Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochemistry ; 59(28): 2608-2615, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32578982

RESUMEN

The virus that causes COVID-19, SARS-CoV-2, has a large RNA genome that encodes numerous proteins that might be targets for antiviral drugs. Some of these proteins, such as the RNA-dependent RNA polymerase, helicase, and main protease, are well conserved between SARS-CoV-2 and the original SARS virus, but several others are not. This study examines one of the proteins encoded by SARS-CoV-2 that is most different, a macrodomain of nonstructural protein 3 (nsp3). Although 26% of the amino acids in this SARS-CoV-2 macrodomain differ from those observed in other coronaviruses, biochemical and structural data reveal that the protein retains the ability to bind ADP-ribose, which is an important characteristic of beta coronaviruses and a potential therapeutic target.


Asunto(s)
Betacoronavirus/química , Proteínas no Estructurales Virales/química , Adenosina Difosfato Ribosa/metabolismo , COVID-19 , Coronavirus/química , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Proteasas Similares a la Papaína de Coronavirus , Cristalografía por Rayos X , Sistemas de Liberación de Medicamentos , Humanos , Modelos Moleculares , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Dominios Proteicos , SARS-CoV-2 , Termodinámica , Proteínas no Estructurales Virales/metabolismo
2.
Anal Biochem ; 595: 113622, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32059949

RESUMEN

Nudix proteins are members of a large family of homologous enzymes that hydrolyze nucleoside diphosphates linked to other compounds. The substrates for a subset of Nudix enzymes are all nucleotides linked to RNA, like the m7G mRNA caps and the more recently discovered NAD(H) RNA caps. However, the RNA affinity and nucleic acid specificity of Nudix proteins has not yet been explored in depth. In this study we designed new fluorescence-based assays to examine the interaction of purified recombinant E. coli NudC and human Nudt1 (aka MTH1) Nudt3, Nudt12, Nudt16, and Nudt20 (aka Dcp2). All Nudix proteins except Nudt1 and Nudt12 bound both RNA and DNA stoichiometrically with high affinity (dissociation constants in the nanomolar range) and no clear sequence specificity. In stark contrast, Nudt12 binds RNA but not similar DNA oligonucleotides. Nudt12 also bound RNAs with 5' NAD+ caps more tightly than those with NADH or m7G cap. NudC was similarly selective against m7G caps but did not differentiate between NAD+ and NADH capped RNA. Nudt3, Nudt16, and Nudt20 bound m7G capped RNA more tightly than RNA with NADH caps.


Asunto(s)
Enzimas Reparadoras del ADN/análisis , ADN/química , Colorantes Fluorescentes/química , Monoéster Fosfórico Hidrolasas/análisis , Pirofosfatasas/análisis , ARN/química , Sitios de Unión , Escherichia coli/enzimología , Humanos , Proteínas Recombinantes/análisis , Hidrolasas Nudix
3.
Adv Exp Med Biol ; 1111: 219-240, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30471070

RESUMEN

This review discusses new developments in Förster resonance energy transfer (FRET) microscopy and its application to cellular receptors. The method is based on the kinetic theory of FRET, which can be used to predict FRET not only in dimers, but also higher order oligomers of donor and acceptor fluorophores. Models based on such FRET predictions can be fit to observed FRET efficiency histograms (also called FRET spectrograms) and used to estimate intracellular binding constants, free energy values, and stoichiometries. These "FRET spectrometry" methods have been used to analyze oligomers formed by various receptors in cell signaling pathways, but until recently such studies were limited to receptors residing on the cell surface. To study complexes residing inside the cell, a technique called Quantitative Micro-Spectroscopic Imaging (Q-MSI) was developed. Q-MSI combines determination of quaternary structure from pixel-level apparent FRET spectrograms with the determination of both donor and acceptor concentrations at the organelle level. This is done by resolving and analyzing the spectrum of a third fluorescent marker, which does not participate in FRET. Q-MSI was first used to study the interaction of a class of cytoplasmic receptors that bind viral RNA and signal an antiviral response via complexes formed mainly on mitochondrial membranes. Q-MSI revealed previously unknown RNA mitochondrial receptor orientations, and the interaction between the viral RNA receptor called LGP2 with the RNA helicase encoded by the hepatitis virus. The biological importance of these new observations is discussed.


Asunto(s)
Supervivencia Celular , Proteína 58 DEAD Box/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Transducción de Señal
4.
Biochemistry ; 57(43): 6247-6255, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30281972

RESUMEN

DECH-box proteins are a subset of DExH/D-box superfamily 2 helicases possessing a conserved Asp-Glu-Cys-His motif in their ATP binding site. The conserved His helps position the Asp and Glu residues, which coordinate the divalent metal cation that connects the protein to ATP and activate the water molecule needed for ATP hydrolysis, but the role of the Cys is still unclear. This study uses site-directed mutants of the model DECH-box helicase encoded by the hepatitis C virus (HCV) to examine the role of the Cys in helicase action. Proteins lacking a Cys unwound DNA less efficiently than wild-type proteins did. For example, at low protein concentrations, a helicase harboring a Gly instead of the DECH-box Cys unwound DNA more slowly than the wild-type helicase did, but at higher protein concentrations, the two proteins unwound DNA at similar rates. All HCV proteins analyzed had similar affinities for ATP and nucleic acids and hydrolyzed ATP in the presence of RNA at similar rates. However, in the absence of RNA, all proteins lacking a DECH-box cysteine hydrolyzed ATP 10-15 times faster with higher Km values, and lower apparent affinities for metal ions, compared to those observed with wild-type proteins. These differences were observed with proteins isolated from HCV genotypes 2a and 1b, suggesting that this role is conserved. These data suggest the helicase needs Cys292 to bind ATP in a state where ATP is not hydrolyzed until RNA binds.


Asunto(s)
Adenosina Trifosfato/metabolismo , Cisteína/química , ADN/metabolismo , Hepacivirus/enzimología , ARN/química , ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Sitios de Unión , Catálisis , Cisteína/genética , Cisteína/metabolismo , ADN/química , Humanos , Hidrólisis , Mutagénesis Sitio-Dirigida , Mutación , Especificidad por Sustrato , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
5.
J Biol Chem ; 292(27): 11165-11177, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28483922

RESUMEN

Human cells detect RNA viruses through a set of helicases called RIG-I-like receptors (RLRs) that initiate the interferon response via a mitochondrial signaling complex. Many RNA viruses also encode helicases, which are sometimes covalently linked to proteases that cleave signaling proteins. One unresolved question is how RLRs interact with each other and with viral proteins in cells. This study examined the interactions among the hepatitis C virus (HCV) helicase and RLR helicases in live cells with quantitative microspectroscopic imaging (Q-MSI), a technique that determines FRET efficiency and subcellular donor and acceptor concentrations. HEK293T cells were transfected with various vector combinations to express cyan fluorescent protein (CFP) or YFP fused to either biologically active HCV helicase or one RLR (i.e. RIG-I, MDA5, or LGP2), expressed in the presence or absence of polyinosinic-polycytidylic acid (poly(I:C)), which elicits RLR accumulation at mitochondria. Q-MSI confirmed previously reported RLR interactions and revealed an interaction between HCV helicase and LGP2. Mitochondria in CFP-RIG-I:YFP-RIG-I cells, CFP-MDA5:YFP-MDA5 cells, and CFP-MDA5:YFP-LGP2 cells had higher FRET efficiencies in the presence of poly(I:C), indicating that RNA causes these proteins to accumulate at mitochondria in higher-order complexes than those formed in the absence of poly(I:C). However, mitochondria in CFP-LGP2:YFP-LGP2 cells had lower FRET signal in the presence of poly(I:C), suggesting that LGP2 oligomers disperse so that LGP2 can bind MDA5. Data support a new model where an LGP2-MDA5 oligomer shuttles NS3 to the mitochondria to block antiviral signaling.


Asunto(s)
Hepacivirus/enzimología , Helicasa Inducida por Interferón IFIH1/metabolismo , Mitocondrias/enzimología , Modelos Biológicos , ARN Helicasas/metabolismo , Transducción de Señal , Proteínas no Estructurales Virales/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hepacivirus/genética , Humanos , Helicasa Inducida por Interferón IFIH1/genética , Microscopía Fluorescente/métodos , Mitocondrias/genética , Poli I-C/farmacología , ARN Helicasas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas no Estructurales Virales/genética
6.
Bioorg Med Chem Lett ; 27(4): 936-940, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28089348

RESUMEN

A ligand-based approach was applied to screen in silico a library of commercially available compounds, with the aim to find novel inhibitors of the HCV replication starting from the study of the viral NS3 helicase. Six structures were selected for evaluation in the HCV subgenomic replicon assay and one hit was found to inhibit the HCV replicon replication in the low micromolar range. A small series of new pyrrolone compounds was designed and synthesised, and novel structures were identified with improved antiviral activity.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Pirroles/farmacología , Antivirales/química , Evaluación Preclínica de Medicamentos , Hepacivirus/fisiología , Pirroles/química , Replicación Viral/efectos de los fármacos
7.
Transpl Infect Dis ; 23(5): e13738, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34590768
8.
J Biol Chem ; 288(27): 19949-57, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23703611

RESUMEN

ATP hydrolysis fuels the ability of helicases and related proteins to translocate on nucleic acids and separate base pairs. As a consequence, nucleic acid binding stimulates the rate at which a helicase catalyzes ATP hydrolysis. In this study, we searched a library of small molecule helicase inhibitors for compounds that stimulate ATP hydrolysis catalyzed by the hepatitis C virus (HCV) NS3 helicase, which is an important antiviral drug target. Two compounds were found that stimulate HCV helicase-catalyzed ATP hydrolysis, both of which are amide derivatives synthesized from the main component of the yellow dye primuline. Both compounds possess a terminal pyridine moiety, which was critical for stimulation. Analogs lacking a terminal pyridine inhibited HCV helicase catalyzed ATP hydrolysis. Unlike other HCV helicase inhibitors, the stimulatory compounds differentiate between helicases isolated from various HCV genotypes and related viruses. The compounds only stimulated ATP hydrolysis catalyzed by NS3 purified from HCV genotype 1b. They inhibited helicases from other HCV genotypes (e.g. 1a and 2a) or related flaviviruses (e.g. Dengue virus). The stimulatory compounds interacted with HCV helicase in the absence of ATP with dissociation constants of about 2 µM. Molecular modeling and site-directed mutagenesis studies suggest that the stimulatory compounds bind in the HCV helicase RNA-binding cleft near key residues Arg-393, Glu-493, and Ser-231.


Asunto(s)
Adenosina Trifosfato/química , Hepacivirus/enzimología , Modelos Moleculares , ARN Helicasas/química , ARN Viral , Tiazoles/química , Proteínas Virales/química , Adenosina Trifosfato/metabolismo , Hepacivirus/genética , Hidrólisis , Mutagénesis Sitio-Dirigida , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
9.
Nucleic Acids Res ; 40(17): 8607-21, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22740655

RESUMEN

Typical assays used to discover and analyze small molecules that inhibit the hepatitis C virus (HCV) NS3 helicase yield few hits and are often confounded by compound interference. Oligonucleotide binding assays are examined here as an alternative. After comparing fluorescence polarization (FP), homogeneous time-resolved fluorescence (HTRF®; Cisbio) and AlphaScreen® (Perkin Elmer) assays, an FP-based assay was chosen to screen Sigma's Library of Pharmacologically Active Compounds (LOPAC) for compounds that inhibit NS3-DNA complex formation. Four LOPAC compounds inhibited the FP-based assay: aurintricarboxylic acid (ATA) (IC50=1.4 µM), suramin sodium salt (IC50=3.6 µM), NF 023 hydrate (IC50=6.2 µM) and tyrphostin AG 538 (IC50=3.6 µM). All but AG 538 inhibited helicase-catalyzed strand separation, and all but NF 023 inhibited replication of subgenomic HCV replicons. A counterscreen using Escherichia coli single-stranded DNA binding protein (SSB) revealed that none of the new HCV helicase inhibitors were specific for NS3h. However, when the SSB-based assay was used to analyze derivatives of another non-specific helicase inhibitor, the main component of the dye primuline, it revealed that some primuline derivatives (e.g. PubChem CID50930730) are up to 30-fold more specific for HCV NS3h than similarly potent HCV helicase inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hepacivirus/enzimología , Ensayos Analíticos de Alto Rendimiento , ARN Helicasas/antagonistas & inhibidores , Proteínas no Estructurales Virales/antagonistas & inhibidores , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Pruebas de Enzimas , Proteínas de Escherichia coli/metabolismo , Polarización de Fluorescencia , ARN Helicasas/metabolismo , Bibliotecas de Moléculas Pequeñas , Proteínas no Estructurales Virales/metabolismo
10.
Biochemistry ; 52(36): 6151-9, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23947785

RESUMEN

Aurintricarboxylic acid (ATA) is a potent inhibitor of many enzymes needed for cell and virus replication, such as polymerases, helicases, nucleases, and topoisomerases. This study examines how ATA interacts with the helicase encoded by the hepatitis C virus (HCV) and reveals that ATA interferes with both nucleic acid and ATP binding to the enzyme. We show that ATA directly binds HCV helicase to prevent the enzyme from interacting with nucleic acids and to modulate the affinity of HCV helicase for ATP, the fuel for helicase action. Amino acid substitutions in the helicase DNA binding cleft or its ATP binding site alter the ability of ATA to disrupt helicase-DNA interactions. These data, along with molecular modeling results, support the notion that an ATA polymer binds between Arg467 and Glu493 to prevent the helicase from binding either ATP or nucleic acids. We also characterize how ATA affects the kinetics of helicase-catalyzed ATP hydrolysis, and thermodynamic parameters describing the direct interaction between HCV helicase and ATA using microcalorimetry. The thermodynamics of ATA binding to HCV helicase reveal that ATA binding does not mimic nucleic acid binding in that ATA binding is driven by a smaller enthalpy change and an increase in entropy.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ácido Aurintricarboxílico/farmacología , ADN/metabolismo , Proteínas no Estructurales Virales/efectos de los fármacos , Sustitución de Aminoácidos , Rastreo Diferencial de Calorimetría , Hepacivirus/enzimología , Modelos Moleculares , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
11.
J Antimicrob Chemother ; 67(8): 1884-94, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22535622

RESUMEN

OBJECTIVES: Infection with yellow fever virus (YFV), the prototypic mosquito-borne flavivirus, causes severe febrile disease with haemorrhage, multi-organ failure and a high mortality. Moreover, in recent years the Flavivirus genus has gained further attention due to re-emergence and increasing incidence of West Nile, dengue and Japanese encephalitis viruses. Potent and safe antivirals are urgently needed. METHODS: Starting from the crystal structure of the NS3 helicase from Kunjin virus (an Australian variant of West Nile virus), we identified a novel, unexploited protein site that might be involved in the helicase catalytic cycle and could thus in principle be targeted for enzyme inhibition. In silico docking of a library of small molecules allowed us to identify a few selected compounds with high predicted affinity for the new site. Their activity against helicases from several flaviviruses was confirmed in in vitro helicase/enzymatic assays. The effect on the in vitro replication of flaviviruses was then evaluated. RESULTS: Ivermectin, a broadly used anti-helminthic drug, proved to be a highly potent inhibitor of YFV replication (EC50 values in the sub-nanomolar range). Moreover, ivermectin inhibited, although less efficiently, the replication of several other flaviviruses, i.e. dengue fever, Japanese encephalitis and tick-borne encephalitis viruses. Ivermectin exerts its effect at a timepoint that coincides with the onset of intracellular viral RNA synthesis, as expected for a molecule that specifically targets the viral helicase. CONCLUSIONS: The well-tolerated drug ivermectin may hold great potential for treatment of YFV infections. Furthermore, structure-based optimization may result in analogues exerting potent activity against flaviviruses other than YFV.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Ivermectina/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Virus de la Fiebre Amarilla/efectos de los fármacos , Animales , Chlorocebus aethiops , Virus del Dengue/efectos de los fármacos , Virus de la Encefalitis Japonesa (Subgrupo)/efectos de los fármacos , Virus de la Encefalitis Transmitidos por Garrapatas/efectos de los fármacos , Simulación de Dinámica Molecular , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/química , Serina Endopeptidasas/química , Células Vero , Proteínas no Estructurales Virales/química , Replicación Viral/efectos de los fármacos
12.
J Infect Dis ; 204(4): 609-16, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21791663

RESUMEN

BACKGROUND: Hepatitis C virus (HCV) chronically infects >170 million persons worldwide and is a leading cause of cirrhosis and hepatocellular carcinoma. The identification of more effective and better-tolerated agents for treating HCV is a high priority. We have reported elsewhere the discovery of the anti-HCV compound ceestatin using a high-throughput screen of a small molecule library. METHODS: To identify host or viral protein targets in an unbiased fashion, we performed affinity chromatography, using tandem liquid chromatography/mass spectrometry to identify specific potential targets. RESULTS. Ceestatin binds to 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) synthase and irreversibly inhibits HMG-CoA synthase in a dose-dependent manner. Ceestatin's anti-HCV effects are reversed by addition of HMG-CoA, mevalonic acid, or geranylgeraniol. Treatment with small interfering RNA against HMG-CoA synthase led to a substantial reduction in HCV replication, further validating HMG-CoA synthase as an enzyme essential for HCV replication. CONCLUSIONS: Ceestatin therefore exerts its anti-HCV effects through inhibition of HMG-CoA synthase. It may prove useful as an antiviral agent, as a probe to study HCV replication, and as a cholesterol-lowering agent. The logical stepwise process employed to discover the mechanism of action of ceestatin can serve as a general experimental strategy to uncover the targets on which novel uncharacterized anti-HCV compounds act.


Asunto(s)
Antivirales/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Hepacivirus/efectos de los fármacos , Hidroximetilglutaril-CoA Sintasa/antagonistas & inhibidores , Lactonas/farmacología , Replicación Viral/efectos de los fármacos , Línea Celular , Cromatografía de Afinidad , Hepacivirus/fisiología , Humanos , Espectrometría de Masas , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño
13.
J Med Chem ; 65(4): 3388-3403, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35133171

RESUMEN

Histone acetylation is a prominent epigenetic modification linked to the memory loss symptoms associated with neurodegenerative disease. The use of existing histone deacetylase inhibitor (HDACi) drugs for treatment is precluded by their weak blood-brain barrier (BBB) permeability and undesirable toxicity. Here, we address these shortcomings by developing a new class of disulfide-based compounds, inspired by the scaffold of the FDA-approved HDACi romidepsin (FK288). Our findings indicate that our novel compound MJM-1 increases the overall level of histone 3 (H3) acetylation in a prostate cancer cell line. In mice, MJM-1 injected intraperitoneally (i.p.) crossed the BBB and could be detected in the hippocampus, a brain region that mediates memory. Consistent with this finding, we found that the post-training i.p. administration of MJM-1 enhanced hippocampus-dependent spatial memory consolidation in male mice. Therefore, MJM-1 represents a potential lead for further optimization as a therapeutic strategy for ameliorating cognitive deficits in aging and neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Memoria Espacial/efectos de los fármacos , Animales , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/farmacología , Ratones , Ratones Endogámicos BALB C
14.
Nucleic Acids Res ; 37(2): 647-57, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074196

RESUMEN

Human DNA polymerase delta (Pol delta4), a key enzyme in chromosomal replication, is a heterotetramer composed of the p125, p50, p68 and p12 subunits. Genotoxic agents such as UV and alkylating chemicals trigger a DNA damage response in which Pol delta4 is converted to a trimer (Pol delta3) by degradation of p12. We show that Pol delta3 has altered enzymatic properties: it is less able to perform translesion synthesis on templates containing base lesions (O(6)-MeG, 8-oxoG, an abasic site or a thymine-thymine dimer); a greater proofreading activity; an increased exonuclease/polymerase activity ratio; a decreased tendency for the insertion of wrong nucleotides, and for the extension of mismatched primers. Overall, our findings indicate that Pol delta3 exhibits an enhanced ability for the detection of errors in both primers and templates over its parent enzyme. These alterations in Pol delta3 show that p12 plays a major role in Pol delta4 catalytic functions, and provides significant insights into the rationale for the conversion of Pol delta4 to Pol delta3 in the cellular response to DNA damage.


Asunto(s)
Disparidad de Par Base , Daño del ADN , ADN Polimerasa III/metabolismo , ADN/biosíntesis , ADN Polimerasa III/genética , Cartilla de ADN , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Células HeLa , Humanos , Mutación , Nucleótidos/metabolismo , Subunidades de Proteína/metabolismo , Dímeros de Pirimidina/química , Proteínas Recombinantes/metabolismo , Moldes Genéticos , Rayos Ultravioleta
15.
Biochemistry ; 49(9): 1822-32, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20108979

RESUMEN

This study examines the effects of 1-N,4-N-bis[4-(1H-benzimidazol-2-yl)phenyl]benzene-1,4-dicarboxamide ((BIP)(2)B) on the NS3 helicase encoded by the hepatitis C virus (HCV). Molecular beacon-based helicase assays were used to show that (BIP)(2)B inhibits the ability of HCV helicase to separate a variety of RNA and DNA duplexes with half-maximal inhibitory concentrations ranging from 0.7 to 5 microM, depending on the nature of the substrate. In single turnover assays, (BIP)(2)B only inhibited unwinding reactions when it was preincubated with the helicase-nucleic acid complex. (BIP)(2)B quenched NS3 intrinsic protein fluorescence with an apparent dissociation constant of 5 microM, and in the presence of (BIP)(2)B, HCV helicase did not appear to interact with a fluorescent DNA oligonucleotide. In assays monitoring HCV helicase-catalyzed ATP hydrolysis, (BIP)(2)B only inhibited helicase-catalyzed ATP hydrolysis in the presence of intermediate concentrations of RNA, suggesting RNA and (BIP)(2)B compete for the same binding site. HCV helicases isolated from various HCV genotypes were similarly sensitive to (BIP)(2)B, with half-maximal inhibitory concentrations ranging from 0.7 to 2.4 microM. (BIP)(2)B also inhibited ATP hydrolysis catalyzed by related helicases from Dengue virus, Japanese encephalitis virus, and humans. (BIP)(2)B appeared to bind the HCV and human proteins with similar affinity (K(i) = 7 and 8 microM, respectively), but it bound the flavivirus proteins up to 270 times more tightly. Results are discussed in light of a molecular model of a (BIP)(2)B-HCV helicase complex, which is unable to bind nucleic acid, thus preventing the enzyme from separating double-stranded nucleic acid.


Asunto(s)
Antivirales/química , Bencimidazoles/química , Hepacivirus/enzimología , ARN Helicasas/antagonistas & inhibidores , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antivirales/metabolismo , Antivirales/farmacología , Secuencia de Bases , Bencimidazoles/metabolismo , Bencimidazoles/farmacología , Catálisis , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , ARN Helicasas/química , ARN Helicasas/metabolismo , Especificidad por Sustrato , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo
16.
Biochemistry ; 49(17): 3545-54, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20334433

RESUMEN

This study examines the role of the p12 subunit in the function of the human DNA polymerase delta (Pol delta) holoenzyme by comparing the kinetics of DNA synthesis and degradation catalyzed by the four-subunit complex, the three-subunit complex lacking p12, and site-directed mutants of each lacking proofreading exonuclease activity. Results show that p12 modulates the rate and fidelity of DNA synthesis by Pol delta. All four complexes synthesize DNA in a rapid burst phase and a slower, more linear phase. In the presence of p12, the burst rates of DNA synthesis are approximately 5 times faster, while the affinity of the enzyme for its DNA and dNTP substrates appears unchanged. The p12 subunit alters Pol delta fidelity by modulating the proofreading 3' to 5' exonuclease activity. In the absence of p12, Pol delta is more likely to proofread DNA synthesis because it cleaves single-stranded DNA twice as fast and transfers mismatched DNA from the polymerase to the exonuclease sites 9 times faster. Pol delta also extends mismatched primers 3 times more slowly in the absence of p12. Taken together, the changes that p12 exerts on Pol delta are ones that can modulate its fidelity of DNA synthesis. The loss of p12, which occurs in cells upon exposure to DNA-damaging agents, converts Pol delta to a form that has an increased capacity for proofreading.


Asunto(s)
ADN Polimerasa III/química , ADN Polimerasa III/metabolismo , ADN/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Replicación del ADN , Exonucleasas/metabolismo , Humanos , Proteínas Nucleares/metabolismo
17.
Antimicrob Agents Chemother ; 54(8): 3187-96, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20516278

RESUMEN

The hepatitis C virus (HCV) NS5B RNA polymerase facilitates the RNA synthesis step during the HCV replication cycle. Nucleoside analogs targeting the NS5B provide an attractive approach to treating HCV infections because of their high barrier to resistance and pan-genotype activity. PSI-7851, a pronucleotide of beta-D-2'-deoxy-2'-fluoro-2'-C-methyluridine-5'-monophosphate, is a highly active nucleotide analog inhibitor of HCV for which a phase 1b multiple ascending dose study of genotype 1-infected individuals was recently completed (M. Rodriguez-Torres, E. Lawitz, S. Flach, J. M. Denning, E. Albanis, W. T. Symonds, and M. M. Berry, Abstr. 60th Annu. Meet. Am. Assoc. Study Liver Dis., abstr. LB17, 2009). The studies described here characterize the in vitro antiviral activity and cytotoxicity profile of PSI-7851. The 50% effective concentration for PSI-7851 against the genotype 1b replicon was determined to be 0.075+/-0.050 microM (mean+/-standard deviation). PSI-7851 was similarly effective against replicons derived from genotypes 1a, 1b, and 2a and the genotype 1a and 2a infectious virus systems. The active triphosphate, PSI-7409, inhibited recombinant NS5B polymerases from genotypes 1 to 4 with comparable 50% inhibitory concentrations. PSI-7851 is a specific HCV inhibitor, as it lacks antiviral activity against other closely related and unrelated viruses. PSI-7409 also lacked any significant activity against cellular DNA and RNA polymerases. No cytotoxicity, mitochondrial toxicity, or bone marrow toxicity was associated with PSI-7851 at the highest concentration tested (100 microM). Cross-resistance studies using replicon mutants conferring resistance to modified nucleoside analogs showed that PSI-7851 was less active against the S282T replicon mutant, whereas cells expressing a replicon containing the S96T/N142T mutation remained fully susceptible to PSI-7851. Clearance studies using replicon cells demonstrated that PSI-7851 was able to clear cells of HCV replicon RNA and prevent viral rebound.


Asunto(s)
Antivirales/farmacología , Nucleótidos de Desoxiuracil/farmacología , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Profármacos/farmacología , Replicación Viral/efectos de los fármacos , Amidas/química , Amidas/farmacología , Antivirales/química , Línea Celular Tumoral , Nucleótidos de Desoxiuracil/química , Inhibidores Enzimáticos/química , Genotipo , Hepacivirus/clasificación , Hepacivirus/enzimología , Humanos , Ácidos Fosfóricos/química , Ácidos Fosfóricos/farmacología , Profármacos/química , ARN Viral/genética , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Replicón/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores
18.
bioRxiv ; 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32676591

RESUMEN

Small molecules that bind the SARS-CoV-2 non-structural protein 3 Mac1 domain in place of ADP-ribose could be useful as molecular probes or scaffolds for COVID-19 antiviral drug discovery because Mac1 has been linked to coronavirus' ability to evade cellular detection. A high-throughput assay based on differential scanning fluorimetry (DSF) was therefore optimized and used to identify possible Mac1 ligands in small libraries of drugs and drug-like compounds. Numerous promising compounds included nucleotides, steroids, beta-lactams, and benzimidazoles. The main drawback to this approach was that a high percentage of compounds in some libraries were found to influence the observed Mac1 melting temperature. To prioritize DSF screening hits, the shapes of the observed melting curves and initial assay fluorescence were examined, and the results were compared with virtual screens performed using Autodock VINA. The molecular basis for alternate ligand binding was also examined by determining a structure of one of the hits, cyclic adenosine monophosphate, with atomic resolution.

19.
SLAS Discov ; 25(10): 1162-1170, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32981460

RESUMEN

Small molecules that bind the SARS-CoV-2 nonstructural protein 3 Mac1 domain in place of ADP-ribose could be useful as molecular probes or scaffolds for COVID-19 antiviral drug discovery because Mac1 has been linked to the ability of coronaviruses to evade cellular detection. A high-throughput assay based on differential scanning fluorimetry (DSF) was therefore optimized and used to identify possible Mac1 ligands in small libraries of drugs and drug-like compounds. Numerous promising compounds included nucleotides, steroids, ß-lactams, and benzimidazoles. The main drawback to this approach was that a high percentage of compounds in some libraries were found to influence the observed Mac1 melting temperature. To prioritize DSF screening hits, the shapes of the observed melting curves and initial assay fluorescence were examined, and the results were compared with virtual screens performed using AutoDock Vina. The molecular basis for alternate ligand binding was also examined by determining a structure of one of the hits, cyclic adenosine monophosphate, with atomic resolution.


Asunto(s)
Proteasas Similares a la Papaína de Coronavirus/química , Proteasas Similares a la Papaína de Coronavirus/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , SARS-CoV-2/química , Antivirales/química , Antivirales/farmacología , Sitios de Unión , Proteasas Similares a la Papaína de Coronavirus/genética , AMP Cíclico/química , AMP Cíclico/metabolismo , Ligandos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Conformación Proteica , Dominios Proteicos , SARS-CoV-2/efectos de los fármacos
20.
J Virol ; 82(7): 3438-51, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18216107

RESUMEN

Failure of the adaptive immune response to control infection with the hepatitis C virus (HCV) can result from mutational escape in targeted T-cell epitopes. Recent studies suggest that T-cell immune pressure is an important factor in the evolution of the nonstructural proteins in HCV. The aim of this study was to characterize the forces that contribute to viral evolution in an HLA-A*01-restricted epitope in HCV NS3. This epitope represents a potentially attractive target for vaccination strategies since it is conserved across all genotypes. In our cohort of subjects with chronic HCV infection (genotype 1b or 3a), it is a frequently recognized CD8 epitope in HLA-A*01-positive subjects. Viral sequence data reveal that an escape variant is the dominant residue in both genotypes. The predominant Y1444F substitution seemingly impairs binding to the HLA-A*01 molecule, which may have an important impact on the ability to prime a functional CD8 response upon infection. Interestingly, a case of evolution toward the prototype sequence was observed during chronic infection, possibly because the helicase activity of the protein containing the Y1444F substitution is reduced compared to the prototype sequence. Comparison of HCV sequences from Asia and Europe suggests that the frequency of the HLA-A*01 allele in a population may influence the frequency of the escape variant in circulating strains. These data suggest a complex interaction of multiple forces shaping the evolution of HCV in which immune pressure both within the individual and also at the population level in addition to functional constraints are important contributing factors.


Asunto(s)
Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Evolución Molecular , Hepacivirus/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Sustitución de Aminoácidos/genética , Asia , ADN Helicasas/metabolismo , Europa (Continente) , Femenino , Antígenos HLA-A/metabolismo , Antígeno HLA-A1 , Hepacivirus/genética , Hepacivirus/aislamiento & purificación , Hepatitis C Crónica/virología , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Filogenia , Unión Proteica , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia , Proteínas no Estructurales Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA