Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biol Eng ; 13: 77, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31649751

RESUMO

BACKGROUND: The baculovirus (BV) Autographa californica multiple nuclear polyhedrosis virus has been used in numerous protein expression systems because of its ability to infect insect cells and serves as a useful vaccination vector with several benefits, such as its low clinical risks and posttranslational modification ability. We recently reported that dendritic cells (DCs) infected with BV stimulated antitumor immunity. The recombinant BV (rBV) also strongly stimulated peptide-specific T-cells and antitumor immunity. In this study, the stimulation of an immune response against EG7-OVA tumors in mice by a recombinant baculovirus-based combination vaccine expressing fragment C-ovalbumin (FrC-OVA-BV; rBV) was evaluated. RESULTS: We constructed an rBV expressing fragment C (FrC) of tetanus toxin containing a promiscuous MHC II-binding sequence and a p30-ovalbumin (OVA) peptide that functions in the MHC I pathway. The results showed that rBV activated the CD8+ T-cell-mediated response much more efficiently than the wild-type BV (wtBV). Experiments with EG7-OVA tumor mouse models showed that rBV significantly decreased tumor volume and increased survival compared with those in the wild-type BV or FrC-OVA DNA vaccine groups. In addition, a significant antitumor effect of classic prophylactic or therapeutic vaccinations was observed for rBV against EG7-OVA-induced tumors compared with that in the controls. CONCLUSION: Our findings showed that FrC-OVA-BV (rBV) induced antitumor immunity, paving the way for its use in BV immunotherapy against malignancies.

2.
J Biol Chem ; 291(9): 4754-62, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26694617

RESUMO

Here, we identify ATP1B3 and fibrillin-1 as novel BST-2-binding proteins. ATP1B3 depletion in HeLa cells (BST-2-positive cells), but not 293T cells (BST-2-negative cells), induced the restriction of HIV-1 production in a BST-2-dependent manner. In contrast, fibrillin-1 knockdown reduced HIV-1 production in 293T and HeLa cells in a BST-2-independent manner. Moreover, NF-κB activation was enhanced by siATP1B3 treatment in HIV-1- and HIV-1ΔVpu-infected HeLa cells. In addition, ATP1B3 silencing induced high level BST-2 expression on the surface of HeLa cells. These results indicate that ATP1B3 is a co-factor that accelerates BST-2 degradation and reduces BST-2-mediated restriction of HIV-1 production and NF-κB activation.


Assuntos
Antígenos CD/metabolismo , HIV-1/fisiologia , Proteínas dos Microfilamentos/metabolismo , NF-kappa B/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/metabolismo , Antígenos CD/química , Antígenos CD/genética , Linhagem Celular , Fibrilina-1 , Fibrilinas , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , NF-kappa B/agonistas , NF-kappa B/genética , NF-kappa B/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Proteólise , Interferência de RNA , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/genética , Técnicas do Sistema de Duplo-Híbrido , Tropismo Viral , Replicação Viral
3.
Cancer Biol Ther ; 16(11): 1616-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26391871

RESUMO

To develop a potent cancer vaccine, it is important to study how to prepare highly immunogenic antigens and to identify the most appropriate adjuvants for the antigens. Here we show that a tumor lysate works as an effective antigen to prime CD4(+) T-cell help when baculovirus is employed as an adjuvant. When immunized intradermally with the combination (BLP) of baculovirus, a CT26 tumor lysate, and a cytotoxic T-cell epitope peptide before a tumor challenge, 60% of mice rejected tumors. In contrast, all mice vaccinated with baculovirus plus a tumor lysate (BL) developed tumors. In addition, flow cytometry showed that tumor-specific, interferon γ-producing CD8(+) cytotoxic T lymphocytes (CTLs) were robustly activated by intradermal immunization with BLP. When BLP was administered therapeutically to tumor-bearing mice, antitumor efficacy was better compared to BL. The established tumor was completely eradicated in 50-60% of BLP-treated mice, and induction of tumor-specific CTLs was observed, suggesting that the antitumor efficacy of BLP is mediated by CD8(+) T cells. Numerous CD4(+) T cells infiltrated the tumors of BLP-treated mice, whereas the antitumor effect of BLP almost disappeared after removal of the tumor lysate from BLP or after depletion of BLP-immunized mice of CD4(+) T cells. Thus, the combination of a peptide, lysate, and baculovirus provides stronger antitumor immunity than does a peptide plus baculovirus or a lysate plus baculovirus; effectiveness of BLP is determined by functioning of CD4(+) T cells stimulated with a tumor lysate.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Neoplasias/terapia , Animais , Baculoviridae/imunologia , Vacinas Anticâncer/administração & dosagem , Extratos Celulares/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/imunologia , Feminino , Humanos , Imunização , Injeções Intradérmicas , Linfócitos do Interstício Tumoral/imunologia , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias/imunologia
4.
AIDS Res Hum Retroviruses ; 31(10): 1023-31, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26178669

RESUMO

Acquired immune deficiency syndrome (AIDS) is mainly caused by infection with human immunodeficiency virus-1 (HIV-1) and still poses a global threat for which we lack a protective or therapeutic vaccine. Dendritic cells (DCs) play a major role in the onset of HIV infection, providing one of the primary sites of HIV replication, and also act as viral reservoirs in vivo. Previous studies have shown that baculovirus (BV) induces strong host immune responses against infections and malignancies. In this study, we infected human monocyte-derived DCs with recombinant BV (AcCAG-gag) and showed that AcCAG-gag-infected human DCs underwent maturation and produced interferon alpha and other proinflammatory cytokines accompanied by increases in the mRNA and protein expression levels of APOBEC3 (A3A, A3F, and A3G), proteins associated with the inhibition of HIV-1 replication. Surprisingly, HIV-1 inhibition is also observed in human DCs infected with a wild-type BV, as determined by the production of inflammatory cytokines, the expression of A3, and a reduction in the p24 level. Our findings outline the mechanism underlying the inhibition of HIV-1 in BV-infected human DCs and pave the way for the use of BV as an effective tool for immunotherapy against HIV-1.


Assuntos
Baculoviridae/crescimento & desenvolvimento , Células Dendríticas/virologia , HIV-1/crescimento & desenvolvimento , Imunidade Inata , Interferência Viral , Células Cultivadas , Humanos
5.
Antiviral Res ; 117: 1-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25666760

RESUMO

Previous studies have demonstrated that cyclopentenone prostaglandins (cyPGs) inhibit the replication of a wide variety of DNA and RNA viruses in different mammalian cell types. We investigated a new role for prostaglandin A1 (PGA1) in the inhibition of hepatitis C virus (HCV)-IRES-mediated translation. PGA1 exhibited dose-dependent inhibitory effects on HCV translation in HCV replicon cells. Furthermore, repetitive PGA1 treatment demonstrated the potential to safely induce the suppression of HCV translation. We also validated a new role for PGA1 in the inhibition of HCV-IRES-mediated translation by targeting cellular translation factors, including the small ribosomal subunit (40S) and eukaryotic initiation factors (eIFs). In pull-down assays, biotinylated PGA1 co-precipitated with the entire HCV IRES RNA/eIF3-40S subunit complex. Moreover, the interactions between PGA1 and the elongation factors and ribosomal subunit were dependent upon HCV IRES RNA binding, and the PGA1/HCV IRES RNA/eIF3-40S subunit complex inhibited HCV-IRES-mediated translation. The novel mechanism revealed in this study may aid in the search for more effective anti-HCV drugs.


Assuntos
Hepacivirus/crescimento & desenvolvimento , Hepacivirus/genética , Hepacivirus/metabolismo , Prostaglandinas A/metabolismo , Prostaglandinas A/farmacologia , Replicon/efeitos dos fármacos , Subunidades Ribossômicas Menores/efeitos dos fármacos , Linhagem Celular Tumoral , Fator de Iniciação 3 em Eucariotos/metabolismo , Humanos , Sítios Internos de Entrada Ribossomal , Biossíntese de Proteínas/efeitos dos fármacos , RNA Viral/genética , Replicon/fisiologia , Subunidades Ribossômicas Menores/metabolismo
6.
J Biol Chem ; 289(38): 26226-26238, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25100733

RESUMO

Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide 1 (APOBEC1) is an intestine-specific RNA-binding protein. However, inflammation or exposure to DNA-damaging agents can induce ectopic APOBEC1 expression, which can result in hepatocellular hyperplasia in animal models. To identify its RNA targets, FLAG-tagged APOBEC1 was immunoprecipitated from transfected HuH7.5 hepatocellular carcinoma cells and analyzed using DNA microarrays. The interleukin-8 (IL8) mRNA was the most abundant co-precipitated RNA. Exogenous APOBEC1 expression increased IL8 production by extending the half-life of the IL8 mRNA. A cluster of AU-rich elements in the 3'-UTR of IL8 was essential to the APOBEC1-mediated increase in IL8 production. Notably, IL8 mRNA did not co-immunoprecipitate with APOBEC1 from lysates of other cell types at appreciable levels; therefore, other factors may enhance the association between APOBEC1 and IL8 mRNA in a cell type-specific manner. A yeast two-hybrid analysis and siRNA screen were used to identify proteins that enhance the interaction between APOBEC1 and IL8 mRNA. Heterogeneous nuclear ribonucleoprotein Q (hnRNPQ) was essential to the APOBEC1/IL8 mRNA association in HuH7.5 cells. Of the seven hnRNPQ isoforms, only hnRNPQ6 enabled APOBEC1 to bind to IL8 mRNA when overexpressed in HEK293 cells, which expressed the lowest level of endogenous hnRNPQ6 among the cell types examined. The results of a reporter assay using a luciferase gene fused to the IL8 3'-UTR were consistent with the hypothesis that hnRNPQ6 is required for APOBEC1-enhanced IL8 production. Collectively, these data indicate that hnRNPQ6 promotes the interaction of APOBEC1 with IL8 mRNA and the subsequent increase in IL8 production.


Assuntos
Citidina Desaminase/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Interleucina-8/genética , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas , Desaminase APOBEC-1 , Linhagem Celular Tumoral , Células HEK293 , Meia-Vida , Humanos , Interleucina-8/metabolismo , Mutação Puntual , Ligação Proteica , Isoformas de Proteínas/metabolismo , Estabilidade de RNA , Transcrição Gênica , Ativação Transcricional , Técnicas do Sistema de Duplo-Híbrido
7.
PLoS One ; 9(4): e94460, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24718268

RESUMO

Most of experiments for HCV infection have been done using lytic infection systems, in which HCV-infected cells inevitably die. Here, to elucidate metabolic alteration in HCV-infected cells in a more stable condition, we established an HCV-persistently-infected cell line, designated as HPI cells. This cell line has displayed prominent steatosis and supported HCV infection for more than 2 years, which is the longest ever reported. It enabled us to analyze metabolism in the HCV-infected cells integrally combining metabolomics and expression arrays. It revealed that rate-limiting enzymes for biosynthesis of cholesterol and fatty acids were up-regulated with actual increase in cholesterol, desmosterol (cholesterol precursor) and pool of fatty acids. Notably, the pentose phosphate pathway was facilitated with marked up-regulation of glucose-6-phosphate dehydrogenase, a rete-limiting enzyme, with actual increase in NADPH. In its downstream, enzymes for purine synthesis were also up-regulated resulting in increase of purine. Contrary to common cancers, the TCA cycle was preferentially facilitated comparing to glycolysis pathway with a marked increase of most of amino acids. Interestingly, some genes controlled by nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a master regulator of antioxidation and metabolism, were constitutively up-regulated in HPI cells. Knockdown of Nrf2 markedly reduced steatosis and HCV infection, indicating that Nrf2 and its target genes play important roles in metabolic alteration and HCV infection. In conclusion, HPI cell is a bona fide HCV-persistently-infected cell line supporting HCV infection for years. This cell line sustained prominent steatosis in a hypermetabolic status producing various metabolites. Therefore, HPI cell is a potent research tool not only for persistent HCV infection but also for liver metabolism, overcoming drawbacks of the lytic infection systems.


Assuntos
Fígado Gorduroso/metabolismo , Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepatite C/virologia , Aminoácidos/metabolismo , Vias Biossintéticas , Linhagem Celular , Colesterol/metabolismo , Células Clonais , Meios de Cultura , Desmosterol/metabolismo , Ácidos Graxos/metabolismo , Técnicas de Silenciamento de Genes , Hepatite C/patologia , Humanos , Espaço Intracelular/metabolismo , Gotículas Lipídicas/metabolismo , Metabolômica , NADP/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Nucleotídeos/metabolismo , Transcrição Gênica , Ativação Transcricional/genética , Triglicerídeos/metabolismo , Proteínas Virais/metabolismo
8.
Int J Oncol ; 43(6): 2023-30, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24101126

RESUMO

Although tumor lysate contains all the potential helper and killer epitopes capable of stimulating T cells, it is difficult to use as a cancer vaccine because it suppresses dendritic cell (DC) function. We report that wild-type baculovirus possesses an adjuvant effect to improve the immunogenicity of tumor lysate. When mice were administered CT26 tumor cell lysate combined with baculovirus intradermally, antitumor immunity was induced and rejection of CT26 tumor growth was observed in 40% of the immunized mice. In contrast, such antitumor immunity was not elicited in mice inoculated with tumor cell lysate or baculovirus alone. In tumor-bearing mice, which had previously received the combined baculovirus and tumor lysate vaccine, the established tumors were completely eradicated by administering a booster dose of the combined vaccine. This antitumor effect was attributed to tumor-specific T cell immunity mediated primarily by CD8⁺ T cells. Baculovirus also strongly activated DCs loaded with tumor lysate. Increased interleukin (IL)-6 and IL-12p70 production were also observed in DCs co-cultured with tumor cell lysate and baculovirus. Our study demonstrates that combined baculovirus and tumor lysate vaccine can effectively stimulate DCs to induce acquired antitumor immunity.


Assuntos
Baculoviridae/imunologia , Vacinas Anticâncer/imunologia , Neoplasias/imunologia , Neoplasias/prevenção & controle , Adjuvantes Farmacêuticos/farmacologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Feminino , Interleucina-12/biossíntese , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Vacinação
9.
Jpn J Infect Dis ; 66(4): 276-83, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23883836

RESUMO

Influenza is a global public health problem that causes a serious respiratory disease. Influenza virus frequently undergoes amino acid substitutions, which result in the emergence of drug-resistant viruses. To control influenza viruses that are resistant to currently available drugs, it is essential to develop new antiviral drugs with a novel molecular target. Here, we report that cyclosporin A (CsA) inhibits the propagation of influenza virus in A549 cells by interfering with a late event in the virus life cycle. CsA did not affect adsorption, internalization, viral RNA replication, or synthesis of viral proteins in A549 cells, but inhibited the step(s) after viral protein synthesis, such as assembly or budding. In addition, siRNA-mediated knockdown of the expression of the major CsA targets, namely cyclophilin A (CypA), cyclophilin B (CypB), and P-glycoprotein (Pgp), did not inhibit influenza virus propagation. These results suggest that CsA inhibits virus propagation by mechanism(s) independent of the inhibition of the function of CypA, CypB, and Pgp. CsA may target an unknown molecule that works as a positive regulator in the propagation of influenza virus. Our findings would contribute to the development of a novel anti-influenza virus therapy and clarification of the regulatory mechanism of influenza virus multiplication.


Assuntos
Antivirais/farmacologia , Ciclosporina/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/fisiologia , Montagem de Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Linhagem Celular , Células Epiteliais/virologia , Humanos
10.
J Virol ; 87(14): 8169-78, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23678168

RESUMO

Inflammatory cytokines and chemokines play important roles in inflammation during viral infection. Hepatitis C virus (HCV) is a hepatotropic RNA virus that is closely associated with chronic liver inflammation, fibrosis, and hepatocellular carcinoma. During the progression of HCV-related diseases, hepatic stellate cells (HSCs) contribute to the inflammatory response triggered by HCV infection. However, the underlying molecular mechanisms that mediate HSC-induced chronic inflammation during HCV infection are not fully understood. By coculturing HSCs with HCV-infected hepatocytes in vitro, we found that HSCs stimulated HCV-infected hepatocytes, leading to the expression of proinflammatory cytokines and chemokines such as interleukin-6 (IL-6), IL-8, macrophage inflammatory protein 1α (MIP-1α), and MIP-1ß. Moreover, we found that this effect was mediated by IL-1α, which was secreted by HSCs. HCV infection enhanced production of CCAAT/enhancer binding protein (C/EBP) ß mRNA, and HSC-dependent IL-1α production contributed to the stimulation of C/EBPß target cytokines and chemokines in HCV-infected hepatocytes. Consistent with this result, knockdown of mRNA for C/EBPß in HCV-infected hepatocytes resulted in decreased production of cytokines and chemokines after the addition of HSC conditioned medium. Induction of cytokines and chemokines in hepatocytes by the HSC conditioned medium required a yet to be identified postentry event during productive HCV infection. The cross talk between HSCs and HCV-infected hepatocytes is a key feature of inflammation-mediated, HCV-related diseases.


Assuntos
Quimiocinas/metabolismo , Citocinas/metabolismo , Células Estreladas do Fígado/metabolismo , Hepatite C/imunologia , Hepatócitos/metabolismo , Inflamação/imunologia , Receptor Cross-Talk/fisiologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular , Quimiocinas/imunologia , Citocinas/imunologia , Primers do DNA/genética , Técnicas de Silenciamento de Genes , Hepatite C/complicações , Humanos , Inflamação/etiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real
11.
PLoS One ; 8(3): e59892, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555825

RESUMO

Influenza is a serious public health problem that causes a contagious respiratory disease. Vaccination is the most effective strategy to reduce transmission and prevent influenza. In recent years, cell-based vaccines have been developed with continuous cell lines such as Madin-Darby canine kidney (MDCK) and Vero. However, wild-type influenza and egg-based vaccine seed viruses will not grow efficiently in these cell lines. Therefore, improvement of virus growth is strongly required for development of vaccine seed viruses and cell-based influenza vaccine production. The aim of our research is to develop novel MDCK cells supporting highly efficient propagation of influenza virus in order to expand the capacity of vaccine production. In this study, we screened a human siRNA library that involves 78 target molecules relating to three major type I interferon (IFN) pathways to identify genes that when knocked down by siRNA lead to enhanced production of influenza virus A/Puerto Rico/8/1934 in A549 cells. The siRNAs targeting 23 candidate genes were selected to undergo a second screening pass in MDCK cells. We examined the effects of knockdown of target genes on the viral production using newly designed siRNAs based on sequence analyses. Knockdown of the expression of a canine gene corresponding to human IRF7 by siRNA increased the efficiency of viral production in MDCK cells through an unknown process that includes the mechanisms other than inhibition of IFN-α/ß induction. Furthermore, the viral yield greatly increased in MDCK cells stably transduced with the lentiviral vector for expression of short hairpin RNA against IRF7 compared with that in control MDCK cells. Therefore, we propose that modified MDCK cells with lower expression level of IRF7 could be useful not only for increasing the capacity of vaccine production but also facilitating the process of seed virus isolation from clinical specimens for manufacturing of vaccines.


Assuntos
Fator Regulador 7 de Interferon/genética , Células Madin Darby de Rim Canino/virologia , Orthomyxoviridae/fisiologia , Cultura de Vírus , Replicação Viral , Animais , Linhagem Celular Tumoral , Cães , Biblioteca Gênica , Humanos , Vacinas contra Influenza/biossíntese , Interferon Tipo I/metabolismo , Plasmídeos/metabolismo , RNA Interferente Pequeno/genética
12.
PLoS One ; 7(12): e51393, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251516

RESUMO

The unspliced human immunodeficiency virus type 1 (HIV-1) RNAs are translated as Gag and Gag-Pol polyproteins or packaged as genomes into viral particles. Efficient translation is necessary before the transition to produce infective virions. The viral protein Rev exports all intron-containing viral RNAs; however, it also appears to enhance translation. Cellular microRNAs target cellular and viral mRNAs to silence their translation and enrich them at discrete cytoplasmic loci that overlap with the putative interim site of Gag and the genome. Here, we analyzed how Rev-mediated transport and the splicing status of the mRNA influenced the silencing status imposed by microRNA. Through identification and mutational analysis of the silencing sites in the HIV-1 genome, we elucidated the effect of silencing on virus production. Renilla luciferase mRNA, which contains a let-7 targeting site in its 3' untranslated region, was mediated when it was transported by Rev and not spliced, but it was either not mediated when it was spliced even in a partial way or it was Rev-independent. The silencing sites in the pol and env-nef regions of the HIV-1 genome, which were repressed in T cells and other cell lines, were Drosha-dependent and could also be modulated by Rev in an unspliced state. Mutant viruses that contained genomic mutations that reflect alterations to show more derepressive effects in the 3' untranslated region of the Renilla luciferase gene replicated more slowly than wild-type virus. These findings yield insights into the HIV-1 silencing sites that might allow the genome to avoid translational machinery and that might be utilized in coordinating virus production during initial virus replication. However, the function of Rev to modulate the silencing sites of unspliced RNAs would be advantageous for the efficient translation that is required to support protein production prior to viral packaging and particle production.


Assuntos
Produtos do Gene rev/metabolismo , HIV-1/genética , Splicing de RNA , RNA Viral/fisiologia , Sequência de Bases , Linhagem Celular , Inativação Gênica , Genes env , Genes pol , Repetição Terminal Longa de HIV , HIV-1/fisiologia , Humanos , MicroRNAs/genética , Regiões Promotoras Genéticas , RNA Viral/genética , Replicação Viral
13.
J Virol ; 84(22): 12048-57, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20826689

RESUMO

Hepatitis C virus (HCV) is a causative agent of chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. HCV in circulating blood associates with lipoproteins such as very low density lipoprotein (VLDL) and low-density lipoprotein (LDL). Although these associations suggest that lipoproteins are important for HCV infectivity, the roles of lipoproteins in HCV production and infectivity are not fully understood. To clarify the roles of lipoprotein in the HCV life cycle, we analyzed the effect of apolipoprotein E (ApoE), a component of lipoprotein, on virus production and infectivity. The production of infectious HCV was significantly reduced by the knockdown of ApoE. When an ApoE mutant that fails to be secreted into the culture medium was used, the amount of infectious HCV in the culture medium was dramatically reduced; the infectious HCV accumulated inside these cells, suggesting that infectious HCV must associate with ApoE prior to virus release. We performed rescue experiments in which ApoE isoforms were ectopically expressed in cells depleted of endogenous ApoE. The ectopic expression of the ApoE2 isoform, which has low affinity for the LDL receptor (LDLR), resulted in poor recovery of infectious HCV, whereas the expression of other isoforms, ApoE3 and ApoE4, rescued the production of infectious virus, raising it to an almost normal level. Furthermore, we found that the infectivity of HCV required both the LDLR and scavenger receptor class B, member I (SR-BI), ligands for ApoE. These findings indicate that ApoE is an essential apolipoprotein for HCV infectivity.


Assuntos
Apolipoproteína E2/metabolismo , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Hepacivirus/fisiologia , Hepatite C/metabolismo , Apolipoproteína E2/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Linhagem Celular Tumoral , Hepatite C/genética , Hepatite C/virologia , Humanos , Ligação Proteica
14.
Cell Mol Immunol ; 7(6): 440-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20871626

RESUMO

A dendritic cell (DC) vaccine strategy has been developed as a new cancer immunotherapy, but the goal of complete tumor eradication has not yet been achieved. We have previously shown that baculoviruses potently infect DCs and induce antitumor immunity against hepatomas in a mouse model. Baculovirus-infected, bone marrow-derived DCs (BMDCs) display increased surface expression of costimulatory molecules, such as CD80, CD86 and major histocompatibility complex (MHC) classes I and II, and secrete interferons and other proinflammatory cytokines. In this study, we evaluated the induction of antitumor immunity in mice by baculovirus-infected BMDCs against lung cancer and melanoma. After treatment with baculovirus-infected BMDCs, murine lung tumors caused by Lewis lung carcinoma (LLC) cells were significantly reduced in size, and the survival of the mice was improved. In addition, experiments using a melanoma mouse model showed that baculovirus-infected BMDCs inhibited tumor growth and improved survival compared with controls. Serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and creatinine levels remained normal in baculovirus-infected BMDC-treated mice. Our findings show that baculovirus-infected DCs induce antitumor immunity and pave the way for the use of this technique as an effective tool for DC immunotherapy against malignancies.


Assuntos
Baculoviridae/fisiologia , Carcinoma/imunologia , Carcinoma/terapia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Imunidade/imunologia , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Carcinoma/sangue , Carcinoma/patologia , Carcinoma Pulmonar de Lewis/sangue , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular Tumoral , Creatinina/sangue , Citotoxicidade Imunológica , Feminino , Imunoterapia , Melanoma/sangue , Melanoma/imunologia , Melanoma/patologia , Melanoma/terapia , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sobrevida
15.
Antivir Chem Chemother ; 20(4): 161-7, 2010 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-20231781

RESUMO

BACKGROUND: Hepatitis C virus (HCV) infection is a major cause of chronic liver disease. Here, we report a new and effective strategy for inhibiting HCV replication using an inhibitor of heat-shock protein 90, 17-AAG (17-allylamino-17demethoxygeldanamycin), and a proteasome inhibitor, MG132. METHODS: To explore the virological basis of combination therapy, we analysed the effects of 17-AAG and MG132, singly and in combination on HCV replication in an HCV replicon cell system. RESULTS: In HCV replicon cells, HCV RNA replication was suppressed by 17-AAG in a dose-dependent manner. As shown in the present study, the 50% inhibitory concentration values were 0.82 nM for 17-AAG and 0.21 nM for MG132. Low concentrations of MG132 had strong synergistic inhibitory effects with low toxicity on HCV replicon cells. CONCLUSIONS: The results of this study suggest that the different effects and synergistic actions of 17-AAG and MG132 could provide a new therapeutic approach to HCV infection.


Assuntos
Benzoquinonas/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Hepacivirus/efeitos dos fármacos , Lactamas Macrocíclicas/farmacologia , Leupeptinas/farmacologia , Inibidores de Proteassoma , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatite C/tratamento farmacológico , Humanos , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Replicon/efeitos dos fármacos , Transfecção , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
16.
Cell Immunol ; 262(1): 35-43, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20060108

RESUMO

We previously reported that the baculovirus induced a strong host immune response against infections and malignancies. Among the immune cells, the dendritic cells were most strongly infected and activated by the baculovirus, although the exact mechanism remained unclear. Here, we evaluated the non-specific immune responses of bone marrow-derived dendritic cells (BMDCs) after infection by a wild-type baculovirus. MHC class I and II molecules and co-stimulation molecules (CD40, CD80, and CD86) on BMDCs were up-regulated by baculovirus infection. At the same time, the BMDCs produced pre-inflammatory cytokines (IL-6, IL12p70, and TNF-alpha) and IFN-alpha. NK cells showed IFN-gamma production, CD69 up-regulation, and enhanced cytotoxicity when they were co-cultured with baculovirus-infected BMDCs. T cells showed IFN-gamma production, CD69 up-regulation, and cell proliferation. Ex vivo analysis performed in vitro produced similar results. These findings suggested that baculovirus-infected dendritic cells induce non-specific immune responses and can be used as an immunotherapeutic agent against viral infections and malignancies, together with present therapeutic drug regimens.


Assuntos
Baculoviridae/imunologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD/imunologia , Baculoviridae/genética , Medula Óssea/imunologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Citocinas/biossíntese , Citocinas/imunologia , DNA Viral/genética , Células Dendríticas/citologia , Feminino , Regulação Viral da Expressão Gênica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Regulação para Cima
17.
Oligonucleotides ; 19(4): 307-16, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19899951

RESUMO

Influenza viruses A and B cause widespread infections of the human respiratory tract; however, existing vaccines and drug therapy are of limited value for their treatment. Here, we show that bispecific short-hairpin small-interfering RNA constructs containing an 8-nucleotide intervening spacer, targeted against influenza virus A or influenza virus B, can inhibit the production of both types of virus in infected cell lines. This multiple vector showed remarkable ability to cope with both influenza viruses A and B. Furthermore, the Autographa californica multiple nuclear polyhedrosis virus can infect a range of mammalian cells, facilitating its use as a baculovirus vector for gene delivery into cells. In this study, baculovirus-mediated bispecific short-hairpin RNA expression markedly inhibited both influenza viruses A and B production.


Assuntos
Vírus da Influenza A/genética , Vírus da Influenza B/genética , Nucleopoliedrovírus/genética , RNA Interferente Pequeno/genética , Animais , Sequência de Bases , Linhagem Celular , Cães , Genes Virais , Engenharia Genética , Terapia Genética/métodos , Vetores Genéticos , Humanos , Vírus da Influenza A/fisiologia , Vírus da Influenza B/fisiologia , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Proteínas do Nucleocapsídeo , Plasmídeos/genética , RNA Interferente Pequeno/química , RNA Viral/genética , Proteínas de Ligação a RNA/genética , Proteínas do Core Viral/genética , Replicação Viral/genética
18.
Virol J ; 6: 156, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19796392

RESUMO

BACKGROUND: Hepatitis C virus (HCV) is one of the main causes of liver-related morbidity and mortality. Although combined interferon-alpha-ribavirin therapy is effective for about 50% of the patients with HCV, better therapies are needed and preventative vaccines have yet to be developed. Short-hairpin RNAs (shRNAs) inhibit gene expression by RNA interference. The application of transient shRNA expression is limited, however, due to the inability of the shRNA to replicate in mammalian cells and its inefficient transduction. The duration of transgene (shRNA) expression in mammalian cells can be significantly extended using baculovirus-based shRNA-expressing vectors that contain the latent viral protein Epstein-Barr nuclear antigen 1 (EBNA1) and the origin of latent viral DNA replication (OriP) sequences. These recombinant vectors contain compatible promoters and are highly effective for infecting primary hepatocyte and hepatoma cell lines, making them very useful tools for studies of hepatitis B and hepatitis C viruses. Here, we report the use of these baculovirus-based vector-derived shRNAs to inhibit core-protein expression in full-length hepatitis C virus (HCV) replicon cells. RESULTS: We constructed a long-term transgene shRNA expression vector that contains the EBV EBNA1 and OriP sequences. We also designed baculovirus vector-mediated shRNAs against the highly conserved core-protein region of HCV. HCV core protein expression was inhibited by the EBNA1/OriP baculovirus vector for at least 14 days, which was considerably longer than the 3 days of inhibition produced by the wild-type baculovirus vector. CONCLUSION: These findings indicate that we successfully constructed a long-term transgene (shRNA) expression vector (Ac-EP-shRNA452) using the EBNA1/OriP system, which was propagated in Escherichia coli and converted into mammalian cells. The potential anti-HCV activity of the long-term transgene (shRNA) expression vector was evaluated with the view of establishing highly effective therapeutic agents that can be further developed for HCV gene therapy applications.


Assuntos
Baculoviridae/fisiologia , Antígenos Nucleares do Vírus Epstein-Barr/genética , Terapia Genética/métodos , Vetores Genéticos , Hepacivirus/genética , Origem de Replicação/genética , Proteínas do Core Viral/antagonistas & inibidores , Replicação Viral , Baculoviridae/genética , Linhagem Celular , Humanos , RNA Interferente Pequeno/genética , Proteínas do Core Viral/genética
19.
Nucleic Acids Symp Ser (Oxf) ; (53): 87-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19749273

RESUMO

Human apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3G (APOBEC3G) is known to play a role in intrinsic cellular immunity against human immunodeficiency virus type 1 (HIV-1). The antiretroviral activity of APOBEC3G is associated with hypermutation of viral DNA through cytidine deamination. APOBEC3G contains two cytidine deaminase domains that are characterized by a highly conserved zinc-coordinating motif. It is known that only the C-terminal domain of APOBEC3G (c-APOBEC3G) is involved in the catalytic activity. Here, we present the solution structure and the interaction with single-stranded DNA of c-APOBEC3G. Furthermore, we have succeeded for the first time in monitoring the deamination reaction of c-APOBEC3G in real-time using NMR signals. The monitoring has demonstrated that the deamination reaction occurs in a strict 3'-->5'


Assuntos
Citidina Desaminase/química , DNA de Cadeia Simples/química , Desaminase APOBEC-3G , HIV-1/genética , Humanos , Cinética , Ressonância Magnética Nuclear Biomolecular
20.
Nucleic Acids Symp Ser (Oxf) ; (53): 287-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19749373

RESUMO

Influenza viruses A and B cause widespread infections of the human respiratory tract; however, existing vaccines and drug therapy are of limited value for their treatment. Here we show that bispecific short hairpin small-interfering RNA constructs containing an eight-nucleotide intervening spacer, targeted against influenza virus A or influenza virus B, can inhibit the production of both types of virus in infected cell lines. This multiple vector showed remarkable ability to cope with both influenza virus A or B. Furthermore, the Autographa californica multiple nuclear polyhedrosis virus can infect a range of mammalian cells, facilitating its use as a baculovirus vector for gene delivery into cells. In this study, baculovirus-mediated bispecific short-hairpin RNA expression markedly inhibited both influenza virus A and B production.


Assuntos
Baculoviridae/genética , Vírus da Influenza A/genética , Vírus da Influenza B/genética , RNA Interferente Pequeno , Animais , Linhagem Celular , Cães , Regulação para Baixo , Vetores Genéticos , Proteínas do Nucleocapsídeo , RNA Interferente Pequeno/química , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas do Core Viral/antagonistas & inibidores , Proteínas do Core Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA