection; these cells produced virus for at least 9 weeks without a decline in titer. CONCLUSIONS: This viral-minimal piggyBac hybrid vector allowed for the rapid generation and purification of packaging cells capable of stably producing self-inactivated gamma-retroviruses. This method can be applied to the large-scale production of viruses for use in research, biotechnology, and potentially, clinical trials.


Assuntos
Elementos de DNA Transponíveis , Vetores Genéticos , Retroviridae/fisiologia , Células HEK293 , Humanos , Retroviridae/genética
5.
Hum Vaccin Immunother ; 10(3): 615-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24326268

RESUMO

Respiratory Syncytial Virus (RSV) is a ubiquitous virus that infects almost all people by age two and is a major source of respiratory illness in infants, the elderly and others with compromised immune systems. Currently there is no available vaccine. Prior efforts using formalin-inactivated RSV (FI-RSV) were associated with enhanced respiratory disease upon viral exposure following clinical vaccine trials. Several researchers and pharmaceutical companies have utilized vector-associated live attenuated RSV vaccines in pre-clinical and clinical studies. Another attractive approach, however, is a subunit vaccine which would be easier to produce and quality control. Our group has previously demonstrated in a murine model of infection that intranasal immunization with nanoemulsion-inactivated and adjuvanted RSV induces humoral and cellular immune responses, resulting in protection against RSV infection. The present studies characterize the immune responses elicited by intranasal RSV F protein adjuvanted with nanoemulsion. Intranasal application of nanoemulsion adjuvanted F protein induced a rapid and robust systemic and mucosal antibody response, as well as protection against subsequent RSV challenge. Importantly, RSV challenge in immunized animals did not elicit airway hyper-reactivity, a Th2-skewed immune response or immunopathology associated with hypersensitivity reactions with formalin-inactivated vaccine. These results suggest that RSV F protein adjuvanted with nanoemulsion may be a good mucosal vaccine candidate. Formulating RSV F protein in nanoemulsion creates a well-defined and well-controlled vaccine that can be delivered intranasally to induce T cell mediated immunity without inducing enhanced disease associated with the mouse model of FI-RSV vaccination and infection.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Imunização/métodos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sinciciais Respiratórios/imunologia , Administração Intranasal , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Sangue/imunologia , Modelos Animais de Doenças , Emulsões/administração & dosagem , Feminino , Imunidade nas Mucosas , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vírus Sinciciais Respiratórios/isolamento & purificação , Células Th2/imunologia , Resultado do Tratamento , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Virais de Fusão/imunologia
6.
Am J Physiol Lung Cell Mol Physiol ; 297(3): L538-45, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19561138

RESUMO

Using adapted retroviruses for gene delivery is a modern and powerful tool in biological research as well as a promising approach for gene therapy. An important limitation for the extensive use of retroviral vectors is the low infection rate in target cells such as pulmonary vascular endothelial cells due to the insufficient infectivity of standard retrovirus supernatants that can only be overcome by complicated methods of virus concentration. This paper describes two easy methods to augment target cell infectivity, first by increasing the retroviral titer in the medium collected from packaging cells by stimulation of viral propagation with dexamethasone, and second, by increasing target cell sensitivity to retroviral infection by the glucocorticoid receptor antagonist, mifepristone. Using this method, we increased the infectivity of pulmonary microvascular endothelial cells from 16% to 85%. We demonstrate that mifepristone increased the susceptibility of target cells to retroviruses without increasing the viral titer of the supernatant. Dexamethasone, but not mifepristone, increased expression of delivered proteins such as GFP that are important for early identification of infected cells. Each, or both step(s), may be included in a standard protocol for retrovirus propagation and infection of target cells.


Assuntos
Dexametasona/farmacologia , Técnicas de Transferência de Genes , Mifepristona/farmacologia , Retroviridae/efeitos dos fármacos , Retroviridae/patogenicidade , Animais , Meios de Cultivo Condicionados , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/virologia , Hormônios/metabolismo , Regiões Promotoras Genéticas/genética , Ratos , Receptores de Glucocorticoides/antagonistas & inibidores , Retroviridae/fisiologia , Soro , Frações Subcelulares/metabolismo , Sequências Repetidas Terminais/genética , Proteínas Virais/metabolismo
7.
J Virol ; 82(16): 7977-87, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550659

RESUMO

The La antigen (SS-B) associates with a wide variety of cellular and viral RNAs to affect gene expression in multiple systems. We show that La is the major cellular protein found to be associated with the abundant 44-nucleotide viral leader RNA (leRNA) early after infection with respiratory syncytial virus (RSV), a nonsegmented negative-strand RNA virus. Consistent with this, La redistributes from the nucleus to the cytoplasm in RSV-infected cells. Upon RNA interference knockdown of La, leRNA is redirected to associate with the RNA-binding protein RIG-I, a known activator of interferon (IFN) gene expression, and this is accompanied by the early induction of IFN mRNA. These results suggest that La shields leRNA from RIG-I, abrogating the early viral activation of type I IFN. We mapped the leRNA binding function to RNA recognition motif 1 of La and showed that while wild-type La greatly enhanced RSV growth, a La mutant defective in RSV leRNA binding also did not support RSV growth. Comparative studies of RSV and Sendai virus and the use of IFN-negative Vero cells indicated that La supports the growth of nonsegmented negative-strand RNA viruses by both IFN suppression and a potentially novel IFN-independent mechanism.


Assuntos
Autoantígenos/fisiologia , RNA Helicases DEAD-box/química , RNA Viral , Ribonucleoproteínas/fisiologia , Motivos de Aminoácidos , Animais , Autoantígenos/química , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Clonagem Molecular , Citoplasma/metabolismo , Proteína DEAD-box 58 , Humanos , Modelos Genéticos , Interferência de RNA , Receptores Imunológicos , Ribonucleoproteínas/química , Células Vero , Replicação Viral , Antígeno SS-B
8.
J Mol Med (Berl) ; 86(3): 313-22, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18193184

RESUMO

MicroRNAs (miRNAs) are endogenous noncoding RNAs that down-regulate gene expression by promoting cleavage or translational arrest of target mRNAs. While most miRNAs are transcribed from their own dedicated genes, some map to introns of 'host' transcripts, the biological significance of which remains unknown. Here, we show that prostate cells are naturally devoid of EGF-like domain 7 (Egfl7) transcripts and hence also deficient in a miRNA, miR-126*, generated from splicing and processing of its ninth intron. Use of recombinant and synthetic miRNAs or a specific antagomir established a role of miR-126* in silencing prostein in non-endothelial cells. We mapped two miR-126*-binding sites in the 3'UTR of the prostein mRNA required for translational repression. Transfection of synthetic miR-126* into prostate cancer LNCaP cells strongly reduced the translation of prostein. Interestingly, loss of prostein correlated with reduction of LNCaP cell migration and invasion. Thus, the robust expression of prostein protein in the prostate cells results from a combination of transcriptional activation of the prostein gene and absence of intronic miRNA-126* due to the prostate-specific repression of the Egfl7 gene. We conclude that intronic miRNAs from tissue-specific transcripts, or their natural absence, make cardinal contributions to cellular gene expression and phenotype. These findings also open the door to tissue-specific miRNA therapy.


Assuntos
Endotélio Vascular/metabolismo , Íntrons/genética , Proteínas de Membrana/genética , MicroRNAs/genética , Proteínas de Neoplasias/genética , Neoplasias da Próstata/patologia , Biossíntese de Proteínas , Regiões 3' não Traduzidas , Sequência de Bases , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Masculino , Proteínas de Membrana/metabolismo , Modelos Genéticos , Dados de Sequência Molecular , Invasividade Neoplásica , Especificidade de Órgãos , Splicing de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno
9.
J Virol ; 81(4): 1786-95, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17151097

RESUMO

The two nonstructural (NS) proteins NS1 and NS2 of respiratory syncytial virus (RSV) are abundantly expressed in the infected cell but are not packaged in mature progeny virions. We found that both proteins were expressed early in infection, whereas the infected cells underwent apoptosis much later. Coincident with NS protein expression, a number of cellular antiapoptotic factors were expressed or activated at early stages, which included NF-kappaB and phosphorylated forms of protein kinases AKT, phosphoinositide-dependent protein kinase, and glycogen synthase kinase. Using specific short interfering RNAs (siRNAs), we achieved significant knockdown of one or both NS proteins in the infected cell, which resulted in abrogation of the antiapoptotic functions and led to early apoptosis. NS-dependent suppression of apoptosis was observed in Vero cells that are naturally devoid of type I interferons (IFN). The siRNA-based results were confirmed by the use of NS-deleted RSV mutants. Early activation of epidermal growth factor receptor (EGFR) in the RSV-infected cell did not require NS proteins. Premature apoptosis triggered by the loss of NS or by apoptosis-promoting drugs caused a severe reduction of RSV growth. Finally, recombinantly expressed NS1 and NS2, individually and together, reduced apoptosis by tumor necrosis factor alpha, suggesting an intrinsic antiapoptotic property of both. We conclude that the early-expressed nonstructural proteins of RSV boost viral replication by delaying the apoptosis of the infected cell via a novel IFN- and EGFR-independent pathway.


Assuntos
Apoptose , Interferons/fisiologia , NF-kappa B/fisiologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Regulação para Baixo , Receptores ErbB/fisiologia , Humanos , Complexo de Endopeptidases do Proteassoma , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Vírus Sinciciais Respiratórios/fisiologia , Células Vero , Replicação Viral
10.
J Virol ; 81(2): 783-90, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17050596

RESUMO

Respiratory syncytial virus (RSV) is the foremost respiratory pathogen in newborns and claims millions of lives annually. However, there has been no methodical study of the pathway(s) of entry of RSV or its interaction with nonrespiratory tissues. We and others have recently established a significant association between allergic conjunctivitis and the presence of RSV in the eye. Here we adopt a BALB/c mouse model and demonstrate that when instilled in the live murine eye, RSV not only replicated robustly in the eye but also migrated to the lung and produced a respiratory disease that is indistinguishable from the standard, nasally acquired RSV disease. Ocularly applied synthetic anti-RSV small interfering RNA prevented infection of the eye as well as the lung. RSV infection of the eye activated a plethora of ocular cytokines and chemokines with profound relevance to inflammation of the eye. Anticytokine treatments in the eye reduced ocular inflammation but had no effect on viral growth in both eye and lung, demonstrating a role of the cytokine response in ocular pathology. These results establish the eye as a major gateway of respiratory infection and a respiratory virus as a bona fide eye pathogen, thus offering novel intervention and treatment options.


Assuntos
Conjuntivite Viral/transmissão , Conjuntivite Viral/virologia , Olho/virologia , Pulmão/virologia , Infecções por Vírus Respiratório Sincicial/transmissão , Vírus Sincicial Respiratório Humano/patogenicidade , Infecções Respiratórias/virologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Conjuntivite Viral/terapia , Modelos Animais de Doenças , Olho/imunologia , Feminino , Humanos , Interleucina-1alfa/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Infecções por Vírus Respiratório Sincicial/terapia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/metabolismo , Vírus Sincicial Respiratório Humano/fisiologia , Infecções Respiratórias/terapia , Resultado do Tratamento , Fator de Necrose Tumoral alfa/imunologia , Replicação Viral
11.
Mol Biochem Parasitol ; 133(2): 297-310, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14698441

RESUMO

Biochemical evidence revealed protein tyrosine kinase and phosphatase activities in the human malarial parasite Plasmodium falciparum, a member of the Apicomplexa. A novel cDNA sequence of a dual-specificity phosphatase was identified in both sexual and asexual stages of P. falciparum, and named PfYVH1, since the predicted primary structure of the 278-amino acid polypeptide showed significant similarity to the human and yeast YVH1 phosphatases. The N-terminal half of PfYVH1 contained a conserved tyrosine phosphatase catalytic domain within a dual-specificity phosphatase domain. The C-terminal region, consisting of one histidine and eight cysteines, represented a zinc-binding domain with a potentially unconventional architecture. Recombinant PfYVH1 contained 2mol of zinc per mol protein and dephosphorylated both phosphoserine and phosphotyrosine residues. Mutation of specific Cys residues in the putative zinc finger region abolished zinc binding and drastically reduced phosphatase activity, suggesting an allosteric role of zinc in catalysis. PfYVH1 was expressed in essentially all erythrocytic stages of the parasite, and shuttled between the nucleus and the cytoplasm in a stage-specific manner. A Plasmodium ortholog of the nuclear pescadillo protein (PfPES) was also characterized and shown to interact with PfYVH1, thus implicating PfYVH1 in the regulation of parasitic development. PfYVH1 represents the first dual-specificity zinc-finger phosphatase characterized in the protozoan kingdom.


Assuntos
Proteínas Nucleares/metabolismo , Plasmodium falciparum/enzimologia , Proteínas Tirosina Fosfatases/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Domínio Catalítico , Núcleo Celular/metabolismo , Clonagem Molecular , Citoplasma/metabolismo , DNA de Protozoário/química , DNA de Protozoário/isolamento & purificação , Fosfatases de Especificidade Dupla , Éxons , Íntrons , Dados de Sequência Molecular , Mutação , Sinais de Localização Nuclear , Proteínas Nucleares/química , Proteínas Nucleares/genética , Plasmodium falciparum/genética , Ligação Proteica , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência , Zinco/metabolismo , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA