Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(26): 7148-53, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27307435

RESUMEN

Trehalose is a disaccharide essential for the survival and virulence of pathogenic fungi. The biosynthesis of trehalose requires trehalose-6-phosphate synthase, Tps1, and trehalose-6-phosphate phosphatase, Tps2. Here, we report the structures of the N-terminal domain of Tps2 (Tps2NTD) from Candida albicans, a transition-state complex of the Tps2 C-terminal trehalose-6-phosphate phosphatase domain (Tps2PD) bound to BeF3 and trehalose, and catalytically dead Tps2PD(D24N) from Cryptococcus neoformans bound to trehalose-6-phosphate (T6P). The Tps2NTD closely resembles the structure of Tps1 but lacks any catalytic activity. The Tps2PD-BeF3-trehalose and Tps2PD(D24N)-T6P complex structures reveal a "closed" conformation that is effected by extensive interactions between each trehalose hydroxyl group and residues of the cap and core domains of the protein, thereby providing exquisite substrate specificity. Disruption of any of the direct substrate-protein residue interactions leads to significant or complete loss of phosphatase activity. Notably, the Tps2PD-BeF3-trehalose complex structure captures an aspartyl-BeF3 covalent adduct, which closely mimics the proposed aspartyl-phosphate intermediate of the phosphatase catalytic cycle. Structures of substrate-free Tps2PD reveal an "open" conformation whereby the cap and core domains separate and visualize the striking conformational changes effected by substrate binding and product release and the role of two hinge regions centered at approximately residues 102-103 and 184-188. Significantly, tps2Δ, tps2NTDΔ, and tps2D705N strains are unable to grow at elevated temperatures. Combined, these studies provide a deeper understanding of the substrate recognition and catalytic mechanism of Tps2 and provide a structural basis for the future design of novel antifungal compounds against a target found in three major fungal pathogens.


Asunto(s)
Candida albicans/enzimología , Cryptococcus neoformans/enzimología , Proteínas Fúngicas/química , Monoéster Fosfórico Hidrolasas/química , Biocatálisis , Candida albicans/química , Candida albicans/genética , Candida albicans/metabolismo , Cryptococcus neoformans/química , Cryptococcus neoformans/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Especificidad por Sustrato , Fosfatos de Azúcar/química , Fosfatos de Azúcar/metabolismo , Trehalosa/análogos & derivados , Trehalosa/química , Trehalosa/metabolismo
2.
Proc Natl Acad Sci U S A ; 113(13): 3669-74, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26976575

RESUMEN

The influenza endonuclease is an essential subdomain of the viral RNA polymerase. It processes host pre-mRNAs to serve as primers for viral mRNA and is an attractive target for antiinfluenza drug discovery. Compound L-742,001 is a prototypical endonuclease inhibitor, and we found that repeated passaging of influenza virus in the presence of this drug did not lead to the development of resistant mutant strains. Reduced sensitivity to L-742,001 could only be induced by creating point mutations via a random mutagenesis strategy. These mutations mapped to the endonuclease active site where they can directly impact inhibitor binding. Engineered viruses containing the mutations showed resistance to L-742,001 both in vitro and in vivo, with only a modest reduction in fitness. Introduction of the mutations into a second virus also increased its resistance to the inhibitor. Using the isolated wild-type and mutant endonuclease domains, we used kinetics, inhibitor binding and crystallography to characterize how the two most significant mutations elicit resistance to L-742,001. These studies lay the foundation for the development of a new class of influenza therapeutics with reduced potential for the development of clinical endonuclease inhibitor-resistant influenza strains.


Asunto(s)
Endonucleasas/antagonistas & inhibidores , Endonucleasas/genética , Inhibidores Enzimáticos/farmacología , Hidroxibutiratos/farmacología , Virus de la Influenza A/efectos de los fármacos , Piperidinas/farmacología , Animales , Dominio Catalítico/genética , Cristalografía por Rayos X , Perros , Farmacorresistencia Viral/genética , Endonucleasas/metabolismo , Variación Genética , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H1N1 del Virus de la Influenza A/genética , Virus de la Influenza A/enzimología , Virus de la Influenza A/genética , Cinética , Células de Riñón Canino Madin Darby , Modelos Moleculares , Mutagénesis , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo
3.
Bioorg Med Chem ; 26(1): 25-36, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29170024

RESUMEN

Within the last decade, the Bromodomain and Extra-Terminal domain family (BET) of proteins have emerged as promising drug targets in diverse clinical indications including oncology, auto-immune disease, heart failure, and male contraception. The BET family consists of four isoforms (BRD2, BRD3, BRD4, and BRDT/BRDT6) which are distinguished by the presence of two tandem bromodomains (BD1 and BD2) that independently recognize acetylated-lysine (KAc) residues and appear to have distinct biological roles. BET BD1 and BD2 bromodomains differ at five positions near the substrate binding pocket: the variation in the ZA channel induces different water networks nearby. We designed a set of congeneric 2- and 3-heteroaryl substituted tetrahydroquinolines (THQ) to differentially engage bound waters in the ZA channel with the goal of achieving bromodomain selectivity. SJ830599 (9) showed modest, but consistent, selectivity for BRD2-BD2. Using isothermal titration calorimetry, we showed that the binding of all THQ analogs in our study to either of the two bromodomains was enthalpy driven. Remarkably, the binding of 9 to BRD2-BD2 was marked by negative entropy and was entirely driven by enthalpy, consistent with significant restriction of conformational flexibility and/or engagement with bound waters. Co-crystallography studies confirmed that 9 did indeed stabilize a water-mediated hydrogen bond network. Finally, we report that 9 retained cytotoxicity against several pediatric cancer cell lines with EC50 values comparable to BET inhibitor (BETi) clinical candidates.


Asunto(s)
Proteínas/antagonistas & inhibidores , Quinolinas/farmacología , Termodinámica , Agua/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Proteínas/metabolismo , Quinolinas/síntesis química , Quinolinas/química , Relación Estructura-Actividad
4.
Bioorg Med Chem Lett ; 26(16): 3950-4, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27423480

RESUMEN

The sulfonamide class of antibiotics has been in continuous use for over 70years. They are thought to act by directly inhibiting dihydropteroate synthase (DHPS), and also acting as prodrugs that sequester pterin pools by forming dead end pterin-sulfonamide conjugates. In this study, eight pterin-sulfonamide conjugates were synthesized using a novel synthetic strategy and their biochemical and microbiological properties were investigated. The conjugates were shown to competitively inhibit DHPS, and inhibition was enhanced by the presence of pyrophosphate that is crucial to catalysis and is known to promote an ordering of the DHPS active site. The co-crystal structure of Yersinia pestis DHPS bound to one of the more potent conjugates revealed a mode of binding that is similar to that of the enzymatic product analog pteroic acid. The antimicrobial activities of the pterin-sulfonamide conjugates were measured against Escherichia coli in the presence and absence of folate precursors and dependent metabolites. These results show that the conjugates have appreciable antibacterial activity and act by an on target, anti-folate pathway mechanism rather than as simple dead end products.


Asunto(s)
Antibacterianos/química , Dihidropteroato Sintasa/antagonistas & inhibidores , Pterinas/química , Sulfonamidas/química , Antibacterianos/síntesis química , Antibacterianos/farmacología , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Dihidropteroato Sintasa/metabolismo , Escherichia coli/efectos de los fármacos , Ácido Fólico/química , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Yersinia pestis/enzimología
5.
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
6.
Anal Bioanal Chem ; 406(7): 1867-75, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24390461

RESUMEN

Herein, we describe the development of a fluorescence-based high throughput assay to determine the small molecule binding towards human serum albumin (HSA). This innovative competition assay is based on the use of a novel fluorescent small molecule Red Mega 500 with unique spectroscopic and binding properties. The commercially available probe displays a large fluorescence intensity difference between the protein-bound and protein-unbound state. The competition of small molecules for HSA binding in the presence of probe resulted in low fluorescence intensities. The assay was evaluated with the library of pharmacological active compounds (LOPAC) small molecule library of 1,280 compounds identifying known high protein binders. The small molecule competition of HSA-Red Mega 500 binding was saturable at higher compound concentrations and exhibited IC50 values between 3 and 24 µM. The compound affinity toward HSA was confirmed by isothermal titration calorimetry indicating that the new protein binding assay is a valid high throughput assay to determine plasma protein binding.


Asunto(s)
Colorantes Fluorescentes/química , Ensayos Analíticos de Alto Rendimiento , Preparaciones Farmacéuticas/metabolismo , Albúmina Sérica/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Unión Competitiva , Colorantes Fluorescentes/metabolismo , Humanos , Unión Proteica , Espectrometría de Fluorescencia
7.
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
8.
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
9.
Sci Rep ; 12(1): 8433, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589938

RESUMEN

CAP256V2LS, a broadly neutralizing monoclonal antibody (bNAb), is being pursued as a promising drug for HIV-1 prevention. The total level of tyrosine-O-sulfation, a post-translational modification, was known to play a key role for antibody biological activity. More importantly, here wedescribe for the first time the significance of the tyrosine-O-sulfation proteoforms. We developed a hydrophobic interaction chromatography (HIC) method to separate and quantify different sulfation proteoforms, which led to the direct functionality assessment of tyrosine-sulfated species. The fully sulfated (4-SO3) proteoform demonstrated the highest in vitro relative antigen binding potency and neutralization efficiency against a panel of HIV-1 viruses. Interestingly, highly variable levels of 4-SO3 were produced by different clonal CHO cell lines, which helped the bNAb process development towards production of a highly potent CAP256V2LS clinical product with high 4-SO3 proteoform. This study presents powerful insight for any biotherapeutic protein development where sulfation may play an important role in product efficacy.


Asunto(s)
VIH-1 , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes , Anticuerpos ampliamente neutralizantes , Células CHO , Cricetinae , Anticuerpos Anti-VIH , Tirosina/química
10.
J Biol Chem ; 285(23): 17292-300, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20360003

RESUMEN

RecD2 from Deinococcus radiodurans is a superfamily 1 DNA helicase that is homologous to the Escherichia coli RecD protein but functions outside the context of RecBCD enzyme. We report here on the kinetics of DNA unwinding by RecD2 under single and multiple turnover conditions. There is little unwinding of 20-bp substrates by preformed RecD2-dsDNA complexes when excess ssDNA is present to trap enzyme molecules not bound to the substrate. A shorter 12-bp substrate is unwound rapidly under single turnover conditions. The 12-bp unwinding reaction could be simulated with a mechanism in which the DNA is unwound in two kinetic steps with rate constant of k(unw) = 5.5 s(-1) and a dissociation step from partially unwound DNA of k(off) = 1.9 s(-1). These results indicate a kinetic step size of about 3-4 bp, unwinding rate of about 15-20 bp/s, and low processivity (p = 0.74). The reaction time courses with 20-bp substrates, determined under multiple turnover conditions, could be simulated with a four-step mechanism and rate constant values very similar to those for the 12-bp substrate. The results indicate that the faster unwinding of a DNA substrate with a forked end versus only a 5'-terminal single-stranded extension can be accounted for by a difference in the rate of enzyme binding to the DNA substrates. Analysis of reactions done with different RecD2 concentrations indicates that the enzyme forms an inactive dimer or other oligomer at high enzyme concentrations. RecD2 oligomers can be detected by glutaraldehyde cross-linking but not by size exclusion chromatography.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Helicasas/química , Deinococcus/metabolismo , Exodesoxirribonucleasa V/química , Animales , Cromatografía/métodos , Reactivos de Enlaces Cruzados/química , ADN/química , ADN Helicasas/metabolismo , ADN de Cadena Simple/química , Dimerización , Relación Dosis-Respuesta a Droga , Exodesoxirribonucleasa V/metabolismo , Glutaral/química , Cinética , Modelos Biológicos , Oligonucleótidos/química , Desnaturalización Proteica
11.
Cancer Res ; 80(17): 3507-3518, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32651255

RESUMEN

Inhibition of members of the bromodomain and extraterminal (BET) family of proteins has proven a valid strategy for cancer chemotherapy. All BET identified to date contain two bromodomains (BD; BD1 and BD2) that are necessary for recognition of acetylated lysine residues in the N-terminal regions of histones. Chemical matter that targets BET (BETi) also interact via these domains. Molecular and cellular data indicate that BD1 and BD2 have different biological roles depending upon their cellular context, with BD2 particularly associated with cancer. We have therefore pursued the development of BD2-selective molecules both as chemical probes and as potential leads for drug development. Here we report the structure-based generation of a novel series of tetrahydroquinoline analogs that exhibit >50-fold selectivity for BD2 versus BD1. This selective targeting resulted in engagement with BD-containing proteins in cells, resulting in modulation of MYC proteins and downstream targets. These compounds were potent cytotoxins toward numerous pediatric cancer cell lines and were minimally toxic to nontumorigenic cells. In addition, unlike the pan BETi (+)-JQ1, these BD2-selective inhibitors demonstrated no rebound expression effects. Finally, we report a pharmacokinetic-optimized, metabolically stable derivative that induced growth delay in a neuroblastoma xenograft model with minimal toxicity. We conclude that BD2-selective agents are valid candidates for antitumor drug design for pediatric malignancies driven by the MYC oncogene. SIGNIFICANCE: This study presents bromodomain-selective BET inhibitors that act as antitumor agents and demonstrates that these molecules have in vivo activity towards neuroblastoma, with essentially no toxicity.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Neoplasias , Factores de Transcripción/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Niño , Femenino , Humanos , Ratones , Ratones SCID , Neoplasias/genética , Neoplasias/metabolismo , Dominios Proteicos , Proteínas Proto-Oncogénicas c-myc/genética , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
12.
ACS Infect Dis ; 5(11): 1915-1925, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31588734

RESUMEN

Acyldepsipeptides are a unique class of antibiotics that act via allosterically dysregulated activation of the bacterial caseinolytic protease (ClpP). The ability of ClpP activators to kill nongrowing bacteria represents a new opportunity to combat deep-seated biofilm infections. However, the acyldepsipeptide scaffold is subject to rapid metabolism. Herein, we explore alteration of the potentially metabolically reactive α,ß unsaturated acyl chain. Through targeted synthesis, a new class of phenyl urea substituted depsipeptide ClpP activators with improved metabolic stability is described. The ureadepsipeptides are potent activators of Staphylococcus aureus ClpP and show activity against Gram-positive bacteria, including S. aureus biofilms. These studies demonstrate that a phenyl urea motif can successfully mimic the double bond, maintaining potency equivalent to acyldepsipeptides but with decreased metabolic liability. Although removal of the double bond from acyldepsipeptides generally has a significant negative impact on potency, structural studies revealed that the phenyl ureadepsipeptides can retain potency through the formation of a third hydrogen bond between the urea and the key Tyr63 residue in the ClpP activation domain. Ureadepsipeptides represent a new class of ClpP activators with improved drug-like properties, potent antibacterial activity, and the tractability to be further optimized.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Depsipéptidos/química , Endopeptidasa Clp/metabolismo , Activadores de Enzimas/química , Staphylococcus aureus/enzimología , Antibacterianos/química , Antibacterianos/metabolismo , Proteínas Bacterianas/agonistas , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Depsipéptidos/metabolismo , Depsipéptidos/farmacología , Endopeptidasa Clp/química , Endopeptidasa Clp/genética , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacología , Dominios Proteicos , Staphylococcus aureus/química , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Urea/química , Urea/metabolismo
13.
J Mol Graph Model ; 81: 197-210, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29605436

RESUMEN

The Bromodomain and Extra-Terminal domain (BET) family of proteins are involved in the regulation of gene transcription, and their dysregulation is implicated in several diseases including cancer. BET proteins contain two tandem bromodomains (BD1 and BD2) that independently recognize acetylated-lysine residues and appear to have distinct biological roles. We compared several published co-crystal structures and found five positions near the substrate binding pocket that vary between BET bromodomains. One position located in the ZA loop has unique properties. In BRD2-4, this residue is glutamine in BD1 and lysine in BD2; in BRDT, this residue is arginine in BD1 and asparagine in BD2. Using molecular modeling, we identified differences in the water-mediated network at this position between bromodomains. Molecular dynamics simulations helped rationalize the observed bromodomain selectivity for exemplar BET inhibitors and a congeneric series of tetrahydroquinolines (THQ) that differed by a single heteroatom near the ZA channel. The 2-furan SJ830599, the most BD2-selective THQ analog, did not disrupt the water-mediated networks in either domain, but was electrostatically-repulsed by the specific arrangement of the W5 water dipole in BD1. Our work underscores the value of exploring water-mediated interactions to study ligand binding, and highlights the difficulty of optimizing polar interactions due to high desolvation penalties. Finally, we suggest further modifications to THQ-based BET inhibitors that would increase BD2-selectivity in BRD2-4, while minimizing affinity for one or both bromodomains of BRDT.


Asunto(s)
Diseño de Fármacos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Secuencia de Aminoácidos , Sitios de Unión , Ligandos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas/antagonistas & inhibidores , Relación Estructura-Actividad Cuantitativa , Agua/química
14.
Eur J Med Chem ; 110: 126-32, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26820555

RESUMEN

Cortisol homeostasis has been linked to the pathogenesis of metabolic syndrome (MetS), since it stimulates hepatic gluconeogenesis and adipogenesis. MetS is classified as a constellation of health conditions that increase the risk of type 2 diabetes and cardiovascular disease. Intracellular cortisol levels are regulated by 11ß-hydroxysteroid dehydrogenase (type 1 and type 2) in a tissue dependent manner. The type 1 enzyme (11ß-HSD1) is widely expressed in glucocorticoid targeted tissues and is responsible for the conversion of cortisone to the active cortisol. Local reduction of cortisol regeneration presents a potential strategy for MetS treatment. Recently we disclosed the total synthesis of (+)-colletoic acid as a potent 11ß-HSD1 inhibitor. Herein, we describe our improved processing chemistry for the synthesis of the colletoic acid core to access a diverse number of derivatives for evaluation against 11ß-HSD1. The Evan's chiral auxiliary was utilized to construct the acyclic precursor 12 to afford the acorane core 9 using a modified Heck reaction in excellent chemical yields. The colletoic acid core derivatives showed modest activity against 11ß-HSD1 and will serve for further biological evaluation.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hidrocortisona/metabolismo , Sesquiterpenos/química , Sesquiterpenos/farmacología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Inhibidores Enzimáticos/síntesis química , Humanos , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/enzimología , Síndrome Metabólico/metabolismo , Modelos Moleculares , Sesquiterpenos/síntesis química
15.
J Biomol Screen ; 18(7): 761-81, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23536547

RESUMEN

Helicases are ubiquitous motor proteins that separate and/or rearrange nucleic acid duplexes in reactions fueled by adenosine triphosphate (ATP) hydrolysis. Helicases encoded by bacteria, viruses, and human cells are widely studied targets for new antiviral, antibiotic, and anticancer drugs. This review summarizes the biochemistry of frequently targeted helicases. These proteins include viral enzymes from herpes simplex virus, papillomaviruses, polyomaviruses, coronaviruses, the hepatitis C virus, and various flaviviruses. Bacterial targets examined include DnaB-like and RecBCD-like helicases. The human DEAD-box protein DDX3 is the cellular antiviral target discussed, and cellular anticancer drug targets discussed are the human RecQ-like helicases and eIF4A. We also review assays used for helicase inhibitor discovery and the most promising and common helicase inhibitor chemotypes, such as nucleotide analogues, polyphenyls, metal ion chelators, flavones, polycyclic aromatic polymers, coumarins, and various DNA binding pharmacophores. Also discussed are common complications encountered while searching for potent helicase inhibitors and possible solutions for these problems.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , ADN Helicasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Proteínas Virales/antagonistas & inhibidores , Animales , Antibacterianos/farmacología , Antineoplásicos/farmacología , Antivirales/farmacología , Proteínas Bacterianas/fisiología , ADN Helicasas/fisiología , Replicación del ADN , Ensayos Analíticos de Alto Rendimiento , Humanos , Unión Proteica , Biosíntesis de Proteínas , Proteínas Virales/fisiología
16.
J Mol Biol ; 425(12): 2083-99, 2013 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-23541589

RESUMEN

The endoplasmic reticulum transmembrane receptor Ire1 senses over-accumulation of unfolded proteins in the endoplasmic reticulum and initiates the unfolded protein response (UPR). The cytoplasmic portion of Ire1 has a protein kinase domain (KD) and a kinase extension nuclease (KEN) domain that cleaves an mRNA for encoding the Hac1 transcription factor needed to express UPR genes. During this UPR signaling, Ire1 proteins self-assemble into an oligomer of dimers, which essentially requires autophosphorylation of a constituent activation loop in the KD. However, it is not clear how dimerization, autophosphorylation, and KEN domain function are precisely coordinated. In this study, we uncoupled the KD and KEN domain functions, by removing the activation loop along with an extended region that we called the auto-inhibitory region (AIR), or by swapping the activation loop with a homologous loop from phosphorylase kinase 1 (Ire1(PHK)). Both Ire1(ΔAIR) and Ire1(PHK) activated the UPR even when either protein contained a mutation (D797A) that abolished the ability of Ire1 KD to transfer phosphates to the AIR. Neither protein functioned when containing mutations in key ATP binding residues (E746A and N749A) or in residues that disrupted Ire1 dimer interface (W426A or R697D). We interpret these results as evidence supporting the notion that the primary function of the kinase domain is to autophosphorylate the AIR in order to relieve auto-inhibition and that ADP acts as a switch to activate the KEN domain-catalyzed HAC1 mRNA cleavage.


Asunto(s)
Estrés del Retículo Endoplásmico , Glicoproteínas de Membrana/metabolismo , Fosforilasa Quinasa/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Glicoproteínas de Membrana/genética , Modelos Biológicos , Fosforilasa Quinasa/genética , Fosforilación , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
17.
Methods Enzymol ; 511: 463-83, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22713333

RESUMEN

This chapter describes two types of FRET-based fluorescence assays that can be used to identify and analyze compounds that inhibit the helicase encoded by the hepatitis C virus (HCV). Both assays use a fluorescently labeled DNA or RNA oligonucleotide to monitor helicase-catalyzed strand separation, and they differ from other real-time helicase assays in that they do not require the presence of other nucleic acids to trap the reaction products. The first assay is a molecular beacon-based helicase assay (MBHA) that monitors helicase-catalyzed displacement of a hairpin-forming oligonucleotide with a fluorescent moiety on one end and a quencher on the other. DNA-based MBHAs have been used extensively for high-throughput screening (HTS), but RNA-based MBHAs are typically less useful because of poor signal to background ratios. In the second assay discussed, the fluorophore and quencher are split between two hairpin-forming oligonucleotides annealed in tandem to a third oligonucleotide. This split beacon helicase assay can be used for HTS with either DNA or RNA oligonucleotides. These assays should be useful to the many labs searching for HCV helicase inhibitors in order to develop new HCV therapies that are still desperately needed.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Hepacivirus/enzimología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Adenosina Trifosfatasas/metabolismo , Transferencia Resonante de Energía de Fluorescencia
18.
Antiviral Res ; 96(2): 245-55, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22940425

RESUMEN

The hepatitis C virus (HCV) multifunctional nonstructural protein 3 (NS3) is a protease that cleaves viral and host proteins and a helicase that separates DNA and RNA structures in reactions fueled by ATP hydrolysis. Li et al. (2012) recently synthesized a series of new NS3 helicase inhibitors from the benzothiazole dimer component of the fluorescent yellow dye primuline. This study further characterizes a subset of these primuline derivatives with respect to their specificity, mechanism of action, and effect on cells harboring HCV subgenomic replicons. All compounds inhibited DNA and RNA unwinding catalyzed by NS3 from different HCV genotypes, but only some inhibited the NS3 protease function, and few had any effect on HCV NS3 catalyzed ATP hydrolysis. A different subset contained potent inhibitors of RNA stimulated ATP hydrolysis catalyzed by the related NS3 protein from Dengue virus. In assays monitoring intrinsic protein fluorescence in the absence of nucleic acids, the compounds cooperatively bound NS3 with K(d)s that reflect their potency in assays. The fluorescent properties of the primuline derivatives both in vitro and in cells are also described. The primuline derivative that was the most active against subgenomic replicons in cells caused a 14-fold drop in HCV RNA levels (IC(50)=5±2µM). In cells, the most effective primuline derivative did not inhibit the cellular activity of NS3 protease but disrupted HCV replicase structures.


Asunto(s)
Antivirales/farmacología , ADN Helicasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Serina Endopeptidasas/metabolismo , Tiazoles/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Línea Celular , Fluorescencia , Hepacivirus/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Unión Proteica , Tiazoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA