Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 591: 130-136, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33454058

RESUMEN

The coronavirus disease (COVID-19) pandemic, resulting from human-to-human transmission of a novel severe acute respiratory syndrome coronavirus (SARS-CoV-2), has led to a global health crisis. Given that the 3 chymotrypsin-like protease (3CLpro) of SARS-CoV-2 plays an indispensable role in viral polyprotein processing, its successful inhibition halts viral replication and thus constrains virus spread. Therefore, developing an effective SARS-CoV-2 3CLpro inhibitor to treat COVID-19 is imperative. A fluorescence resonance energy transfer (FRET)-based method was used to assess the proteolytic activity of SARS-CoV-2 3CLpro using intramolecularly quenched fluorogenic peptide substrates corresponding to the cleavage sequence of SARS-CoV-2 3CLpro. Molecular modeling with GEMDOCK was used to simulate the molecular interactions between drugs and the binding pocket of SARS-CoV-2 3CLpro. This study revealed that the Vmax of SARS-CoV-2 3CLpro was about 2-fold higher than that of SARS-CoV 3CLpro. Interestingly, the proteolytic activity of SARS-CoV-2 3CLpro is slightly more efficient than that of SARS-CoV 3CLpro. Meanwhile, natural compounds PGG and EGCG showed remarkable inhibitory activity against SARS-CoV-2 3CLpro than against SARS-CoV 3CLpro. In molecular docking, PGG and EGCG strongly interacted with the substrate binding pocket of SARS-CoV-2 3CLpro, forming hydrogen bonds with multiple residues, including the catalytic residues C145 and H41. The activities of PGG and EGCG against SARS-CoV-2 3CLpro demonstrate their inhibition of viral protease activity and highlight their therapeutic potentials for treating SARS-CoV-2 infection.


Asunto(s)
Catequina/análogos & derivados , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Taninos Hidrolizables/farmacología , Simulación del Acoplamiento Molecular , SARS-CoV-2/efectos de los fármacos , Sitios de Unión , COVID-19/epidemiología , COVID-19/prevención & control , COVID-19/virología , Catequina/química , Catequina/metabolismo , Catequina/farmacología , Proteasas 3C de Coronavirus/química , Proteasas 3C de Coronavirus/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Humanos , Taninos Hidrolizables/química , Taninos Hidrolizables/metabolismo , Cinética , Modelos Moleculares , Estructura Molecular , Pandemias , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Unión Proteica , Dominios Proteicos , SARS-CoV-2/enzimología , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos
2.
J Gastroenterol Hepatol ; 29(5): 1073-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24325631

RESUMEN

BACKGROUND AND AIM: Vascular endothelial (VEGF) and fibroblast growth factor (FGF)-induced hepatic stellate (HSCs) and liver endothelial cells (LECs) activation accelerates hepatic fibrogenesis and angiogenesis, and hemodynamic dysarrangements in cirrhosis. VEGF targeting agents had been reported as potential drugs for cirrhosis. However, the evaluation of effects of dual VEGF/FGF targeting agent in cirrhosis is still limited. METHODS: Using hemodynamic parameters, blood chemistry, primary isolated HSCs and LECs, histology, and digital imaging, we assess the effects of 2-week brivanib alaninate, a dual VEGFR/FGFR inhibitor, treatment in the pathophysiology of bile duct-ligated-cirrhotic rats. RESULTS: Fibrogenic and angiogenic markers in the serum and liver of bile duct-ligated-cirrhotic rats, including hydroxyproline, transforming growth factor-ß1, angiopoietin-1, VEGF, FGF-2, endocan and phosphorylated-VEGFR2/VEGFR2, and phosphorylated-FGFR/FGFR together with hepatic CD31/angiopoietin-1 expressions (immunohistochemistry staining), angiogenesis (micro-computed tomography scan), microcirculatory dysfunction (in vivo miscroscopy and in situ liver perfusion study), portal hypertension, and hyperdynamic circulations (colored microsphere methods) were markedly suppressed and ameliorated by brivanib alaninate treatment. In in vitro study, acute brivanib alaninate incubation inhibited the transforming growth factor-ß1-induced HSCs contraction/migration and VEGF-induced LECs angiogenesis. Concomitantly, the overexpression of various fibrogenic and angiogenic markers in HSCs and LECs, and in their culture media, was increased in parallel and these changes were suppressed by acute brivanib alaninate incubation. CONCLUSIONS: This study demonstrated that brivanib alaninate targeting multiple mechanisms and working in the different pathogenic steps of the complications of cirrhotic rats with portal hypertension.


Asunto(s)
Alanina/análogos & derivados , Hipertensión Portal/tratamiento farmacológico , Hipertensión Portal/genética , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/genética , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Triazinas/uso terapéutico , Alanina/farmacología , Alanina/uso terapéutico , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/efectos de los fármacos , Cirrosis Hepática/patología , Masculino , Terapia Molecular Dirigida , Ratas , Ratas Sprague-Dawley , Triazinas/farmacología
3.
Mol Ther ; 20(2): 298-305, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22068428

RESUMEN

Sindbis virus (SBV) has been shown to possess oncolytic potential in many human xenograft tumor models in immunocompromised mice. However, the mechanism underlying the tumor selectivity of SBV remains undetermined. In this study, we provide evidence that the tumor tropism of SBV infection is not determined by the levels of SBV receptor but by the status of the type I interferon (IFN) response in the tumors. Our results demonstrate that cells with defects in the IFN response (in either IFN-ß production or IFN signaling) were highly susceptible to SBV infection in vitro. The results of oncolysis experiments conducted in immunocompetent animals further confirmed that the success of SBV-mediated oncolysis is greatly dependent on the presence of defects in IFN signaling in tumors. In all cases, viral titers rapidly declined in tumors due to host immune responses in immunocompetent animals. Interestingly, however, tumor-specific immune responses were concomitantly elicited, which might contribute to the sustained antitumor effect observed after the clearance of SBV. These findings indicate that SBV-mediated virotherapy is a promising therapeutic strategy for cancers defective in the IFN response and underscore the importance of bystander antitumor immunity in the efficacy of this virotherapy.


Asunto(s)
Efecto Espectador/inmunología , Vectores Genéticos/genética , Interferones/genética , Neoplasias/inmunología , Neoplasias/terapia , Virus Oncolíticos/genética , Virus Sindbis/genética , Animales , Línea Celular , Terapia Genética , Humanos , Interferón beta/genética , Interferón beta/inmunología , Interferón beta/metabolismo , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Receptores Virales/metabolismo , Transducción de Señal , Virus Sindbis/fisiología , Tropismo Viral
4.
Proc Natl Acad Sci U S A ; 107(33): 14769-74, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20679198

RESUMEN

Cytokine and antiangiogenic gene therapies have proved effective in implanted hepatocellular carcinoma (HCC) models in which small tumor burdens were established in small rodents. These models, however, may not reflect human HCCs, which are frequently detected at a stage when tumors are large and multifocal. In addition, HCC in patients is often associated with viral hepatitis. To investigate the effectiveness of a mixture type of gene therapy strategy on large tumor burdens, we used the woodchuck model in which woodchuck hepatitis virus-induced HCCs are large and multifocal, simulating the conditions in humans. Adenoviruses encoding antiangiogenic factors (pigment epithelium-derived factor and endostatin) or cytokines (GM-CSF and IL-12) were delivered via the hepatic artery separately or in combination into woodchuck livers bearing HCCs. Our results showed that the mixture type of strategy, which contained two cytokines and two antiangiogenic factors, had better antitumor effects on large tumors as compared with monotherapy either with antiangiogenic or cytokine genes. The immunotherapy recruited significant levels of CD3(+) T cells that infiltrated the tumors, whereas the antiangiogenesis-based therapy significantly reduced tumor vasculature. The mixture type of gene therapy achieved both effects. In addition, it induced high levels of natural killer cells and apoptotic cells and reduced the levels of immunosuppressive effectors in the tumor regions. Hence, antiangiogenic therapy may provide the advantage of reducing immune tolerance in large tumors, making them more vulnerable to the immune reactions. Our study implies that in the future, the combination therapy may prove effective for the treatment of patients with advanced HCC.


Asunto(s)
Terapia Genética/métodos , Inmunoterapia/métodos , Neoplasias Hepáticas Experimentales/terapia , Enfermedades de los Roedores/terapia , Alanina Transaminasa/sangre , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Animales , Aspartato Aminotransferasas/sangre , Terapia Combinada , Endostatinas/genética , Endostatinas/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Hepatitis B/complicaciones , Hepatitis B/virología , Virus de la Hepatitis B de la Marmota/crecimiento & desarrollo , Humanos , Neoplasias Hepáticas Experimentales/sangre , Neoplasias Hepáticas Experimentales/etiología , Marmota , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Enfermedades de los Roedores/sangre , Enfermedades de los Roedores/etiología , Serpinas/genética , Serpinas/metabolismo , Resultado del Tratamiento , Carga Tumoral , gamma-Glutamiltransferasa/sangre
5.
Int J Cancer ; 130(12): 2892-902, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21805477

RESUMEN

Tumor-induced angiogenesis has been shown to suppress immune responses. One mechanism is to suppress leukocyte-endothelial cell interaction by down-regulating the expression of adhesion molecules, such as intercellular adhesion molecule (ICAM)-1, vascular cell adhesion molecule (VCAM)-1 and E-selectin on the tumor endothelium, which enables tumor cells to escape immune surveillance. Calreticulin (CRT), a chaperone protein mainly located in the endoplasmic reticulum, has been shown to exert anti-angiogenic activity and inhibit tumor growth. Here, we demonstrate that in addition to inhibiting angiogenesis, CRT also enhances the expression of both ICAM-1 and VCAM-1 on tumor endothelial cells. This expression results in enhanced leukocyte-endothelial cell interactions and increased lymphocyte infiltration into tumors. Therefore, combining intramuscular CRT gene transfer with intratumoral cytokine gene therapies significantly improves the antitumor effects of immunotherapy by markedly increasing the levels of tumor-infiltrating lymphocytes. This combined treatment increased the levels of infiltrating lymphocytes to those achieved using four times the cytokine dosage. The combined therapy also resulted in lower levels of immunosuppressive molecules and higher levels of activated T-cells in the tumor microenvironment than immunotherapy alone. In conclusion, this study describes a new antitumor mechanism of CRT that involves the up-regulation of tumor endothelial adhesion molecules and the enhanced infiltration of tumor-specific lymphocytes. Thus, CRT treatment can make tumor cells more vulnerable to immunotherapy and improve the therapeutic efficacy of immunotherapy.


Asunto(s)
Calreticulina/metabolismo , Selectina E/metabolismo , Inmunoterapia , Molécula 1 de Adhesión Intercelular/metabolismo , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/terapia , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Calreticulina/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Cricetinae , Selectina E/biosíntesis , Selectina E/genética , Células Endoteliales/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Técnicas de Transferencia de Gen , Molécula 1 de Adhesión Intercelular/genética , Leucocitos/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Neovascularización Patológica , Ratas , Ratas Wistar , Linfocitos T/inmunología , Linfocitos T/metabolismo , Escape del Tumor , Molécula 1 de Adhesión Celular Vascular/genética
6.
J Biol Chem ; 285(36): 28183-90, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20601427

RESUMEN

The NS5A protein of the hepatitis C virus (HCV) is an integral component of the viral replicase. It also modulates cellular signaling and perturbs host interferon responses. The multifunctional characteristics of NS5A are mostly attributed to its ability to interact with various cellular proteins. This study aimed to identify the novel cellular factors that interact with NS5A and decipher the significance of this interaction in viral replication. The NS5A-interacting proteins were purified by the tandem affinity purification (TAP) procedure from cells expressing NS5A and identified by mass spectrometry. The chaperone protein Hsp72 was identified herein. In vivo protein-protein interaction was verified by co-immunoprecipitation and an in situ proximity ligation assay. In addition to NS5A, Hsp72 was also associated with other members of the replicase complex, NS3 and NS5B, suggesting that it might be directly involved in the HCV replication complex. Hsp72 plays a positive regulatory role in HCV RNA replication by increasing levels of the replicase complex, which was attributed either to the increased stability of the viral proteins in the replicase complex or to the enhanced translational activity of the internal ribosome entry site of HCV. The fact that the host chaperone protein Hsp72 is involved in HCV RNA replication may represent a therapeutic target for controlling virus production.


Asunto(s)
Proteínas del Choque Térmico HSP72/metabolismo , Hepacivirus/enzimología , ARN Viral/biosíntesis , ARN Polimerasa Dependiente del ARN/metabolismo , Línea Celular , Proteínas del Choque Térmico HSC70/metabolismo , Hepacivirus/metabolismo , Hepacivirus/fisiología , Humanos , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
7.
Front Microbiol ; 12: 664955, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025620

RESUMEN

Enterovirus A71 (EV-A71) is a human pathogen causing hand, foot, and mouth disease (HFMD) in children. Its infection can lead to severe neurological diseases or even death in some cases. While being produced in a large quantity during infection, viral proteins often require the assistance from cellular chaperones for proper folding. In this study, we found that heat shock protein A6 (HSPA6), whose function in viral life cycle is scarcely studied, was induced and functioned as a positive regulator for EV-A71 infection. Depletion of HSPA6 led to the reductions of EV-A71 viral proteins, viral RNA and virions as a result of the downregulation of internal ribosomal entry site (IRES)-mediated translation. Unlike other HSP70 isoforms such as HSPA1, HSPA8, and HSPA9, which regulate all phases of the EV-A71 life, HSPA6 was required for the IRES-mediated translation only. Unexpectedly, the importance of HSPA6 in the IRES activity could be observed in the absence of viral proteins, suggesting that HSPA6 facilitated IRES activity through cellular factor(s) instead of viral proteins. Intriguingly, the knockdown of HSPA6 also caused the reduction of luciferase activity driven by the IRES from coxsackievirus A16, echovirus 9, encephalomyocarditis virus, or hepatitis C virus, supporting that HSPA6 may assist the function of a cellular protein generally required for viral IRES activities.

8.
Front Microbiol ; 11: 1656, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32760390

RESUMEN

Enterovirus A71 (EV-A71) is one of the major etiologic agents causing hand, foot, and mouth disease (HFMD) in children and occasionally causes severe neurological diseases or even death. EV-A71 replicates rapidly in host cells. For a successful infection, viruses produce large quantities of viral proteins in a short period, which requires cellular chaperone proteins for viral protein folding and viral particle assembly. In this study, we explored the roles of the heat shock protein 70 (HSP70) chaperone subnetwork in the EV-A71 life cycle. Our results revealed that EV-A71 exploits multiple HSP70s at each step of the viral life cycle, i.e., viral entry, translation, replication, assembly and release, and that each HSP70 typically functions in several stages of the life cycle. For example, the HSP70 isoforms HSPA1, HSPA8, and HSPA9 are required for viral entry and the translational steps of the infection. HSPA8 and HSPA9 may facilitate folding and stabilize viral proteins 3D and 2C, respectively, thus contributing to the formation of a replication complex. HSPA8 and HSPA9 also promote viral particle assembly, whereas HSPA1 and HSPA8 are involved in viral particle release. Because of the importance of various HSP70s at distinct steps of the viral life cycle, an allosteric inhibitor, JG40, which targets all HSP70s, significantly blocks EV-A71 infection. JG40 also blocks the replication of several other enteroviruses, such as coxsackievirus (CV) A16, CVB1, CVB3, and echovirus 11. Thus, targeting HSP70s may be a means of providing broad-spectrum antiviral therapy.

9.
J Gene Med ; 11(10): 933-40, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19639605

RESUMEN

BACKGROUND: Ultrasound (US) is a novel and effective tool for the local delivery of genes into target tissues. US can temporarily change the permeability of a cell membrane and thus enhance the delivery of naked DNA into cells. In the present study, the efficiencies of gene expression mediated by US delivery in orthotopic liver tumor, subcutaneous tumor and muscle tissue were evaluated by changing the contrast agent concentrations and US exposure durations. METHODS: Plasmid DNA coding for luciferase, interleukin-12 or enhanced green fluorescence protein was mixed with SonoVue and injected intratumorally or intramuscularly. The injection sites were then exposed to US (20% duty cycle and 0.4 W/cm(2) intensity). RESULTS: The results obtained showed that the optimal condition was 50% SonoVue for tumors and 30% for muscle, with 10 min of US exposure. The expression levels of the transfected DNAs were in the order: muscle > subcutaneous tumor > orthotopic liver tumor. CONCLUSIONS: The present study indicates that muscle tissue is a good target site for producing large amounts of gene products for the purpose of gene therapy.


Asunto(s)
Expresión Génica , Neoplasias Hepáticas Experimentales/diagnóstico por imagen , Neoplasias Hepáticas Experimentales/metabolismo , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/metabolismo , Transfección/métodos , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/biosíntesis , Inhibidores de la Angiogénesis/genética , Animales , Línea Celular , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/metabolismo , Terapia Genética/métodos , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Interleucina-12/administración & dosificación , Interleucina-12/biosíntesis , Interleucina-12/genética , Neoplasias Hepáticas Experimentales/terapia , Luciferasas/administración & dosificación , Luciferasas/biosíntesis , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Esquelético/efectos de los fármacos , Especificidad de Órganos , Plásmidos/administración & dosificación , Plásmidos/genética , Plásmidos/metabolismo , Ultrasonografía
10.
Mol Ther ; 16(10): 1681-1687, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28189003

RESUMEN

Human hepatocellular carcinoma (HCC) often arises from a background of liver cirrhosis. Therefore, in order to develop therapeutic strategies for HCC, an animal model bearing multifocal liver tumors accompanied by liver cirrhosis is a preferred experimental setting. In this study, we developed a rapid and reproducible method for generating such a model in rats by weekly administration of diethylnitrosamine (DEN) at doses based on body weight (BW). By adjusting the duration of administration of DEN, the animals could be induced to develop HCC alone, or HCC and liver cirrhosis simultaneously. The latter model was used for evaluating the therapeutic effects of adenoviral delivery of interferon-α (IFN-α). Our results demonstrated that targeting of IFN-α expression to the liver significantly reduced liver tumor volume and ameliorated liver cirrhosis. Mechanistic studies revealed that IFN-α gene therapy induced immunomodulatory, antiproliferative, and proapoptotic activities that were effective in the control of tumor growth, and reduced the expressions of transforming growth factor-ß (TGF-ß) and tissue inhibitor of metalloproteinase-1 (TIMP-1), leading to amelioration of liver cirrhosis. These results suggest that IFN-α gene therapy is a promising strategy to treat HCC patients who have concomitant liver cirrhosis.

11.
Mol Ther ; 16(10): 1681-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18665156

RESUMEN

Human hepatocellular carcinoma (HCC) often arises from a background of liver cirrhosis. Therefore, in order to develop therapeutic strategies for HCC, an animal model bearing multifocal liver tumors accompanied by liver cirrhosis is a preferred experimental setting. In this study, we developed a rapid and reproducible method for generating such a model in rats by weekly administration of diethylnitrosamine (DEN) at doses based on body weight (BW). By adjusting the duration of administration of DEN, the animals could be induced to develop HCC alone, or HCC and liver cirrhosis simultaneously. The latter model was used for evaluating the therapeutic effects of adenoviral delivery of interferon-alpha (IFN-alpha). Our results demonstrated that targeting of IFN-alpha expression to the liver significantly reduced liver tumor volume and ameliorated liver cirrhosis. Mechanistic studies revealed that IFN-alpha gene therapy induced immunomodulatory, antiproliferative, and proapoptotic activities that were effective in the control of tumor growth, and reduced the expressions of transforming growth factor-beta (TGF-beta) and tissue inhibitor of metalloproteinase-1 (TIMP-1), leading to amelioration of liver cirrhosis. These results suggest that IFN-alpha gene therapy is a promising strategy to treat HCC patients who have concomitant liver cirrhosis.


Asunto(s)
Terapia Genética , Interferón-alfa/genética , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas Experimentales/terapia , Animales , Apoptosis , Carcinógenos/toxicidad , Proliferación Celular , Dietilnitrosamina/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/complicaciones , Neoplasias Hepáticas Experimentales/patología , Ratas , Inhibidor Tisular de Metaloproteinasa-1/genética , Factor de Crecimiento Transformador beta/genética
12.
Front Microbiol ; 9: 1324, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29971060

RESUMEN

The translation of enterovirus 71 (EV71) is mediated by an internal ribosome entry site (IRES)-dependent manner. EV71 IRES comprises five highly structured domains (domains II-VI) in the 5'-untranslated region of the viral mRNA. A conserved AUG triplet residing in domain VI is proposed to be the ribosome entry site. It is thus envisaged that the highly structured conformation of domain VI may actually reduce the accessibility of the AUG triplet to the ribosome. This study identified a DEAD-box family RNA helicase, DDX3X, that positively regulated the EV71 IRES-dependent translation. The helicase activity of DDX3X was required for the stimulation of EV71 IRES activity; however, DDX3X was no longer important for the IRES activity when the secondary structure of domain VI was destabilized. DDX3X interacted with the truncated eIF4G which bound specifically to domain V. Thus, we proposed that DDX3X might bind to domain VI or a region nearby via the interaction with the truncated eIF4G, and subsequently unwound the secondary structure of domain VI to facilitate ribosome entry. Additionally, we demonstrated that the viral 2Apro and 3Cpro enhanced the IRES-dependent translation via their protease activities. Together, these results indicate that DDX3X is an important RNA helicase involved in EV71 IRES-dependent translation and that IRES translation is enhanced by viral infection, partly mediated by viral protease activity.

13.
Front Microbiol ; 9: 3253, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30687256

RESUMEN

Hepatitis B virus (HBV) infection causes acute and chronic liver inflammation. Recent studies have demonstrated that some viral antigens can suppress host innate and adaptive immunity, and thus lead to HBV liver persistency. However, the cellular factors that can help host cells to clear HBV during acute infection remain largely unknown. Here, we used HBV-cleared and HBV-persistent mouse models to seek for cellular factors that might participate in HBV clearance. HBV replicon DNA was delivered into the mouse liver by hydrodynamic injection. RNA-Seq analysis was conducted to identify immune-related genes that were differentially expressed in HBV-persistent and HBV-cleared mouse models. A cellular factor, B cell lymphoma 6 (BCL6), was found to be significantly upregulated in the liver of HBV-cleared mice upon HBV clearance. Co-expression of BCL6 and a persistent HBV clone rendered the clone largely cleared, implicating an important role of BCL6 in controlling HBV clearance. Mechanistic studies demonstrated that BCL6 functioned as a repressor, binding to and suppressing the activities of the four HBV promoters. Correspondingly, BCL6 expression significantly reduced the levels of HBV viral RNA, DNA, and proteins. BCL6 expression could be stimulated by inflammatory cytokines such as TNF-α; the BCL6 in turn synergized TNF-α signaling to produce large amounts of CXCL9 and CXCL10, leading to increased infiltrating immune cells and elevated cytokine levels in the liver. Thus, positive feedback loops on BCL6 expression and immune responses could be produced. Together, our results demonstrate that BCL6 is a novel host restriction factor that exerts both anti-HBV and immunomodulatory activities. Induction of BCL6 in the liver may ultimately assist host immune responses to clear HBV.

14.
Front Immunol ; 8: 972, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28848563

RESUMEN

Using shRNA high-throughput screening, we identified H-Ras as a regulator of antiviral activity, whose depletion could enhance Sindbis virus replication. Further analyses indicated that depletion of H-Ras results in a robust increase in vesicular stomatitis virus infection and a decrease in Sendai virus (SeV)-induced retinoic acid-inducible gene-I-like receptor (RLR) signaling. Interestingly, however, ectopic expression of wild-type H-Ras results in a biphasic mode of RLR signaling regulation: while low-level expression of H-Ras enhances SeV-induced RLR signaling, high-level expression of H-Ras significantly inhibits this signaling. The inhibitory effects correlate with the activation status of H-Ras. As a result, oncogenic H-Ras, H-RasV12, strongly inhibits SeV-induced IFN-ß promoter activity and type I interferon signaling. Conversely, the positive effects exerted by H-Ras on RLR signaling are independent of its signaling activity, as a constitutively inactive form of H-Ras, H-RasN17, also positively regulates RLR signaling. Mechanistically, we demonstrate that depletion of H-Ras reduces the formation of MAVS-TNF receptor-associated factor 3 signaling complexes. These results reveal that the H-Ras protein plays a role in promoting MAVS signalosome assembly in the mitochondria, whereas oncogenic H-Ras exerts a negative effect on type I IFN responses.

15.
Clin Cancer Res ; 11(4): 1639-49, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15746069

RESUMEN

PURPOSE: To evaluate antitumor effects on intracerebral gliomas of genetically engineered tumor vaccines expressing granulocyte-macrophage colony-timulating factor (GM-CSF), B7.1, or both (combination). EXPERIMENTAL DESIGN: A rat glioma cell line, RT-2, was engineered with a retroviral vector to express GM-CSF, B7.1, or combination. Tumorigenicity of engineered cells and therapeutic effects of s.c. given irradiated or live tumor vaccines on parental intracerebral gliomas were studied. Immune cell infiltration induced at vaccine and tumor sites was examined by histologic and immunohistochemical staining. Apoptosis of T cells from vaccine sites was analyzed with fluorescence-activated cell sorting. RESULTS: Engineered RT-2 cells exhibited reduced s.c. tumorigenicity in rats with reduced tumor growth and prolonged animal survival time compared with control rats. Rats with intracerebral gliomas s.c. treated with irradiated or live GM-CSF-expressing vaccines had 60% and 100% survival rates, respectively, significantly better than the control groups (P < 0.05). In contrast, rats treated with vaccines expressing B7.1 or the combination had no or mild therapeutic effects. Studies revealed less T-cell infiltration at both vaccine and tumor sites in rats treated with vaccines expressing B7.1 or the combination than in rats treated with a vaccine expressing GM-CSF. Cell sorting analyses revealed higher proportions of apoptotic T cells at vaccine sites of rats treated with the combination than those treated with vaccine expressing GM-CSF. CONCLUSIONS: Combination of GM-CSF- and B7.1-expressing tumor vaccines exerted no synergistic, or even worse, therapeutic effects on gliomas compared with single GM-CSF-secreting tumor vaccine. The worse therapeutic effects of the GM-B7.1-expressing tumor vaccine than the GM-CSF-expressing tumor vaccine were related to the reduced T-cell amount and increased T-cell apoptosis in the former.


Asunto(s)
Apoptosis/inmunología , Antígeno B7-1/inmunología , Glioma/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Linfocitos T/inmunología , Animales , Antígeno B7-1/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Citometría de Flujo , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Glioma/genética , Glioma/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Ratas , Retroviridae/genética , Análisis de Supervivencia , Linfocitos T/metabolismo , Factores de Tiempo , Transfección
16.
PLoS One ; 7(5): e36984, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615863

RESUMEN

The mechanism underlying the chronicity of hepatitis B virus (HBV) infection has long been an interesting question. However, this mechanism remains unclear largely due to the lack of an animal model that can support persistent HBV replication and allow for the investigation of the relevant immune responses. In this study, we used hydrodynamic injection to introduce HBV replicon DNA into the livers of three different mouse strains: BALB/c, C57BL/6, and FVB/N. Interestingly, we found that an HBV clone persistently replicated in the livers of FVB/N mice for up to 50 weeks but was rapidly cleared from the livers of BALB/c and C57BL/6 mice. Flow cytometric analysis and quantitative reverse transcription PCR analysis of the mouse livers indicated that after DNA injection, FVB/N mice had few intrahepatic activated cytotoxic T lymphocytes (CTLs) and produced low levels of alanine aminotransferase, interferon (IFN)-γ, tumor necrosis factor (TNF)-α, and the CXCL9 and CXCL10 chemokines. These findings were in sharp contrast with those observed in BALB/c and C57BL/6 mice, reflecting a strong correlation between the degree of liver inflammation and viral clearance. Mutational analysis further demonstrated that a change of Asn-214 to Ser-214 in the HBV surface antigen rendered the persistent HBV clone clearable in FVB/N mice, which was accompanied by increased levels of activated CTL and upregulated expression of IFN-γ, CXCL9, and CXCL10 in the livers. These results indicate that the heterogeneity of the host factors and viral sequences may influence the immune responses against HBV. An inadequate activation of immune or inflammatory responses can lead to persistent HBV replication in vivo.


Asunto(s)
Virus de la Hepatitis B/fisiología , Hepatitis B/inmunología , Hepatitis B/virología , Replicación Viral/genética , Alanina Transaminasa/genética , Alanina Transaminasa/inmunología , Alanina Transaminasa/metabolismo , Animales , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Quimiocina CXCL9/metabolismo , Replicación del ADN/genética , Replicación del ADN/inmunología , Hepatitis B/genética , Hepatitis B/metabolismo , Antígenos de Superficie de la Hepatitis B/genética , Antígenos de Superficie de la Hepatitis B/inmunología , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Interacciones Huésped-Patógeno , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/virología , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Hígado/inmunología , Hígado/metabolismo , Hígado/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Replicación Viral/inmunología
17.
Cancer Lett ; 320(1): 23-30, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22266191

RESUMEN

We have recently demonstrated that a 4-in-1 gene therapy strategy that contains two anti-angiogenic genes [endostatin and pigment epithelium-derived factor] and two cytokine genes [granulocyte macrophage colony-stimulating factor and interleukin 12] has a considerable antitumor effect on large tumors in a woodchuck hepatoma model. The current study further investigates the underlying mechanisms for the antitumor effect observed by using small rodent models. We found that immunotherapy alone increased immunosuppressive cells in large tumors over time, whereas the anti-angiogenic therapy contained in the 4-in-1 strategy alleviated immunosuppression and made tumors vulnerable to immunotherapy, thus resulting in a synergistic antitumor effect.


Asunto(s)
Endostatinas/genética , Endostatinas/inmunología , Proteínas del Ojo/genética , Proteínas del Ojo/inmunología , Terapia Genética/métodos , Inmunoterapia/métodos , Neoplasias Hepáticas Experimentales/terapia , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/inmunología , Serpinas/genética , Serpinas/inmunología , Adenoviridae/genética , Animales , Apoptosis/genética , Apoptosis/inmunología , Línea Celular Tumoral , Terapia Combinada , Endostatinas/biosíntesis , Proteínas del Ojo/biosíntesis , Humanos , Neoplasias Hepáticas Experimentales/irrigación sanguínea , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/terapia , Factores de Crecimiento Nervioso/biosíntesis , Serpinas/biosíntesis , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología
18.
J Med Chem ; 52(9): 2716-23, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19419203

RESUMEN

Hepatitis C virus nonstructural protein 3 (HCV NS3) helicase is believed to be essential for viral replication and has become an attractive target for the development of antiviral drugs. A fluorescence resonant energy transfer helicase assay was established for fast screening of putative inhibitors selected from virtual screening using the program DOCK. Soluble blue HT (1) was first identified as a novel HCV helicase inhibitor. Crystal structure of the NS3 helicase in complex with soluble blue HT shows that the inhibitor bears a significantly higher binding affinity mainly through a 4-sulfonatophenylaminophenyl group, and this is consistent with the activity assay. Subsequently, fragment-based searches were utilized to identify triphenylmethane derivatives for more potent inhibitors. Lead optimization resulted in a 3-bromo-4-hydroxyl substituted derivative 12 with an EC(50) value of 2.72 microM to Ava.5/Huh-7 cells and a lower cytotoxicity to parental Huh-7 cells (CC(50) = 10.5 microM), and it indeed suppressed HCV replication in the HCV replicon cells. Therefore, these inhibitors with structural novelty may serve as a useful scaffold for the discovery of new HCV NS3 helicase inhibitors.


Asunto(s)
Descubrimiento de Drogas , Hepacivirus/enzimología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Compuestos de Tritilo/química , Compuestos de Tritilo/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Sitios de Unión , Línea Celular , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Hepacivirus/efectos de los fármacos , Modelos Moleculares , Replicón/efectos de los fármacos , Programas Informáticos , Proteínas no Estructurales Virales/química
19.
J Hepatol ; 49(6): 899-907, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18842320

RESUMEN

BACKGROUND/AIMS: Patients infected with hepatitis C virus (HCV) genotype 2 or 3 usually respond better to interferon (IFN) treatment than those infected with genotype 1. In this study, we investigated whether the non-structural 5A protein (NS5A) of HCV genotypes 1 and 2 (1b-NS5A and 2a-NS5A, respectively) exerted differential counteractivities against IFN treatment. METHODS: We compared the inhibitory effects of 1b-NS5As and 2a-NS5As on IFN activity. We also investigated the replication inhibition of HCV subgenomic replicons containing 1b-NS5A or 2a-NS5A in response to IFN treatment. RESULTS: 1b-NS5As exerted more profound inhibitory effects on IFN activity than 2a-NS5As. The replication of the 2a-NS5A-containing replicons was more sensitive to IFN treatment than that of the 1b-NS5A-containing replicons. Deletion of the interferon sensitivity-determining region/protein kinase R-binding domain (PKR-BD), the V3 domain, or the C-terminus region of NS5A significantly abrogated its anti-IFN activity. Domain swapping between 1b-NS5A and 2a-NS5A in the V3 domain and/or the C-terminus region resulted in a transfer of their anti-IFN activity. CONCLUSIONS: 1b-NS5As exert higher magnitudes of IFN antagonism than do 2a-NS5As. The V3 and the C-terminus regions are responsible for the differential anti-IFN effects. This phenomenon may partly explain the genotype-linked differences in the response of HCV to IFN treatment.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Hepatitis C/virología , Interferón-alfa/farmacología , Proteínas no Estructurales Virales/genética , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Farmacorresistencia Viral/genética , Genotipo , Células HeLa , Hepacivirus/genética , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/fisiología , Estructura Terciaria de Proteína/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transfección , Células Vero , Proteínas no Estructurales Virales/química
20.
Hepatology ; 45(3): 746-54, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17326190

RESUMEN

UNLABELLED: Immunotherapy is often effective only for small tumor burdens and, in many cases, is restricted to subcutaneous tumors. Here, we investigated the antitumor effects of combination therapy with GM-CSF and IL-12 on orthotopic liver tumors with intermediate or large tumor volumes, or on chemically-induced multifocal liver tumors in animals. Adenoviruses encoding GM-CSF or IL-12 were injected intratumorally to animals bearing transplanted tumors, or injected via intrahepatic artery in animals with primary multifocal liver tumors induced by diethylnitrosamine. Our results demonstrated that IL-12, but not GM-CSF, monotherapy displayed significant therapeutic effects, whereas combination therapy with both cytokines displayed synergistic antitumor effects not only on transplanted tumor models with intermediate or large tumor loads, but also on carcinogen-induced multifocal liver tumors. Effector cell analyses, revealed by in vivo cell subset depletion, flow cytometry analysis, and immunohistochemical staining of tumor infiltrates, indicated that NK cells were the prominent antitumor effectors for the IL-12-mediated antitumor activity, whereas CD8+ T cells, NKT cells, and macrophages were more important than NK cells in the combination therapy-mediated antitumor effects. Both IL-12 monotherapy and combination therapy could induce various types of effectors and high levels of IFN-gamma; however, the latter induced much higher levels than the former, which may explain why combination therapy is superior to IL-12 monotherapy. CONCLUSION: Combination therapy with GM-CSF and IL-12 represents a promising immunotherapy strategy for treating orthotopic, widespread liver tumors.


Asunto(s)
Inhibidores de la Angiogénesis/genética , Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interleucina-12/genética , Neoplasias Hepáticas/terapia , Adenoviridae/genética , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Interferón gamma/metabolismo , Interleucina-12/uso terapéutico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA