Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
J Biol Chem ; 300(4): 107168, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38490434

RESUMEN

Lipids have been previously implicated in the lifecycle of neuroinvasive viruses. However, the role of lipids in programmed cell death and the relationship between programmed cell death and lipid droplets (LDs) in neuroinvasive virus infection remains unclear. Here, we found that the infection of neuroinvasive virus, such as rabies virus and encephalomyocarditis virus could enhance the LD formation in N2a cells, and decreasing LDs production by targeting diacylglycerol acyltransferase could suppress viral replication. The lipidomics analysis revealed that arachidonic acid (AA) was significantly increased after reducing LD formation by restricting diacylglycerol acyltransferase, and AA was further demonstrated to induce ferroptosis to inhibit neuroinvasive virus replication. Moreover, lipid peroxidation and viral replication inhibition could be significantly alleviated by a ferroptosis inhibitor, ferrostatin-1, indicating that AA affected neuroinvasive virus replication mainly through inducing ferroptosis. Furthermore, AA was demonstrated to activate the acyl-CoA synthetase long-chain family member 4-lysophosphatidylcholine acyltransferase 3-cytochrome P450 oxidoreductase axis to induce ferroptosis. Our findings highlight novel cross-talks among viral infection, LDs, and ferroptosis for the first time, providing a potential target for antiviral drug development.


Asunto(s)
Ácido Araquidónico , Ferroptosis , Gotas Lipídicas , Replicación Viral , Ferroptosis/efectos de los fármacos , Gotas Lipídicas/metabolismo , Gotas Lipídicas/efectos de los fármacos , Animales , Replicación Viral/efectos de los fármacos , Ratones , Ácido Araquidónico/metabolismo , Ácido Araquidónico/farmacología , Virus de la Encefalomiocarditis/efectos de los fármacos , Diacilglicerol O-Acetiltransferasa/metabolismo , Diacilglicerol O-Acetiltransferasa/antagonistas & inhibidores , Peroxidación de Lípido/efectos de los fármacos , Coenzima A Ligasas/metabolismo , Línea Celular Tumoral , Humanos
2.
Antimicrob Agents Chemother ; 60(10): 6402-6, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27480860

RESUMEN

Encephalomyocarditis virus (EMCV), like hepatitis C virus (HCV), requires phosphatidylinositol 4-kinase IIIα (PI4KA) for genome replication. Here, we demonstrate that tyrphostin AG1478, a known epidermal growth factor receptor (EGFR) inhibitor, also inhibits PI4KA activity, both in vitro and in cells. AG1478 impaired replication of EMCV and HCV but not that of an EMCV mutant previously shown to escape PI4KA inhibition. This work uncovers novel cellular and antiviral properties of AG1478, a compound previously regarded only as a cancer chemotherapy agent.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Antivirales/farmacología , Virus de la Encefalomiocarditis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Quinazolinas/farmacología , Tirfostinos/farmacología , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Relación Dosis-Respuesta a Droga , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/fisiología , Células HeLa/efectos de los fármacos , Células HeLa/virología , Hepacivirus/fisiología , Humanos , Terapia Molecular Dirigida/métodos , Mutación , Replicación Viral/efectos de los fármacos
3.
J Interferon Cytokine Res ; 36(3): 192-203, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26824268

RESUMEN

Human serum albumin (HSA)-free formulation of Escherichia coli-derived human interferon beta (IFNß-1b) with a high percentage of monomeric protein and low immunogenicity is developed and characterized in the current study. UV spectroscopy, fluorescence spectroscopy, dynamic light scattering, sodium dodecyl sulfate polyacrylamide gel electrophoresis, Western blotting, Micro-Flow Imaging, resonant mass measurement, size exclusion, and reversed-phase high performance liquid chromatographies were applied to assess the effect of excipients on the stability of IFNß-1b to establish a HSA-free formulation. The antiviral activity of IFNß-1b was evaluated using human lung carcinoma cell line. Immune tolerant mice to hIFNß were used to assess the immunogenicity of the HSA-free formulated IFNß-1b in comparison to Betaferon drug product and Avonex drug substance as standards through IgG titering of plasma. HSA-free formulated IFNß-1b, including 200 mM L-arginine, 200 mM trehalose, and 0.1% n-dodecyl ß-D-maltoside in 10 mM sodium acetate buffer, pH 7.4, showed the highest biological activity. The stability of IFNß-1b in the HSA-free formulation was monitored for 3 weeks at 4°C and 37°C with relative humidity of 10% and 75%, respectively. Protein aggregation and immunogenicity in transgenic mice were decreased in the HSA-free formulated IFNß-1b compared to Betaferon. The stability, biological activity, and immunogenicity of the HSA-free formulation and Betaferon were evaluated. Incubation of formulations at 4°C and 37°C for 3 weeks showed that the HSA-free formulated IFNß-1b was more stable and less immunogenic in transgenic FVB/N mice. Low immunogenicity and the absence of HSA, which reduces the potential risk of viral infection (eg, HIV and HCV), are promising for clinical studies.


Asunto(s)
Anticuerpos Antivirales/sangre , Antivirales/farmacología , Virus de la Encefalomiocarditis/efectos de los fármacos , Interferon beta-1b/farmacología , Células A549 , Animales , Antivirales/inmunología , Antivirales/aislamiento & purificación , Clonación Molecular , Virus de la Encefalomiocarditis/crecimiento & desarrollo , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Tolerancia Inmunológica , Interferon beta-1b/biosíntesis , Interferon beta-1b/aislamiento & purificación , Ratones , Ratones Transgénicos , Estabilidad Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología
4.
J Interferon Cytokine Res ; 36(3): 180-91, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26700737

RESUMEN

Type I interferons (IFNs) exhibit broad-spectrum antiviral activity, with potential utility against emerging acute virus infections that pose a threat to global health. Recombinant IFN-αs that have been approved for clinical use require cold storage and are administered through intramuscular or subcutaneous injection, features that are problematic for global distribution, storage, and administration. Cognizant that the biological potency of an IFN-α subtype is determined by its binding affinity to the type I IFN receptor, IFNAR, we identified a panel of small molecule nonpeptide compounds using an in silico screening strategy that incorporated specific structural features of amino acids in the receptor-binding domains of the most potent IFN-α, IFN alfacon-1. Hit compounds were selected based on ease of synthesis and formulation properties. In preliminary biological assays, we provide evidence that these compounds exhibit antiviral activity. This proof-of-concept study validates the strategy of in silico design and development for IFN mimetics.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalomiocarditis/efectos de los fármacos , Interferón-alfa/química , Peptidomiméticos/farmacología , Receptor de Interferón alfa y beta/agonistas , Bibliotecas de Moléculas Pequeñas/farmacología , Antivirales/síntesis química , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Linfocitos B/virología , Línea Celular Tumoral , Simulación por Computador , Diseño de Fármacos , Virus de la Encefalomiocarditis/crecimiento & desarrollo , Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Modelos Moleculares , Peptidomiméticos/síntesis química , Estructura Secundaria de Proteína , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Proteínas Recombinantes/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Relación Estructura-Actividad , Interfaz Usuario-Computador
5.
PLoS One ; 10(7): e0130797, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26158644

RESUMEN

Type I interferons are multi-potent cytokines that serve as first line of defense against viruses and other pathogens, posses immunomudolatory functions and elicit a growth inhibitory response. In recent years it has been shown that interferons are also detrimental, for example in lupus, AIDS, tuberculosis and cognitive decline, highlighted the need to develop interferon antagonists. We have previously developed the antagonist IFN-1ant, with much reduced binding to the IFNAR1 receptor and enhanced binding to IFNAR2. Here, we further tune the IFN-1ant by producing three additional antagonists based on IFN-1ant but with altered activity profiles. We show that in all three cases the antiproliferative activity of interferons is blocked and the induction of gene transcription of immunomudolatory and antiproliferative associated genes are substantially decreased. Conversely, each of the new antagonists elicits a different degree of antiviral response, STAT phosphorylation and related gene induction. Two of the new antagonists promote decreased activity in relation to the original IFN-1ant, while one of them promotes increased activity. As we do not know the exact causes of the detrimental effects of IFNs, the four antagonists that were produced and analyzed provide the opportunity to investigate the extent of antagonistic and agonistic activity optimal for a given condition.


Asunto(s)
Interferón Tipo I/genética , Proteínas Mutantes/genética , Mutación , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Virus de la Encefalomiocarditis/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/metabolismo , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , Proteínas Mutantes/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Receptor de Interferón alfa y beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos
6.
Int J Biol Macromol ; 79: 459-68, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26003302

RESUMEN

In the report, three bioactive fractions from Cerrena unicolor: laccase (LAC), endopolysaccharides (c-EPL), and low molecular weight (ex-LMS) were tested for the first time towards their antiviral, immunostimulatory, cytotoxic and antiproliferative effect. The immunomodulatory activity was studied by means of THP-1-derived macrophages able to synthesize and secrete IL-6 and TNF-α. We used cervical carcinoma cell lines SiHa (ATCC, HTB-35) and CaSki (ATCC, CRL 1550) to determine antitumor activity and human skin fibroblasts (HSF) as a control. SiHa and L929 cell lines were used in the antiviral activity assay to propagate HHV-1 and EMCV, respectively. LAC was the most active against HSV at an early stage of viral replication, whereas the activity of laccase against EMCV was evident after incubation of the virus with LAC before and after the adsorption step. Moreover, the investigations showed that the fungal c-EPL fraction stimulated the production and secretion of TNF-α and IL-6 by THP-1-derived macrophages up to a level of 2000 pg/ml and 400 pg/ml, respectively. It was indicated for the first time that the LAC and ex-LMS fractions exhibited anticancer activity. This resulted from their cytotoxic or antiproliferative action against the investigated tumor cells at concentrations above 250 µg/ml and 10 µg/ml, respectively.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Antivirales/aislamiento & purificación , Polisacáridos Fúngicos/aislamiento & purificación , Proteínas Fúngicas/aislamiento & purificación , Factores Inmunológicos/aislamiento & purificación , Lacasa/aislamiento & purificación , Polyporaceae/química , Antineoplásicos/farmacología , Antivirales/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/fisiología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibroblastos/virología , Polisacáridos Fúngicos/farmacología , Proteínas Fúngicas/farmacología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/fisiología , Humanos , Factores Inmunológicos/farmacología , Interleucina-6/biosíntesis , Interleucina-6/metabolismo , Lacasa/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Macrófagos/virología , Polyporaceae/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Replicación Viral/efectos de los fármacos
7.
J Pept Sci ; 21(7): 554-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25810135

RESUMEN

Interferon-alpha (IFNα) is a cytokine that orchestrates innate and adaptive immune responses and potently inhibits proliferation of normal and tumor cells. These properties have warranted the use of IFNα in clinical practice for the treatment of several viral infections and malignancies. However, overexpression of IFNα leads to immunopathology observed in the context of chronic viral infections and autoimmune conditions. Thus, it is desirable to develop therapeutic approaches that aim at suppressing excessive IFNα production. To that end, artificial evolution of peptides from phage display libraries represents a strategy that seeks to disrupt the interaction between IFNα and its cell surface receptor and thus inhibit the ensuing biological effects. Mirror-image phage display that screens peptide libraries against the D-enantiomer is particularly attractive because it allows for identification of proteolysis-resistant D-peptide inhibitors. This approach, however, relies on the availability of chemically synthesized D-IFNα composed entirely of D-amino acids. Here, we describe the synthesis and biological properties of IFNα2b of 165 amino acid residues produced by native chemical ligation, which represents an important first step toward the discovery of D-peptide antagonists with potential therapeutic applications.


Asunto(s)
Interferón-alfa/síntesis química , Biblioteca de Péptidos , Péptidos/síntesis química , Técnicas de Síntesis en Fase Sólida/métodos , Secuencia de Aminoácidos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Disulfuros/química , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/crecimiento & desarrollo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/virología , Humanos , Interferón alfa-2 , Interferón-alfa/farmacología , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/farmacología , Cultivo Primario de Células , Pliegue de Proteína , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/farmacología , Estereoisomerismo , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
8.
AAPS PharmSciTech ; 15(6): 1619-29, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25142823

RESUMEN

Understanding the mechanism of aggregation of a therapeutic protein would not only ease the manufacturing processing but could also lead to a more stable finished product. Aggregation of recombinant interferon (IFNß-1b) was studied by heating, oxidizing, or seeding of unformulated monomeric solution. The formation of aggregates was monitored by dynamic light scattering (DLS) and UV spectroscopy. The autocatalytic monomer loss model was used to fit the data on aggregation rates. The influence of pre-nucleation on aggregation step was demonstrated by inducing the liquid samples containing a monomer form of folded IFNß-1b by heat and also an oxidizing agent. Results tend to suggest that the nucleus includes a single protein molecule which has been probably deformed. Seeding tests showed that aggregation of IFNß-1b was probably initiated when 1.0% (w/w) of monomers converted to nucleus form. Chemiluminescence spectroscopy analysis of the sample indicated the generation of 3.0 µM of hydrogen peroxide (H2O2) during nucleation stage of IFNß-1b aggregation. Arginine with a concentration of 200 mM was sufficient to suppress aggregation of IFNß-1b by decreasing the rate of pre-nucleation step. We proposed the formation of pre-nucleus structures prior to nucleation as the mechanism of aggregation of IFNß-1b. Furthermore, we have showed the positive anti-aggregation effect of arginine on pre-nucleation step.


Asunto(s)
Antivirales/química , Arginina/química , Excipientes/química , Interferón beta/química , Antivirales/farmacología , Línea Celular Tumoral , Efecto Citopatogénico Viral/efectos de los fármacos , Virus de la Encefalomiocarditis/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/química , Interferon beta-1b , Interferón beta/farmacología , Cinética , Luz , Modelos Químicos , Oxidación-Reducción , Agregado de Proteínas , Pliegue de Proteína , Proteínas Recombinantes/química , Dispersión de Radiación , Espectrofotometría Ultravioleta , Tecnología Farmacéutica/métodos
9.
J Virol ; 88(19): 11091-107, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25008939

RESUMEN

UNLABELLED: Few drugs targeting picornaviruses are available, making the discovery of antivirals a high priority. Here, we identified and characterized three compounds from a library of kinase inhibitors that block replication of poliovirus, coxsackievirus B3, and encephalomyocarditis virus. Using an in vitro translation-replication system, we showed that these drugs inhibit different stages of the poliovirus life cycle. A4(1) inhibited both the formation and functioning of the replication complexes, while E5(1) and E7(2) were most effective during the formation but not the functioning step. Neither of the compounds significantly inhibited VPg uridylylation. Poliovirus resistant to E7(2) had a G5318A mutation in the 3A protein. This mutation was previously found to confer resistance to enviroxime-like compounds, which target a phosphatidylinositol 4-kinase IIIß (PI4KIIIß)-dependent step in viral replication. Analysis of host protein recruitment showed that E7(2) reduced the amount of GBF1 on the replication complexes; however, the level of PI4KIIIß remained intact. E7(2) as well as another enviroxime-like compound, GW5074, interfered with viral polyprotein processing affecting both 3C- and 2A-dependent cleavages, and the resistant G5318A mutation partially rescued this defect. Moreover, E7(2) induced abnormal recruitment to membranes of the viral proteins; thus, enviroxime-like compounds likely severely compromise the interaction of the viral polyprotein with membranes. A4(1) demonstrated partial protection from paralysis in a murine model of poliomyelitis. Multiple attempts to isolate resistant mutants in the presence of A4(1) or E5(1) were unsuccessful, showing that effective broad-spectrum antivirals could be developed on the basis of these compounds. IMPORTANCE: Diverse picornaviruses can trigger multiple human maladies, yet currently, only hepatitis A virus and poliovirus can be controlled with vaccination. The development of antipicornavirus therapeutics is also facing significant difficulties because these viruses readily generate resistance to compounds targeting either viral or cellular factors. Here, we describe three novel compounds that effectively block replication of distantly related picornaviruses with minimal toxicity to cells. The compounds prevent viral RNA replication after the synthesis of the uridylylated VPg primer. Importantly, two of the inhibitors are strongly refractory to the emergence of resistant mutants, making them promising candidates for further broad-spectrum therapeutic development. Evaluation of one of the compounds in an in vivo model of poliomyelitis demonstrated partial protection from the onset of paralysis.


Asunto(s)
Antivirales/farmacología , Poliomielitis/tratamiento farmacológico , Poliovirus/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/genética , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Antivirales/química , Sistema Libre de Células , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/metabolismo , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Regulación Viral de la Expresión Génica , Células HeLa , Humanos , Ratones , Mutación , Poliomielitis/virología , Poliovirus/genética , Poliovirus/crecimiento & desarrollo , Poliproteínas/antagonistas & inhibidores , Poliproteínas/genética , Poliproteínas/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Proteínas Virales/genética , Proteínas Virales/metabolismo
10.
PLoS One ; 9(4): e94491, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24722666

RESUMEN

Most antiviral treatment options target the invading pathogen and unavoidably encounter loss of efficacy as the pathogen mutates to overcome replication restrictions. A good strategy for circumventing drug resistance, or for pathogens without treatment options, is to target host cell proteins that are utilized by viruses during infection. The small molecule WP1130 is a selective deubiquitinase inhibitor shown previously to successfully reduce replication of noroviruses and some other RNA viruses. In this study, we screened a library of 31 small molecule derivatives of WP1130 to identify compounds that retained the broad-spectrum antiviral activity of the parent compound in vitro but exhibited improved drug-like properties, particularly increased aqueous solubility. Seventeen compounds significantly reduced murine norovirus infection in murine macrophage RAW 264.7 cells, with four causing decreases in viral titers that were similar or slightly better than WP1130 (1.9 to 2.6 log scale). Antiviral activity was observed following pre-treatment and up to 1 hour postinfection in RAW 264.7 cells as well as in primary bone marrow-derived macrophages. Treatment of the human norovirus replicon system cell line with the same four compounds also decreased levels of Norwalk virus RNA. No significant cytotoxicity was observed at the working concentration of 5 µM for all compounds tested. In addition, the WP1130 derivatives maintained their broad-spectrum antiviral activity against other RNA viruses, Sindbis virus, LaCrosse virus, encephalomyocarditis virus, and Tulane virus. Thus, altering structural characteristics of WP1130 can maintain effective broad-spectrum antiviral activity while increasing aqueous solubility.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Nitrilos/farmacología , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Línea Celular , Cianoacrilatos , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/fisiología , Inhibidores Enzimáticos/química , Interacciones Huésped-Patógeno , Humanos , Virus La Crosse/efectos de los fármacos , Virus La Crosse/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/virología , Ratones , Nitrilos/química , Norovirus/efectos de los fármacos , Norovirus/fisiología , Virus Norwalk/efectos de los fármacos , Virus Norwalk/fisiología , Cultivo Primario de Células , Piridinas/química , Virus Sindbis/efectos de los fármacos , Virus Sindbis/fisiología , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Proteasas Ubiquitina-Específicas/metabolismo
11.
Elife ; 3: e01535, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550253

RESUMEN

The RIG-I-like receptors RIG-I, LGP2, and MDA5 initiate an antiviral response that includes production of type I interferons (IFNs). The nature of the RNAs that trigger MDA5 activation in infected cells remains unclear. Here, we purify and characterise LGP2/RNA complexes from cells infected with encephalomyocarditis virus (EMCV), a picornavirus detected by MDA5 and LGP2 but not RIG-I. We show that those complexes contain RNA that is highly enriched for MDA5-stimulatory activity and for a specific sequence corresponding to the L region of the EMCV antisense RNA. Synthesis of this sequence by in vitro transcription is sufficient to generate an MDA5 stimulatory RNA. Conversely, genomic deletion of the L region in EMCV generates viruses that are less potent at stimulating MDA5-dependent IFN production. Thus, the L region antisense RNA of EMCV is a key determinant of innate immunity to the virus and represents an RNA that activates MDA5 in virally-infected cells. DOI: http://dx.doi.org/10.7554/eLife.01535.001.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Virus de la Encefalomiocarditis/metabolismo , ARN Helicasas/metabolismo , ARN sin Sentido/metabolismo , ARN Viral/metabolismo , Animales , Antivirales/farmacología , Chlorocebus aethiops , ARN Helicasas DEAD-box/genética , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/inmunología , Regulación Viral de la Expresión Génica , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferones/genética , Interferones/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , ARN Helicasas/genética , ARN sin Sentido/genética , ARN Viral/genética , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Transducción de Señal , Transfección , Células Vero , Replicación Viral
12.
PLoS One ; 8(5): e63940, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696859

RESUMEN

The type III interferons (IFNs), comprising IFN-λ1, IFN-λ2, and IFN-λ3, behave similarly to IFN-α in eliciting antiviral, antitumor, and immune-modulating activities. Due to their more restricted cellular targets, IFN-λs are attractive as potential alternatives to existing therapeutic regimens based on IFN-αs. We have applied the DOCK-AND-LOCK™ method to improve the anti-proliferative potency of IFN-λ1 up to 1,000-fold in targeted cancer cell lines by tethering stabilized Fab dimers, derived from hRS7 (humanized anti-Trop-2), hMN-15 (humanized anti-CEACAM6), hL243 (humanized anti-HLA-DR), and c225 (chimeric anti-EGFR), to IFN-λ1 site-specifically, resulting in novel immunocytokines designated (E1)-λ1, (15)-λ1, (C2)-λ1, and (c225)-λ1, respectively. Targeted delivery of IFN-λ1 via (15)-λ1 or (c225)-λ1 to respective antigen-expressing cells also significantly increased antiviral activity when compared with non-targeting (C2)-λ1, as demonstrated in human lung adenocarcinoma cell line A549 by (15)-λ1 against encephalomyocarditis virus (EC50 = 22.2 pM versus 223 pM), and in human hepatocarcinoma cell line Huh-7 by (c225)-λ1 against hepatitis C virus (EC50 = 0.56 pM versus 91.2 pM). These promising results, which are attributed to better localization and stronger binding of IFN-λ1 to antibody-targeted cells, together with the favorable pharmacokinetic profile of (E1)-λ1 in mice (T(1/2) = 8.6 h), support further investigation of selective prototypes as potential antiviral and antitumor therapeutic agents.


Asunto(s)
Antineoplásicos/farmacología , Antivirales/farmacología , Fragmentos Fab de Inmunoglobulinas/química , Interferones/farmacología , Animales , Antineoplásicos/química , Antivirales/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Electroforesis en Gel de Poliacrilamida , Virus de la Encefalomiocarditis/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Hepacivirus/efectos de los fármacos , Humanos , Interferones/química , Ratones , Ratones Desnudos
13.
PLoS Pathog ; 8(7): e1002783, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792064

RESUMEN

Ubiquitin (Ub) is a vital regulatory component in various cellular processes, including cellular responses to viral infection. As obligate intracellular pathogens, viruses have the capacity to manipulate the ubiquitin (Ub) cycle to their advantage by encoding Ub-modifying proteins including deubiquitinases (DUBs). However, how cellular DUBs modulate specific viral infections, such as norovirus, is poorly understood. To examine the role of DUBs during norovirus infection, we used WP1130, a small molecule inhibitor of a subset of cellular DUBs. Replication of murine norovirus in murine macrophages and the human norovirus Norwalk virus in a replicon system were significantly inhibited by WP1130. Chemical proteomics identified the cellular DUB USP14 as a target of WP1130 in murine macrophages, and pharmacologic inhibition or siRNA-mediated knockdown of USP14 inhibited murine norovirus infection. USP14 is a proteasome-associated DUB that also binds to inositol-requiring enzyme 1 (IRE1), a critical mediator of the unfolded protein response (UPR). WP1130 treatment of murine macrophages did not alter proteasome activity but activated the X-box binding protein-1 (XBP-1) through an IRE1-dependent mechanism. In addition, WP1130 treatment or induction of the UPR also reduced infection of other RNA viruses including encephalomyocarditis virus, Sindbis virus, and La Crosse virus but not vesicular stomatitis virus. Pharmacologic inhibition of the IRE1 endonuclease activity partially rescued the antiviral effect of WP1130. Taken together, our studies support a model whereby induction of the UPR through cellular DUB inhibition blocks specific viral infections, and suggest that cellular DUBs and the UPR represent novel targets for future development of broad spectrum antiviral therapies.


Asunto(s)
Antivirales/farmacología , Nitrilos/farmacología , Norovirus/efectos de los fármacos , Piridinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/virología , Línea Celular , Línea Celular Tumoral , Cianoacrilatos , Proteínas de Unión al ADN/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/patogenicidad , Inhibidores Enzimáticos/farmacología , Humanos , Virus La Crosse/efectos de los fármacos , Virus La Crosse/patogenicidad , Macrófagos/virología , Proteínas de la Membrana/metabolismo , Ratones , Norovirus/fisiología , Virus Norwalk/efectos de los fármacos , Virus Norwalk/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción del Factor Regulador X , Virus Sindbis/efectos de los fármacos , Virus Sindbis/patogenicidad , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética , Replicación Viral/efectos de los fármacos , Proteína 1 de Unión a la X-Box
14.
J Interferon Cytokine Res ; 32(8): 368-77, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22509977

RESUMEN

The nuclear factor-kappa B (NFκB) signal transduction pathway plays an important role in immunity, inflammation, cell growth, and survival. Since dysregulation of this pathway results in high, constitutive NFκB activation in various cancers and immune disorders, the development of specific drugs to target this pathway has become a focus for treating these diseases. NFκB regulates various aspects of the cellular response to interferon (IFN). However, the role of the upstream regulator of the NFκB signaling pathway, the inhibitor of κB kinase (IKK) complex, on IFN function has not been examined. In the present study, we examined the effects of 2 IKK inhibitors, N-(1,8-Dimethylimidazo[1,2-a]quinoxalin-4-yl)-1,2-ethanediamine hydrochloride (BMS-345541) and 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA-1), on IFN action in several human glioma cell lines. IKK inhibitors inhibit glioma cell proliferation, as well as TNF-induced RelA (p65) nuclear translocation and NFκB-dependent IL8 gene expression. Importantly, BMS-345541 and TPCA-1 differentially inhibit IFN-induced gene expression, completely suppressing MX1 and GBP1 gene expression, while having only a minor effect on ISG15 expression. Furthermore, these IKK inhibitors displayed marked differences in blocking IFN-induced antiviral action against cytopathic effects and replication of vesicular stomatitis virus (VSV) and encephalomyocarditis virus (EMCV). Our results show that the IKK complex plays an important function in IFN-induced gene expression and antiviral activity. Since VSV and EMCV are oncolytic viruses used in cancer therapy, our results indicate the potential synergy in combining IKK inhibitors with oncolytic viruses.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Glioma/metabolismo , Quinasa I-kappa B/antagonistas & inhibidores , Imidazoles/farmacología , Interferón Tipo I/farmacología , Quinoxalinas/farmacología , Tiofenos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/fisiología , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/virología , Humanos , Quinasa I-kappa B/metabolismo , Interferón Tipo I/inmunología , Interleucina-8/genética , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/fisiología
15.
Int J Immunopathol Pharmacol ; 24(2): 499-504, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21658324

RESUMEN

In order to determine whether Blastoferon®, a biosimilar interferon (IFN)- beta 1a formulation, shares epitopes with other known IFN-beta products, a series of neutralization bioassays were performed with a set of well-characterized anti-IFN- beta monoclonal antibodies and human sera (World Health Organization Reference Reagents). The bioassay was the interferon-induced inhibition of virus cytopathic effect on human cells in culture (EMC virus and A-549 cells). Computer-calculated results were reported as Tenfold Reduction Units (TRU)/ml. To further assess Blastoferon® immunogenicity, in vivo production of anti-IFN beta antibodies was determined in sera of patients included in the pharmacovigilance plan of Blastoferon® by the level of IFN- beta 1a binding antibodies (by enzyme immunoassay -EIA) and neutralizing antibodies (in the Wish-VSV system). The highly characterized neutralizing monoclonal antibodies A1 and A5 that bind to specific regions of the IFN- beta molecule reacted positively with the three beta 1a IFNs: Blastoferon®, Rebif®, and the IFN- beta WHO Second International Standard 00/572. As expected, the non-neutralizing monoclonal antibodies B4 and B7 did not neutralize any of the IFN- beta preparations. The commercially available monoclonal antibody B-02 reacted essentially equally with Rebif® and Blastoferon®. The WHO Reference Reagent human serum anti-IFN- beta polyclonal antibody neutralized all the IFN- beta products, whereas the WHO Reference Reagent human serum anti-IFN-alpha polyclonal antibody G037-501-572 appropriately failed to react with any of the IFN- beta products. On the basis of in vitro reactivity with known, well-characterized monoclonal and polyclonal antibody preparations, Blastoferon® shares immunological determinants with other human interferon- beta products, especially IFN- beta 1a. In vivo antibodies were detected by EIA in 72.9% of 37 chronically treated multiple sclerosis patients, whereas neutralizing antibodies were found in 8.1% of them. Blastoferon® appears to have immunological characteristics comparable to other IFN- beta 1a products.


Asunto(s)
Adyuvantes Inmunológicos , Epítopos , Interferón beta/inmunología , Anticuerpos Monoclonales , Anticuerpos Neutralizantes/sangre , Línea Celular Tumoral , Efecto Citopatogénico Viral/efectos de los fármacos , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/patogenicidad , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Humanos , Interferón beta-1a , Interferon beta-1b , Interferón beta/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Pruebas de Neutralización
16.
Bioconjug Chem ; 22(6): 1017-20, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21618993

RESUMEN

Here, we describe a novel method for the site-specific C-terminal PEGylation of recombinant proteins. This general approach exploits chemical cleavage of precursor intein-fusion proteins with hydrazine to directly produce recombinant protein hydrazides. This unique functionality within the protein sequence then facilitates site-specific C-terminal modification by hydrazone-forming ligation reactions. This approach was used to generate folded, site-specifically C-terminal PEGylated IFNalpha2b and IFNbeta1b, which retained excellent antiviral activity, demonstrating the utility of this technology in the PEGylation of therapeutic proteins. As this methodology is straightforward to perform, is compatible with disulfide bonds, and is exclusively selective for the protein C-terminus, it shows great potential as general technology for the site-specific engineering and labeling of recombinant proteins.


Asunto(s)
Antivirales/química , Hidrazonas/química , Interferón-alfa/química , Interferón beta/química , Polietilenglicoles/química , Ingeniería de Proteínas/métodos , Proteínas Recombinantes/química , Antivirales/síntesis química , Antivirales/farmacología , Línea Celular Tumoral , Virus de la Encefalomiocarditis/efectos de los fármacos , Humanos , Interferón alfa-2 , Interferon beta-1b , Interferón-alfa/farmacología , Interferón beta/farmacología , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad
17.
Planta Med ; 77(7): 718-25, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21104606

RESUMEN

EPs® 7630 is an aqueous-ethanolic extract of the roots of Pelargonium sidoides, employed in the treatment of upper respiratory tract infections. Its anti-infective activity is supposed to be associated with the activation of the nonspecific immune system. Using Leishmania major GFP-infected murine BMMΦ, the NO production of EPs® 7630-activated macrophages was correlated with the reduction of the GFP signal measured at single cell levels using flow cytometry. The anti-infectious effect of EPs® 7630 (3-10 µg/mL) on its own (NO production: 4-13 µM; signal reduction: 25-73 %) was less prominent than that in combination with IFN- γ (100 U/mL) (NO production: 20-27 µM; signal reduction: 35-78 %). Furthermore, supernatants of EPs® 7630-stimulated BMMΦ (10 µg/mL) significantly reduced the cytopathic effect of EMCV on L929 fibroblasts (antiviral activity 80 U/mL) when compared with an IFN- γ standard (100 U/mL). Direct addition of EPs® 7630 to L929 did not mediate cytoprotective effects. The antiviral components induced in BMMΦ by EPs® 7630 remain to be identified. Detection of any IFNs by ELISA was unsuccessful, which may be due to their very low concentrations in cell supernatants. The current data provide convincing support for the induction of anti-infectious responses by EPs® 7630.


Asunto(s)
Antiinfecciosos/farmacología , Leishmania major/efectos de los fármacos , Leishmania major/metabolismo , Macrófagos/efectos de los fármacos , Óxido Nítrico/biosíntesis , Extractos Vegetales/farmacología , Animales , Antivirales/farmacología , Betaretrovirus/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Virus de la Encefalomiocarditis/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Humanos , Leishmaniasis Cutánea/tratamiento farmacológico , Leishmaniasis Cutánea/parasitología , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL , Pelargonium/química , Raíces de Plantas/química
18.
J Gen Virol ; 90(Pt 3): 536-545, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19218198

RESUMEN

The interferon (IFN) system is a major effector of the innate immunity that allows time for the subsequent establishment of an adaptive immune response against a wide-range of pathogens. Their diverse biological actions are thought to be mediated by the products of specific but usually overlapping sets of cellular genes induced in the target cells. Ubiquitin ligase members of the tripartite motif (TRIM) protein family have emerged as IFN-induced proteins involved in both innate and adaptive immunity. In this report, we provide evidence that TRIM22 is a functional E3 ubiquitin ligase that is also ubiquitinated itself. We demonstrate that TRIM22 expression leads to a viral protection of HeLa cells against encephalomyocarditis virus infections. This effect is dependent upon its E3 ubiquitinating activity, since no antiviral effect was observed in cells expressing a TRIM22-deletion mutant defective in ubiquitinating activity. Consistent with this, TRIM22 interacts with the viral 3C protease (3C(PRO)) and mediates its ubiquitination. Altogether, our findings demonstrate that TRIM22 E3 ubiquitin ligase activity represents a new antiviral pathway induced by IFN against picornaviruses.


Asunto(s)
Antivirales/metabolismo , Cisteína Endopeptidasas/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Proteasas Virales 3C , Antivirales/farmacología , Línea Celular , Cisteína Endopeptidasas/genética , Virus de la Encefalomiocarditis/enzimología , Virus de la Encefalomiocarditis/genética , Células HeLa , Humanos , Antígenos de Histocompatibilidad Menor , Proteínas Represoras/genética , Proteínas Represoras/farmacología , Proteínas de Motivos Tripartitos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/farmacología , Proteínas Virales/genética
19.
Mol Gen Mikrobiol Virusol ; (3): 37-40, 2008.
Artículo en Ruso | MEDLINE | ID: mdl-18756823

RESUMEN

The avian recombinant adenovirus of serotype 1 (CELO) was obtained. The recombinant adenovirus of serotype 1 (CELO) induces expression of human beta-interferon (IB). The expression cassette containing IB gene was placed at the right end of the CELO genome under control of hybrid promoter hEF-1alpha/HTLV. The resulting recombinant adenovirus CELO-IB transduced the avian cell culture LMH. The level of production of the recombinant IB was 0.15 micro/ml. The IB protein yield after affine chromatography purification using Ni-NTA agarose was 50%. The biological activity of the purified IB was high (7.8 x 10(8) MU/microg protein). The purified IB inhibited replication of murine encephalomyocarditis virus (VMEC) in cell culture of human diploid fibroblasts (HDF). Thus, expression system based on avian cell culture is an effective system for producing biologically active protein of human interferon beta.


Asunto(s)
Aviadenovirus/genética , Interferón Tipo I/biosíntesis , Animales , Células Cultivadas , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/fisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/farmacología , Proteínas Recombinantes , Replicación Viral
20.
Proc Natl Acad Sci U S A ; 105(12): 4808-13, 2008 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18339807

RESUMEN

Multiple signaling pathways are engaged by the type I and II IFN receptors, but their specific roles and possible coordination in the generation of IFN-mediated biological responses remain unknown. We provide evidence that activation of Akt kinases is required for IFN-inducible engagement of the mTOR/p70 S6 kinase pathway. Our data establish that Akt activity is essential for up-regulation of key IFN-alpha- and IFN-gamma-inducible proteins, which have important functional consequences in the induction of IFN responses. Such effects of the Akt pathway are unrelated to regulatory activities on IFN-dependent STAT phosphorylation/activation or transcriptional regulation. By contrast, they reflect regulatory activities on mRNA translation via direct control of the mTOR pathway. In studies using Akt1 and Akt2 double knockout cells, we found that the absence of Akt kinases results in dramatic reduction in IFN-induced antiviral responses, establishing a critical role of the Akt pathway in IFN signaling. Thus, activation of the Akt pathway by the IFN receptors complements the function of IFN-activated JAK-STAT pathways, by allowing mRNA translation of IFN-stimulated genes and, ultimately, the induction of the biological effects of IFNs.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Interferón-alfa/farmacología , Interferón gamma/farmacología , Biosíntesis de Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/genética , Citocinas/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/inmunología , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/virología , Ratones , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Transcripción Genética/efectos de los fármacos , Ubiquitinas/genética , Ubiquitinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA