Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 13(6): e1006343, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28594932

RESUMEN

Hepatitis C virus (HCV) RNA is synthesized by the replicase complex (RC), a macromolecular assembly composed of viral non-structural proteins and cellular co-factors. Inhibitors of the HCV NS5A protein block formation of new RCs but do not affect RNA synthesis by pre-formed RCs. Without new RC formation, existing RCs turn over and are eventually lost from the cell. We aimed to use NS5A inhibitors to estimate the half-life of the functional RC of HCV. We compared different cell culture-infectious strains of HCV that may be grouped based on their sensitivity to lipid peroxidation: robustly replicating, lipid peroxidation resistant (LPOR) viruses (e.g. JFH-1 or H77D) and more slowly replicating, lipid peroxidation sensitive (LPOS) viruses (e.g. H77S.3 and N.2). In luciferase assays, LPOS HCV strains declined under NS5A inhibitor therapy with much slower kinetics compared to LPOR HCV strains. This difference in rate of decline was not observed for inhibitors of the NS5B RNA-dependent RNA polymerase suggesting that the difference was not simply a consequence of differences in RNA stability. In further analyses, we compared two isoclonal HCV variants: the LPOS H77S.3 and the LPOR H77D that differ only by 12 amino acids. Differences in rate of decline between H77S.3 and H77D following NS5A inhibitor addition were not due to amino acid sequences in NS5A but rather due to a combination of amino acid differences in the non-structural proteins that make up the HCV RC. Mathematical modeling of intracellular HCV RNA dynamics suggested that differences in RC stability (half-lives of 3.5 and 9.9 hours, for H77D and H77S.3, respectively) are responsible for the different kinetics of antiviral suppression between LPOS and LPOR viruses. In nascent RNA capture assays, the rate of RNA synthesis decline following NS5A inhibitor addition was significantly faster for H77D compared to H77S.3 indicating different half-lives of functional RCs.


Asunto(s)
Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Hepatitis C/virología , Replicación Viral/efectos de los fármacos , Semivida , Hepacivirus/química , Hepacivirus/clasificación , Humanos , Cinética , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Ensamble de Virus/efectos de los fármacos
3.
Nat Microbiol ; 2: 16247, 2016 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-27991882

RESUMEN

With a single exception, all isolates of hepatitis C virus (HCV) require adaptive mutations to replicate efficiently in cell culture. Here, we show that a major class of adaptive mutations regulates the activity of a cellular lipid kinase, phosphatidylinositol 4-kinase IIIα (PI4KA). HCV needs to stimulate PI4KA to create a permissive phosphatidylinositol 4-phosphate-enriched membrane microenvironment in the liver and in primary human hepatocytes (PHHs). In contrast, in Huh7 hepatoma cells, the virus must acquire loss-of-function mutations that prevent PI4KA overactivation. This adaptive mechanism is necessitated by increased PI4KA levels in Huh7 cells compared with PHHs, and is conserved across HCV genotypes. PI4KA-specific inhibitors promote replication of unadapted viral isolates and allow efficient replication of patient-derived virus in cell culture. In summary, this study has uncovered a long-sought mechanism of HCV cell-culture adaptation and demonstrates how a virus can adapt to changes in a cellular environment associated with tumorigenesis.

4.
Eur J Immunol ; 46(7): 1622-32, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27129615

RESUMEN

IL-10 is an anti-inflammatory cytokine that inhibits maturation and cytokine production of dendritic cells (DCs). Although mature DCs have the unique capacity to prime CD8(+) CTL, IL-10 can promote CTL responses. To understand these paradoxic findings, we analyzed the role of IL-10 produced by human APC subsets in T-cell responses. IL-10 production was restricted to CD1c(+) DCs and CD14(+) monocytes. Interestingly, it was differentially regulated, since R848 induced IL-10 in DCs, but inhibited IL-10 in monocytes. Autocrine IL-10 had only a weak inhibitory effect on DC maturation, cytokine production, and CTL priming with high-affinity peptides. Nevertheless, it completely blocked cross-priming and priming with low-affinity peptides of a self/tumor-antigen. IL-10 also inhibited CD1c(+) DC-induced CD4(+) T-cell priming and enhanced Foxp3 induction, but was insufficient to induce T-cell IL-10 production. CD1c(+) DC-derived IL-10 had also no effect on DC-induced secondary expansions of memory CTL. However, IL-15-driven, TCR-independent proliferation of memory CTL was enhanced by IL-10. We conclude that DC-derived IL-10 selects high-affinity CTL upon priming. Moreover, IL-10 preserves established CTL memory by enhancing IL-15-dependent homeostatic proliferation. These combined effects on CTL priming and memory maintenance provide a plausible mechanism how IL-10 promotes CTL responses in humans.


Asunto(s)
Antígenos CD1/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Glicoproteínas/metabolismo , Memoria Inmunológica/inmunología , Interleucina-10/metabolismo , Activación de Linfocitos/inmunología , Presentación de Antígeno/inmunología , Comunicación Autocrina/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Reactividad Cruzada/inmunología , Citocinas/metabolismo , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Monocitos/inmunología , Monocitos/metabolismo
5.
PLoS One ; 10(9): e0138546, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26405760

RESUMEN

Hepatitis C virus (HCV) variants characterized by genomic deletions in the structural protein region have been sporadically detected in liver and serum of hepatitis C patients. These defective genomes are capable of autonomous RNA replication and are packaged into infectious viral particles in cells co-infected with the wild-type virus. The prevalence of such forms in the chronically HCV-infected population and the impact on the severity of liver disease or treatment outcome are currently unknown. In order to determine the prevalence of HCV defective variants and to study their association with clinical characteristics, a screening campaign was performed on pre-therapy serum samples from a well-characterized cohort of previously untreated genotype 1 HCV-infected patients who received treatment with PEG-IFNα and RBV. 132 subjects were successfully analyzed for the presence of defective species exploiting a long-distance nested PCR assay. HCV forms with deletions predominantly affecting E1, E2 and p7 proteins were found in a surprising high fraction of the subjects (25/132, 19%). Their presence was associated with patient older age, higher viral load and increased necroinflammatory activity in the liver. While the presence of circulating HCV carrying deletions in the E1-p7 region did not appear to significantly influence sustained virological response rates to PEG-IFNα/RBV, our study indicates that the presence of these subgenomic HCV mutants could be associated with virological relapse in patients who did not have detectable viremia at the end of the treatment.


Asunto(s)
Hepacivirus/genética , Hepatitis C Crónica/virología , Hígado/patología , Eliminación de Secuencia , Proteínas Virales/genética , Adulto , Factores de Edad , Anciano , Femenino , Genotipo , Hepatitis C Crónica/sangre , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/patología , Humanos , Interferón alfa-2 , Interferón-alfa/uso terapéutico , Hígado/virología , Masculino , Persona de Mediana Edad , Polietilenglicoles/uso terapéutico , Prevalencia , Proteínas Recombinantes/uso terapéutico , Recurrencia , Ribavirina/uso terapéutico , Proteínas del Envoltorio Viral/genética , Carga Viral
6.
Gastroenterology ; 147(2): 453-62.e7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24768676

RESUMEN

BACKGROUND & AIMS: All-oral regimens combining different classes of direct-acting antivirals (DAA) are highly effective for treatment of patients with chronic hepatitis C. NS5A inhibitors will likely form a component of future interferon-sparing treatment regimens. However, despite their potential, the detailed mechanism of action of NS5A inhibitors is unclear. To study their mechanisms, we compared their kinetics of antiviral suppression with those of other classes of DAA, using the hepatitis C virus genotype 1a cell culture-infectious virus H77S.3. METHODS: We performed detailed kinetic analyses of specific steps in the hepatitis C virus life cycle using cell cultures incubated with protease inhibitors, polymerase inhibitors, or NS5A inhibitors. Assays were designed to measure active viral RNA synthesis and steady-state RNA abundance, polyprotein synthesis, virion assembly, and infectious virus production. RESULTS: Despite their high potency, NS5A inhibitors were slow to inhibit viral RNA synthesis compared with protease or polymerase inhibitors. By 24 hours after addition of an NS5A inhibitor, polyprotein synthesis was reduced <50%, even at micromolar concentrations. In contrast, inhibition of virus release by NS5A inhibitors was potent and rapid, with onset of inhibition as early as 2 hours. Cells incubated with NS5A inhibitors were rapidly depleted of intracellular infectious virus and RNA-containing hepatitis C virus particles, indicating a block in virus assembly. CONCLUSIONS: DAAs that target NS5A rapidly inhibit intracellular assembly of genotype 1a virions. They also inhibit formation of functional replicase complexes, but have no activity against preformed replicase, thereby resulting in slow shut-off of viral RNA synthesis.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Ensamble de Virus/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral , Genotipo , Hepacivirus/enzimología , Hepacivirus/genética , Hepacivirus/crecimiento & desarrollo , Humanos , Cinética , Mutación , Inhibidores de Proteasas/farmacología , ARN Viral/biosíntesis , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
7.
Gastroenterology ; 146(5): 1373-85.e1-11, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24512803

RESUMEN

BACKGROUND & AIMS: Positive-sense RNA viruses remodel intracellular membranes to generate specialized membrane compartments for viral replication. Several RNA viruses, including poliovirus and hepatitis C virus (HCV), require phosphatidylinositol (PI) 4-kinases for their replication. However, it is not known how PI 4-kinases and their product, PI(4)P, facilitate host membrane reorganization and viral replication. In addition, although the HCV replication compartment, known as the membranous web, is believed to be cholesterol enriched, the mechanisms by which this occurs have not been elucidated. We aimed to identify and characterize a PI 4-kinase effector in HCV replication. METHODS: We used a combination of microscopic and biochemical methods to study HCV replication, web morphology, the distribution of intracellular protein and PI(4)P, along with cholesterol trafficking in HCV-infected cells. PI 4-kinase and oxysterol-binding protein (OSBP) were inhibited using RNA interference or small molecules in cells expressing a full-length genotype 1b replicon or infected with the JFH-1 strain of HCV. RESULTS: OSBP was required for HCV replication and membranous web integrity. OSBP was recruited to membranous webs in a PI 4-kinase-dependent manner, and both these factors were found to regulate cholesterol trafficking to the web. We also found OSBP to be required for poliovirus infection but dispensable for dengue virus. CONCLUSIONS: OSBP is a PI 4-kinase effector in HCV infection, and contributes to the integrity and cholesterol enrichment of the membranous web. OSBP might also be a PI 4-kinase effector in poliovirus infection and could be involved in replication of other viruses that require PI 4-kinases.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/metabolismo , Membrana Celular/enzimología , Colesterol/metabolismo , Hepacivirus/enzimología , Receptores de Esteroides/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , Transporte Biológico , Membrana Celular/efectos de los fármacos , Genotipo , Células HEK293 , Células HeLa , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Hepacivirus/crecimiento & desarrollo , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Fenotipo , Fosfatos de Fosfatidilinositol/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Viral/biosíntesis , Receptores de Esteroides/genética , Transfección , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
8.
Blood ; 122(6): 932-42, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23794066

RESUMEN

Dendritic cells (DC) have the unique capacities to induce primary T-cell responses. In mice, CD8α(+)DC are specialized to cross-prime CD8(+) T cells and produce interleukin-12 (IL-12) that promotes cytotoxicity. Human BDCA-3(+)DC share several relevant characteristics with CD8α(+)DC, but the capacities of human DC subsets to induce CD8(+) T-cell responses are incompletely understood. Here we compared CD1c(+) myeloid DC (mDC)1, BDCA-3(+)mDC2, and plasmacytoid DC (pDC) in peripheral blood and lymphoid tissues for phenotype, cytokine production, and their capacities to prime cytotoxic T cells. mDC1 were surprisingly the only human DC that secreted high amounts of IL-12p70, but they required combinational Toll-like receptor (TLR) stimulation. mDC2 and pDC produced interferon-λ and interferon-α, respectively. Importantly, mDC1 and mDC2 required different combinations of TLR ligands to cross-present protein antigens to CD8(+) T cells. pDC were inefficient and also expressed lower levels of major histocompatibility complex and co-stimulatory molecules. Nevertheless, all DC induced CD8(+) memory T-cell expansions upon licensing by CD4(+) T cells, and primed naive CD8(+) T cells following appropriate TLR stimulation. However, because mDC1 produced IL-12, they induced the highest levels of cytotoxic molecules. In conclusion, CD1c(+)mDC1 are the relevant source of IL-12 for naive T cells and are fully equipped to cross-prime cytotoxic T-cell responses.


Asunto(s)
Antígenos CD1/metabolismo , Células Dendríticas/citología , Glicoproteínas/metabolismo , Interleucina-12/metabolismo , Linfocitos T Citotóxicos/citología , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Proliferación Celular , Separación Celular , Citocinas/metabolismo , Células Dendríticas/metabolismo , Humanos , Memoria Inmunológica , Interferón-alfa/metabolismo , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Fenotipo , Receptores Toll-Like/metabolismo
9.
Curr Top Microbiol Immunol ; 369: 289-320, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23463206

RESUMEN

The major targets for direct-acting antivirals (DAAs) are the NS3/4A protease, the NS5A protein, and the NS5B polymerase. The latter enzyme offers several target sites: the catalytic domain for nucleoside/nucleotide analogs and different allosteric sites for non-nucleoside inhibitors. Two protease inhibitors have already been approved and more than 40 new NS3/4A, NS5A, or NS5B inhibitors are in development pipeline. Not only these agents can achieve very high cure rates when combined with PEG-IFN and RBV, but have also started to provide promising results when combined in IFN-free, all-oral combinations. In addition to the more canonical drug targets, new alternative viral targets for small molecule drug development are emerging, such as p7 or NS4B. Current research is focusing on defining the most efficacious DAA combination regimens, i.e., those which provide the highest rates of viral eradication, broadest spectrum of action, minimal or no clinical resistance, shortest treatment duration, and good tolerability.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Animales , Hepacivirus/enzimología , Hepacivirus/genética , Hepacivirus/fisiología , Hepatitis C/virología , Humanos , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/farmacología , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
10.
PLoS Pathog ; 8(3): e1002576, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22412376

RESUMEN

4-anilino quinazolines have been identified as inhibitors of HCV replication. The target of this class of compounds was proposed to be the viral protein NS5A, although unequivocal proof has never been presented. A 4-anilino quinazoline moiety is often found in kinase inhibitors, leading us to formulate the hypothesis that the anti-HCV activity displayed by these compounds might be due to inhibition of a cellular kinase. Type III phosphatidylinositol 4-kinase α (PI4KIIIα) has recently been identified as a host factor for HCV replication. We therefore evaluated AL-9, a compound prototypical of the 4-anilino quinazoline class, on selected phosphatidylinositol kinases. AL-9 inhibited purified PI4KIIIα and, to a lesser extent, PI4KIIIß. In Huh7.5 cells, PI4KIIIα is responsible for the phosphatidylinositol-4 phosphate (PI4P) pool present in the plasma membrane. Accordingly, we observed a gradual decrease of PI4P in the plasma membrane upon incubation with AL-9, indicating that this agent inhibits PI4KIIIα also in living cells. Conversely, AL-9 did not affect the level of PI4P in the Golgi membrane, suggesting that the PI4KIIIß isoform was not significantly inhibited under our experimental conditions. Incubation of cells expressing HCV proteins with AL-9 induced abnormally large clusters of NS5A, a phenomenon previously observed upon silencing PI4KIIIα by RNA interference. In light of our findings, we propose that the antiviral effect of 4-anilino quinazoline compounds is mediated by the inhibition of PI4KIIIα and the consequent depletion of PI4P required for the HCV membranous web. In addition, we noted that HCV has a profound effect on cellular PI4P distribution, causing significant enrichment of PI4P in the HCV-membranous web and a concomitant depletion of PI4P in the plasma membrane. This observation implies that HCV--by recruiting PI4KIIIα in the RNA replication complex--hijacks PI4P metabolism, ultimately resulting in a markedly altered subcellular distribution of the PI4KIIIα product.


Asunto(s)
1-Fosfatidilinositol 4-Quinasa/metabolismo , Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Fosfatos de Fosfatidilinositol/metabolismo , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/química , Dominio Catalítico/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Inhibidores Enzimáticos/farmacología , Hepacivirus/patogenicidad , Hepatocitos/metabolismo , Hepatocitos/virología , Interacciones Huésped-Patógeno , Humanos , Quinazolinas/farmacología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
11.
Bioorg Med Chem Lett ; 19(6): 1779-83, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19216075

RESUMEN

The RNA replication machinery of HCV is a multi-subunit membrane-associated complex. NS5A has emerged as an active component of HCV replicase, possibly involved in regulation of viral replication and resistance to the antiviral effect of interferon. We report here substituted piperazinyl-N-(aryl)benzamides as potent inhibitors of HCV replication exerted via modulation of the dimerization of NS5A.


Asunto(s)
Benzamidas/síntesis química , Hepacivirus/genética , Hepatitis C/tratamiento farmacológico , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Antivirales/química , Benzamidas/farmacología , Cristalografía por Rayos X/métodos , Dimerización , Hepacivirus/fisiología , Humanos , Interferones/química , Modelos Químicos , Conformación Molecular , Mutación , Relación Estructura-Actividad
12.
Methods Mol Biol ; 510: 95-110, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19009256

RESUMEN

NS5A phosphorylation can be studied in two ways: in living cells and in vitro. The former has several advantages: NS5A phosphorylation takes place in a cellular background and therefore might mimic more closely the real in vivo situation. Viral proteins and cellular kinases are in the correct cellular compartments, and dynamic processes like viral polyprotein processing and cellular signaling are in place. The disadvantage of this system is its great complexity, which makes limiting an observed effect to a single, well-defined agent, for example, a kinase, very difficult. NS5A phosphorylation in cells can easily be followed by metabolic labeling with either (35)S-methionine or (32)P-orthophosphate. The effect of a single, well-defined kinase on NS5A phosphorylation can be investigated in cells either by overexpression of this kinase in the presence of NS5A or by RNA interference of this kinase. If available, specific kinase inhibitors can be used to reveal the effect of this inhibition on NS5A phosphorylation. The problem with this approach is that only very few really specific kinase inhibitors are available. Biochemical in vitro experiments use purified components. This type of experiment allows direct investigation of the activity of a single kinase on NS5A as a substrate. In addition, the precise phosphorylation sites of a kinase can be mapped when NS5A-derived peptides are used instead of a full-length recombinant protein. Kinase inhibitors, which show a particular effect on NS5A phosphorylation in cells, can be retested in vitro on a particular kinase candidate. The problem with this approach is that purified components, like the purified NS5A substrate and the kinase of interest, are not always available.


Asunto(s)
Fosfoproteínas/análisis , Proteínas no Estructurales Virales/metabolismo , Extractos Celulares/inmunología , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Expresión Génica , Hepatitis C/metabolismo , Humanos , Inmunoprecipitación , Péptidos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Fosfotransferasas/genética , Fosfotransferasas/aislamiento & purificación , Fosfotransferasas/metabolismo , Interferencia de ARN , Proteínas Recombinantes/análisis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Coloración y Etiquetado , Transfección , Vaccinia/genética , Proteínas no Estructurales Virales/análisis , Proteínas no Estructurales Virales/inmunología , Replicación Viral
13.
J Biol Chem ; 283(39): 26694-704, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18614528

RESUMEN

Histone deacetylases (HDACs) regulate chromatin status and gene expression, and their inhibition is of significant therapeutic interest. To date, no biological substrate for class IIa HDACs has been identified, and only low activity on acetylated lysines has been demonstrated. Here, we describe inhibitor-bound and inhibitor-free structures of the histone deacetylase-4 catalytic domain (HDAC4cd) and of an HDAC4cd active site mutant with enhanced enzymatic activity toward acetylated lysines. The structures presented, coupled with activity data, provide the molecular basis for the intrinsically low enzymatic activity of class IIa HDACs toward acetylated lysines and reveal active site features that may guide the design of class-specific inhibitors. In addition, these structures reveal a conformationally flexible structural zinc-binding domain conserved in all class IIa enzymes. Importantly, either the mutation of residues coordinating the structural zinc ion or the binding of a class IIa selective inhibitor prevented the association of HDAC4 with the N-CoR.HDAC3 repressor complex. Together, these data suggest a key role of the structural zinc-binding domain in the regulation of class IIa HDAC functions.


Asunto(s)
Histona Desacetilasas/química , Proteínas Represoras/química , Zinc/química , Acetilación , Sitios de Unión/fisiología , Línea Celular , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Cristalografía por Rayos X , Histona Desacetilasas/metabolismo , Humanos , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Proteínas Represoras/metabolismo , Zinc/metabolismo
14.
Bioorg Med Chem Lett ; 18(6): 1814-9, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18308563

RESUMEN

It has been widely debated whether class IIa HDACs have catalytic deacetylase activity, and whether this plays any part in controlling gene expression. Herein, it has been demonstrated that class IIa HDACs isolated from mammalian cells are contaminated with other deacetylases, but can be prepared cleanly in Escherichia coli. These bacteria preparations have weak but measurable deacetylase activity. The low efficiency can be restored either by: mutation of an active site histidine to tyrosine, or by the use of a non-acetylated lysine substrate, allowing the development of assays to identify class IIa HDAC inhibitors.


Asunto(s)
Histona Desacetilasas/metabolismo , Acetilación , Benzamidas/farmacología , Catálisis , Células Cultivadas , Escherichia coli/enzimología , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/clasificación , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/farmacología , Riñón/enzimología , Estructura Molecular , Mutación , Péptidos Cíclicos/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Sulfonamidas
15.
J Biol Chem ; 282(8): 5536-44, 2007 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-17166835

RESUMEN

The hepatitis C virus encodes a single polyprotein that is processed by host and viral proteases to yield at least 10 mature viral proteins. The nonstructural (NS) protein 5A is a phosphoprotein, and experimental data indicate that the phosphorylation state of NS5A is important for the outcome of viral RNA replication. We were able to identify kinase inhibitors that specifically inhibit the formation of the hyperphosphorylated form of NS5A (p58) in cells. These kinase inhibitors were used for inhibitor affinity chromatography in order to identify the cellular targets of these compounds. The kinases casein kinase I (CKI), p38 MAPK, CIT (Citron Rho-interacting kinase), GAK, JNK2, PKA, RSK1/2, and RIPK2 were identified in the high affinity binding fractions of two NS5A hyperphosphorylation inhibitors (NS5A-p58-i). Even though these kinases are targets of the NS5A-p58-i, the only kinase showing an effect on NS5A hyperphosphorylation was confirmed to be CKI-alpha. Although this finding does not exclude the possibility that other kinase(s) might be involved in basal or regulatory phosphorylation of NS5A, we show here that NS5A is a direct substrate of CKI-alpha. Moreover, in vitro phosphorylation of NS5A by CKI-alpha resulted for the first time in the production of basal and hyperphosphorylated forms resembling those produced in cells. In vitro kinase reactions performed with NS5A peptides show that Ser-2204 is a preferred substrate residue for CKI-alpha after pre-phosphorylation of Ser-2201.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Hepacivirus/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología , Animales , Caseína Quinasa Ialfa/química , Caseína Quinasa Ialfa/aislamiento & purificación , Línea Celular , Cromatografía de Afinidad , Hepacivirus/química , Humanos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/aislamiento & purificación , Replicación Viral/efectos de los fármacos
16.
J Virol ; 80(22): 11305-12, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16943283

RESUMEN

Hepatitis C virus (HCV) has been the subject of intensive studies for nearly two decades. Nevertheless, some aspects of the virus life cycle are still a mystery. The HCV nonstructural protein 5A (NS5A) has been shown to be a modulator of cellular processes possibly required for the establishment of viral persistence. NS5A is heavily phosphorylated, and a switch between a basally phosphorylated form of NS5A (p56) and a hyperphosphorylated form of NS5A (p58) seems to play a pivotal role in regulating HCV replication. Using kinase inhibitors that specifically inhibit the formation of NS5A-p58 in cells, we identified the CKI kinase family as a target. NS5A-p58 increased upon overexpression of CKI-alpha, CKI-delta, and CKI-epsilon, whereas the RNA interference of only CKI-alpha reduced NS5A hyperphosphorylation. Rescue of inhibition of NS5A-p58 was achieved by CKI-alpha overexpression, and we demonstrated that the CKI-alpha isoform is targeted by NS5A hyperphosphorylation inhibitors in living cells. Finally, we showed that down-regulation of CKI-alpha attenuates HCV RNA replication.


Asunto(s)
Caseína Quinasa Ialfa/metabolismo , Hepacivirus/fisiología , Proteínas no Estructurales Virales/metabolismo , Caseína Cinasa 1 épsilon/genética , Caseína Cinasa 1 épsilon/metabolismo , Caseína Quinasa Ialfa/genética , Quinasa Idelta de la Caseína/genética , Quinasa Idelta de la Caseína/metabolismo , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Inhibidores Enzimáticos/farmacología , Humanos , Fosforilación , Interferencia de ARN
17.
J Virol ; 78(23): 13306-14, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15542681

RESUMEN

Efficient replication of hepatitis C virus (HCV) subgenomic RNA in cell culture requires the introduction of adaptive mutations. In this report we describe a system which enables efficient replication of the Con1 subgenomic replicon in Huh7 cells without the introduction of adaptive mutations. The starting hypothesis was that high amounts of the NS5A hyperphosphorylated form, p58, inhibit replication and that reduction of p58 by inhibition of specific kinase(s) below a certain threshold enables HCV replication. Upon screening of a panel of kinase inhibitors, we selected three compounds which inhibited NS5A phosphorylation in vitro and the formation of NS5A p58 in cell culture. Cells, transfected with the HCV Con1 wild-type sequence, support HCV RNA replication upon addition of any of the three compounds. The effect of the kinase inhibitors was found to be synergistic with coadaptive mutations in NS3. This is the first direct demonstration that the presence of high amounts of NS5A-p58 causes inhibition of HCV RNA replication in cell culture and that this inhibition can be relieved by kinase inhibitors.


Asunto(s)
Hepacivirus/genética , Proteínas Quinasas/fisiología , ARN Viral/biosíntesis , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Células Cultivadas , Humanos , Fosforilación , Replicón
18.
J Virol ; 78(7): 3502-13, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15016873

RESUMEN

The hepatitis C virus NS5A protein plays a critical role in virus replication, conferring interferon resistance to the virus through perturbation of multiple intracellular signaling pathways. Since NS5A is a phosphoprotein, it is of considerable interest to understand the role of phosphorylation in NS5A function. In this report, we investigated the phosphorylation of NS5A by taking advantage of 119 glutathione S-transferase-tagged protein kinases purified from Saccharomyces cerevisiae to perform a global screening of yeast kinases capable of phosphorylating NS5A in vitro. A database BLAST search was subsequently performed by using the sequences of the yeast kinases that phosphorylated NS5A in order to identify human kinases with the highest sequence homologies. Subsequent in vitro kinase assays and phosphopeptide mapping studies confirmed that several of the homologous human protein kinases were capable of phosphorylating NS5A. In vivo phosphopeptide mapping revealed phosphopeptides common to those generated in vitro by AKT, p70S6K, MEK1, and MKK6, suggesting that these kinases may phosphorylate NS5A in mammalian cells. Significantly, rapamycin, an inhibitor commonly used to investigate the mTOR/p70S6K pathway, reduced the in vivo phosphorylation of specific NS5A phosphopeptides, strongly suggesting that p70S6 kinase and potentially related members of this group phosphorylate NS5A inside the cell. Curiously, certain of these kinases also play a major role in mRNA translation and antiapoptotic pathways, some of which are already known to be regulated by NS5A. The findings presented here demonstrate the use of high-throughput screening of the yeast kinome to facilitate the major task of identifying human NS5A protein kinases for further characterization of phosphorylation events in vivo. Our results suggest that this novel approach may be generally applicable to the screening of other protein biochemical activities by mechanistic class.


Asunto(s)
Hepacivirus/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Homología de Secuencia , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Sistema Libre de Células , Biología Computacional , Humanos , Mapeo Peptídico , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Sirolimus/farmacología , Especificidad por Sustrato , Serina-Treonina Quinasas TOR
19.
EMBO Rep ; 4(2): 159-65, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12612605

RESUMEN

Bacteria adapt their pattern of gene expression in response to a variety of external cues, including fluctuations in population density. This type of bacterial cell-to-cell communication is referred to as quorum-sensing. Quorum-sensing systems are present in many bacterial species and constitute a large collection of ligands and cognate receptors. The availability of such diversity offers interesting opportunities for biotechnological exploitation. We describe here the transformation of the quorum-sensing system of Agrobacterium tumefaciens into a transcription regulatory system that works in mammalian cells. The A. tumefaciens TraR protein was fused to the eukaryotic activation domain of NF-kappaB p65, generating a novel chimaeric transcriptional activator that stimulates gene transcription in different human cell lines from a minimal promoter containing the TraR DNA recognition sequence in the presence of the Agrobacterium quorum-sensing signal molecule N-(3-oxo-octanoyl)homoserine lactone (3-oxo-C(8)-HSL). The basal level of transcription was low in the absence of 3-oxo-C(8)-HSL, and gene expression was stimulated up to 1,000-fold at a saturating concentration of 3-oxo-C(8)-HSL.


Asunto(s)
Proteínas Bacterianas/fisiología , ADN/fisiología , Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transcripción Genética/fisiología
20.
Anal Biochem ; 309(1): 11-8, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12381356

RESUMEN

An electrospray mass spectrometry-based methodology has been developed to have a fast and sensitive method for protein-cofactor stoichiometry determination. As model systems, we used two proteins which require the presence of cofactors for activity: TraR, a member of the LuxR family of quorum-sensing transcriptional regulators, which requires an acyl-homoserine lactone molecule called Agrobacterium autoinducer (AAI) as coinducer and the NS3 protease of hepatitis C virus which complexes with a NS4A cofactor peptide. Both TraR/AAI and NS3/NS4A are noncovalent complexes. Our method requires only nanomolar concentration of sample. A calibration curve of the cofactor is determined by high-performance liquid chromatography (HPLC) coupled on-line with an ion trap mass spectrometer operated in selected reaction monitoring mode. Subsequently, the complex is analyzed using the same experimental setup. During the HPLC run, the complex dissociates, and cofactor and protein elute at different retention times. The peak area of the cofactor is integrated and the molar concentration of cofactor in the complex is extrapolated from the calibration curve. The stoichiometry is consequently calculated by dividing the molar concentration of protein injected by that of cofactor measured. Both TraR/AAI and NS3/NS4A complexes have 1:1 stoichiometries, in line with those already reported in the literature.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Hepacivirus/enzimología , Espectrometría de Masa por Ionización de Electrospray/métodos , Agrobacterium tumefaciens/química , Agrobacterium tumefaciens/aislamiento & purificación , Secuencia de Aminoácidos , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/aislamiento & purificación , Calibración , Proteínas Portadoras/química , Proteínas Portadoras/aislamiento & purificación , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/aislamiento & purificación , Homoserina/análogos & derivados , Homoserina/aislamiento & purificación , Interacciones Hidrofóbicas e Hidrofílicas , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/aislamiento & purificación , Proteínas Virales/química , Proteínas Virales/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...