Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem J ; 481(6): 437-460, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38372302

RESUMEN

Catalytic poly(ADP-ribose) production by PARP1 is allosterically activated through interaction with DNA breaks, and PARP inhibitor compounds have the potential to influence PARP1 allostery in addition to preventing catalytic activity. Using the benzimidazole-4-carboxamide pharmacophore present in the first generation PARP1 inhibitor veliparib, a series of 11 derivatives was designed, synthesized, and evaluated as allosteric PARP1 inhibitors, with the premise that bulky substituents would engage the regulatory helical domain (HD) and thereby promote PARP1 retention on DNA breaks. We found that core scaffold modifications could indeed increase PARP1 affinity for DNA; however, the bulk of the modification alone was insufficient to trigger PARP1 allosteric retention on DNA breaks. Rather, compounds eliciting PARP1 retention on DNA breaks were found to be rigidly held in a position that interferes with a specific region of the HD domain, a region that is not targeted by current clinical PARP inhibitors. Collectively, these compounds highlight a unique way to trigger PARP1 retention on DNA breaks and open a path to unveil the pharmacological benefits of such inhibitors with novel properties.


Asunto(s)
Antineoplásicos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Bencimidazoles/farmacología , Reparación del ADN , Roturas del ADN , Daño del ADN
2.
Bioorg Med Chem Lett ; 88: 129288, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37094724

RESUMEN

NIMA Related Kinase 2 (Nek2) kinase is an attractive target for the development of therapeutic agents for several types of highly invasive cancers. Despite this, no small molecule inhibitor has advanced to the late clinical stages thus far. In this work, we have identified a novel spirocyclic inhibitor (V8) of Nek2 kinase, utilizing a high-throughput virtual screening (HTVS) approach. Using recombinant Nek2 enzyme assays, we show that V8 can inhibit Nek2 kinase activity (IC50 = 2.4 ± 0.2 µM) by binding to the enzyme's ATP pocket. The inhibition is selective, reversible and is not time dependent. To understand the key chemotype features responsible for Nek2 inhibition, a detailed structure-activity relationships (SAR) was performed. Using molecular models of the energy-minimized structures of Nek2-inhibitory complexes, we identify key hydrogen-bonding interactions, including two from the hinge-binding region, likely responsible for the observed affinity. Finally, using cell-based studies, we show that V8 attenuates (a) pAkt/PI3 Kinase signaling in a dose-dependent manner, and (b) proliferative and migratory phenotypes of highly aggressive human MDA-MB-231 breast and A549 lung cancer cell lines. Thus, V8 is an important novel lead compound for the development of highly potent and selective Nek2 inhibitory agents.


Asunto(s)
Quinasas Relacionadas con NIMA , Humanos , Línea Celular Tumoral , Neoplasias Pulmonares , Modelos Moleculares , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Fosforilación , Relación Estructura-Actividad
3.
Molecules ; 27(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35056661

RESUMEN

Cell cycle kinases represent an important component of the cell machinery that controls signal transduction involved in cell proliferation, growth, and differentiation. Nek2 is a mitotic Ser/Thr kinase that localizes predominantly to centrosomes and kinetochores and orchestrates centrosome disjunction and faithful chromosomal segregation. Its activity is tightly regulated during the cell cycle with the help of other kinases and phosphatases and via proteasomal degradation. Increased levels of Nek2 kinase can promote centrosome amplification (CA), mitotic defects, chromosome instability (CIN), tumor growth, and cancer metastasis. While it remains a highly attractive target for the development of anti-cancer therapeutics, several new roles of the Nek2 enzyme have recently emerged: these include drug resistance, bone, ciliopathies, immune and kidney diseases, and parasitic diseases such as malaria. Therefore, Nek2 is at the interface of multiple cellular processes and can influence numerous cellular signaling networks. Herein, we provide a critical overview of Nek2 kinase biology and discuss the signaling roles it plays in both normal and diseased human physiology. While the majority of research efforts over the last two decades have focused on the roles of Nek2 kinase in tumor development and cancer metastasis, the signaling mechanisms involving the key players associated with several other notable human diseases are highlighted here. We summarize the efforts made so far to develop Nek2 inhibitory small molecules, illustrate their action modalities, and provide our opinion on the future of Nek2-targeted therapeutics. It is anticipated that the functional inhibition of Nek2 kinase will be a key strategy going forward in drug development, with applications across multiple human diseases.


Asunto(s)
Enfermedades Óseas/patología , Inhibidores Enzimáticos/farmacología , Enfermedades del Sistema Inmune/patología , Enfermedades Renales/patología , Malaria/patología , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Neoplasias/patología , Enfermedades Óseas/tratamiento farmacológico , Enfermedades Óseas/enzimología , Resistencia a Medicamentos , Humanos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Enfermedades del Sistema Inmune/enzimología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/enzimología , Malaria/tratamiento farmacológico , Malaria/enzimología , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología
4.
Mol Med ; 27(1): 79, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34271850

RESUMEN

BACKGROUND: High mobility group box 1 protein (HMGB1) is an alarmin following its release by immune cells upon cellular activation or stress. High levels of extracellular HMGB1 play a critical role in impairing the clearance of invading pulmonary pathogens and dying neutrophils in the injured lungs of cystic fibrosis (CF) and acute respiratory distress syndrome (ARDS). A heparin derivative, 2-O, 3-O desulfated heparin (ODSH), has been shown to inhibit HMGB1 release from a macrophage cell line and is efficacious in increasing bacterial clearance in a mouse model of pneumonia. Thus, we hypothesized that ODSH can attenuate the bacterial burden and inflammatory lung injury in CF and we conducted experiments to determine the underlying mechanisms. METHODS: We determined the effects of ODSH on lung injury produced by Pseudomonas aeruginosa (PA) infection in CF mice with the transmembrane conductance regulator gene knockout (CFTR-/-). Mice were given ODSH or normal saline intraperitoneally, followed by the determination of the bacterial load and lung injury in the airways and lung tissues. ODSH binding to HMGB1 was determined using surface plasmon resonance and in silico docking analysis of the interaction of the pentasaccharide form of ODSH with HMGB1. RESULTS: CF mice given 25 mg/kg i.p. of ODSH had significantly lower PA-induced lung injury compared to mice given vehicle alone. The CF mice infected with PA had decreased levels of nitric oxide (NO), increased levels of airway HMGB1 and HMGB1-impaired macrophage phagocytic function. ODSH partially attenuated the PA-induced alteration in the levels of NO and airway HMGB1 in CF mice. In addition, ODSH reversed HMGB1-impaired macrophage phagocytic function. These effects of ODSH subsequently decreased the bacterial burden in the CF lungs. In a surface plasmon resonance assay, ODSH interacted with HMGB1 with high affinity (KD = 3.89 × 10-8 M) and induced conformational changes that may decrease HMGB1's binding to its membrane receptors, thus attenuating HMGB1-induced macrophage dysfunction. CONCLUSIONS: The results suggest that ODSH can significantly decrease bacterial infection-induced lung injury in CF mice by decreasing both HMGB1-mediated impairment of macrophage function and the interaction of HMGB1 with membrane receptors. Thus, ODSH could represent a novel approach for treating CF and ARDS patients that have HMGB1-mediated lung injury.


Asunto(s)
Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Proteína HMGB1/genética , Heparina/análogos & derivados , Macrófagos/inmunología , Macrófagos/metabolismo , Neumonía Bacteriana/etiología , Neumonía Bacteriana/metabolismo , Animales , Carga Bacteriana , Biomarcadores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Proteína HMGB1/química , Proteína HMGB1/metabolismo , Heparina/química , Heparina/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Óxido Nítrico/metabolismo , Fagocitosis/inmunología , Neumonía Bacteriana/patología , Unión Proteica , Células RAW 264.7 , Relación Estructura-Actividad
5.
Bioorg Chem ; 102: 104075, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32777641

RESUMEN

Poly(ADP-ribose) polymerase 1 (PARP1), a widely explored anticancer drug target, plays an important role in single-strand DNA break repair processes. High-throughput virtual screening (HTVS) of a Maybridge small molecule library using the PARP1-benzimidazole-4-carboxamide co-crystal structure and pharmacophore model led to the identification of eleven compounds. These compounds were evaluated using recombinant PARP1 enzyme assay that resulted in the acquisition of three PARP1 inhibitors: 3 (IC50 = 12 µM), 4 (IC50 = 5.8 µM), and 10 (IC50 = 0.88 µM). Compound 4 (2,3-dihydro-1,4-benzodioxine-5-carboxamide) was selected as a lead and was subjected to further chemical modifications, involving analogue synthesis and scaffold hopping. These efforts led to the identification of (Z)-2-(4-hydroxybenzylidene)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazine-8-carboxamide (49, IC50 = 0.082 µM) as the most potent inhibitor of PARP1 from the series.


Asunto(s)
Dioxinas/síntesis química , Dioxinas/uso terapéutico , Ensayos Analíticos de Alto Rendimiento/métodos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/síntesis química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Dioxinas/farmacología , Humanos , Simulación del Acoplamiento Molecular , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Relación Estructura-Actividad
6.
Virus Genes ; 53(4): 522-531, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28425034

RESUMEN

The molluscum contagiosum virus (MCV) uses a variety of immune evasion strategies to antagonize host immune responses. Two MCV proteins, MC159 and MC160, contain tandem death effector domains (DEDs). They are reported to inhibit innate immune signaling events such as NF-κB and IRF3 activation, and apoptosis. The RxDL motif of MC159 is required for inhibition of both apoptosis and NF-κB activation. However, the role of the conserved RxDL motif in the MC160 DEDs remained unknown. To answer this question, we performed alanine mutations to neutralize the arginine and aspartate residues present in the MC160 RxDL in both DED1 and DED2. These mutations were further modeled against the structure of the MC159 protein. Surprisingly, the RxDL motif was not required for MC160's ability to inhibit MAVS-induced IFNß activation. Further, unlike previous results with the MC159 protein, mutations within the RxDL motif of MC160 had no effect on the ability of MC160 to dampen TNF-α-induced NF-κB activation. Molecular modeling predictions revealed no overall changes to the structure in the MC160 protein when the amino acids of both RxDL motifs were mutated to alanine (DED1 = R67A D69A; DED2 = R160A D162A). Taken together, our results demonstrate that the RxDL motifs present in the MC160 DEDs are not required for known functions of the viral protein.


Asunto(s)
Evasión Inmune , Molusco Contagioso/virología , Virus del Molusco Contagioso/inmunología , Proteínas Virales/química , Proteínas Virales/inmunología , Secuencias de Aminoácidos , Apoptosis , Humanos , Interferón beta/genética , Interferón beta/inmunología , Molusco Contagioso/genética , Molusco Contagioso/inmunología , Molusco Contagioso/fisiopatología , Virus del Molusco Contagioso/química , Virus del Molusco Contagioso/genética , Dominios Proteicos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Virales/genética
8.
Biochem Biophys Res Commun ; 469(3): 743-7, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26697747

RESUMEN

Dengue virus (DENV) infection is a significant health threat to the global population with no therapeutic option. DENV NS5 RNA-dependent RNA polymerase (RdRp) is the key replicating protein of the virus and thus an attractive target for drug development. Herein, we report on the synthesis and biological evaluation of a series of hybrid thiazolidinone-thiadiazole derivatives as a new class of DENV-2 NS5 RdRp inhibitors. This yielded compounds 12 and 21 with IC50 values of 2.3 µM and 2.1 µM, respectively, as promising leads. Limited SAR analysis indicated 3-fluorobenzylidene as the optimal substituent at C5-position of the thiazolidinone core, whereas both 2-chlorophenyl and 3-fluorophenyl substituents were equally effective at C5-position of the 1,3,4-thiadiazole core. Biophysical characterization and molecular docking studies conferred the binding site of this scaffold on DENV NS5 polymerase. Binding mode of compound 21 in Thumb pocket-II of DENV-2 NS5 polymerase will form the basis for future structure-activity relationship optimization.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/ultraestructura , Tiadiazoles/química , Tiazolidinedionas/química , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/ultraestructura , Sitios de Unión , Combinación de Medicamentos , Descubrimiento de Drogas , Modelos Químicos , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Tiadiazoles/administración & dosificación , Tiazolidinedionas/administración & dosificación
9.
Pharm Res ; 33(6): 1456-71, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26951566

RESUMEN

PURPOSE: The purpose of this study was to develop an amorphous solid dispersion (SD) of an extremely water-insoluble and very weakly basic drug, itraconazole (ITZ), by interaction with weak organic acids and then drying that would enhance dissolution rate of drug and physical stability of formulation. METHODS: Aqueous solubility of ITZ in concentrated solutions of weak organic acids, such as glutaric, tartaric, malic and citric acid, was determined. Solutions with high drug solubility were dried using vacuum oven and the resulting SDs having 2 to 20% drug load were characterized by differential scanning calorimetry (DSC), powder X-ray diffractometry (PXRD) and attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy. The dissolution of SDs was initially studied in 250 mL of 0.1 N HCl (pH 1.1), and any undissolved solids were collected and analyzed by PXRD. The pH of the dissolution medium was then changed from 1.1 to 5.5, particle size of precipitates were measured, and drug concentrations in solution were determined by filtration through membrane filters of varying pore sizes. RESULTS: The aqueous solubility of ITZ was greatly enhanced in presence of weak acids. While the solubility of ITZ in water was ~4 ng/ mL, it increased to 25-40 mg per g of solution at 25°C and 200 mg per g of solution at 65°C at a high acid concentration leading to extremely high solubilization. PXRD of SDs indicated that ITZ was present in the amorphous form, wherein the acid formed a partially crystalline matrix. ATR-FTIR results showed possible weak interactions, such as hydrogen bonding, between drug and acid but there was no salt formation. SDs formed highly supersaturated solutions at pH 1.1 and had superior dissolution rate as compared to amorphous drug and physical mixtures of drug and acids. Following the change in pH from 1.1 to 5.5, ITZ precipitated as mostly nanoparticles, providing high surface area for relatively rapid redissolution. CONCLUSIONS: A method of highly solubilizing an extremely water-insoluble drug, ITZ, in aqueous media and converting it into an amorphous form in a physically stable SD was successfully investigated. The dissolution rate and the extent of supersaturation of the drug in dissolution media improved greatly, and any precipitate formed at high pH had very small particle size.


Asunto(s)
Antifúngicos/química , Ácido Cítrico/química , Desecación , Itraconazol/química , Tecnología Farmacéutica/métodos , Rastreo Diferencial de Calorimetría , Cristalización , Cristalografía por Rayos X , Ácidos Dicarboxílicos/química , Composición de Medicamentos , Estabilidad de Medicamentos , Glutaratos/química , Ácido Clorhídrico/química , Concentración de Iones de Hidrógeno , Cinética , Malatos/química , Tamaño de la Partícula , Difracción de Polvo , Solubilidad , Espectroscopía Infrarroja por Transformada de Fourier , Tartratos/química
10.
Bioorg Med Chem ; 23(18): 6125-37, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26320664

RESUMEN

Methicillin resistant Staphylococcus aureus (MRSA) is a major drug resistant bacteria that persists in both community and clinical settings due to growing resistance to current drug regimens. Thus, there is a continued need for novel compounds that are active against this organism. Previously, we reported that various rhodanine derivatives inhibited the supercoiling activity of DNA gyrase. In this study, we determined the effect of new phenylalanine-derived (Z)-5-arylmethylidene rhodanines (which are efficacious against MRSA) on the activity of the two type II bacterial topoisomerases, DNA gyrase and topoisomerase IV (Topo IV). Compounds 1 and 9 showed the greatest efficacy against DNA gyrase with a minimal inhibitory concentration (MIC) of 5 µM while compounds 2 and 3 were the most efficacious against Topo IV with MIC values of 0.75 µM and 0.5 µM, respectively. Induced fit docking, using the crystallographic structures of the target enzymes, indicated that these rhodanine derivatives bind to the ATPase domain of gyrB and ParE subunits on DNA gyrase and Topo IV, respectively. These new compounds were efficacious against both DNA gyrase and Topo IV. The increased efficacy of these new rhodanine compounds, as compared to other rhodanine derivatives, results from their dual inhibition of DNA gyrase and Topo IV, thereby making them good candidates for further drug design and development.


Asunto(s)
Antibacterianos/química , Girasa de ADN/química , Topoisomerasa de ADN IV/antagonistas & inhibidores , Rodanina/química , Staphylococcus aureus/enzimología , Inhibidores de Topoisomerasa II/química , Secuencia de Aminoácidos , Antibacterianos/farmacología , Sitios de Unión , Dominio Catalítico , Girasa de ADN/metabolismo , Topoisomerasa de ADN IV/metabolismo , Diseño de Fármacos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Fenilalanina/química , Rodanina/farmacología , Alineación de Secuencia , Staphylococcus aureus/efectos de los fármacos , Inhibidores de Topoisomerasa II/farmacología
11.
J Enzyme Inhib Med Chem ; 30(5): 778-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25676325

RESUMEN

Arachidonic acid is an unsaturated fatty acid liberated from phospholipids of cell membranes. NSAIDs are known as targets of cyclooxygenase enzyme (COX-1, COX-2 and COX-3) in arachidonic acid metabolism. This mechanism of COX-2 in carcinogenesis causes cancer. In addition, COX-2 plays a role in the early stages of hepatocarcinogenesis. Hepatitis C virus (HCV) infection is cause of liver cirrhosis and hepatocellular carcinoma (HCC). The aim of our study was to improve effective agents against HCV. A novel series of new etodolac 1,2,4-triazoles derivatives (4a-h) have been synthesized and investigated for their activity against HCV NS5B polymerase. Compound 4a was found to be the most active with IC(50) value of 14.8 µM. In accordance with these results, compound 4a was screened for anti-cancer activity on liver cancer cell lines (Huh7, Mahlavu, HepG2, FOCUS). Compound 4a showed anti-cancer activity against Huh7 human hepatoma cell line with IC(50) value of 4.29 µM. Therefore, compound 4a could be considered as a new anti-cancer and anti-HCV lead compound.


Asunto(s)
Antineoplásicos/farmacología , Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Etodolaco/análogos & derivados , Hepacivirus/efectos de los fármacos , Triazoles/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Antivirales/síntesis química , Antivirales/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Etodolaco/síntesis química , Etodolaco/química , Etodolaco/farmacología , Hepacivirus/enzimología , Humanos , Estructura Molecular , Relación Estructura-Actividad , Triazoles/síntesis química , Triazoles/química , Proteínas no Estructurales Virales/metabolismo
12.
Arch Pharm (Weinheim) ; 348(1): 10-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25449674

RESUMEN

In continuation of our efforts to develop new derivatives as hepatitis C virus (HCV) NS5B inhibitors, we synthesized novel 5-arylidene-4-thiazolidinones. The novel compounds 29-42, together with their synthetic precursors 22-28, were tested for HCV NS5B inhibitory activity; 12 of these compounds displayed IC50 values between 25.3 and 54.1 µM. Compound 33, an arylidene derivative, was found to be the most active compound in this series with an IC50 value of 25.3 µM. Molecular docking studies were performed on the thumb pocket-II of NS5B to postulate the binding mode for these compounds.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Tiazolidinas/síntesis química , Tiazolidinas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antivirales/metabolismo , Sitios de Unión , Línea Celular Tumoral , Diseño Asistido por Computadora , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Hepacivirus/enzimología , Ensayos Analíticos de Alto Rendimiento , Humanos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Molecular , Conformación Proteica , Relación Estructura-Actividad , Tiazolidinas/metabolismo , Proteínas no Estructurales Virales/metabolismo
13.
J Cell Biochem ; 115(8): 1381-91, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24939447

RESUMEN

ARRY-334543 is a small molecule inhibitor of ErbB1 and ErbB2 tyrosine kinases. We conducted this study to determine whether ARRY-334543 can enhance the efficacy of conventional anticancer drugs through interaction with ABC transporters. Lung cancer cell line NCI-H460 and its ABCG2-overexpressing NCI-H460/MX20, as well as the ABCG2-, ABCB1-, and ABCC10-overexpressing transfected cell lines were used for the reversal study. Our results demonstrated that ARRY-334543 (1.0 µM) significantly reversed ABCG2-mediated multidrug resistance (MDR) by directly inhibiting the drug efflux function of ABCG2, resulting in the elevated intracellular accumulation of chemotherapeutic drugs in the ABCG2-overexpressing cell lines. In addition, in isolated membranes, ARRY-334543 stimulated ATPase activity and inhibited photolabeling of ABCG2 with [(125)I]-iodoarylazidoprazosin in a concentration-dependent manner indicating that this drug directly interacts at the drug-binding pocket of this transporter. ARRY-334543 (1.0 µM) only slightly reversed ABCB1- and partially reversed ABCC10-mediated MDR suggesting that it exhibits high affinity toward ABCG2. Moreover, homology modeling predicted the binding conformation of ARRY-334543 at Arg482 centroid-based grid of ABCG2. However, ARRY-334543 at reversal concentrations did not affect the expression level of ABCG2, AKT and ERK1/2 and regulate the re-localization of ABCG2. We conclude that ARRY-334543 significantly reverses drug resistance mediated by ABCG2.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinazolinas/administración & dosificación , Tiazoles/administración & dosificación , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/química , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Proteínas de Neoplasias/química , Proteínas Oncogénicas v-erbB/antagonistas & inhibidores , Proteínas Oncogénicas v-erbB/genética , Paclitaxel/administración & dosificación , Unión Proteica , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética
14.
Cancer Sci ; 105(8): 1071-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24903205

RESUMEN

ATP-binding cassette (ABC) transmembrane proteins evidently decrease the intracellular accumulation of substrate chemotherapeutic drugs by extruding them against a concentration gradient, thereby inducing drug resistance. Here we reported the effect of WHI-P154, an irreversible inhibitor of Janus kinase 3 and epidermal growth factor receptor tyrosine kinases, on reversing ABC transporters-mediated drug resistance. We found that WHI-P154 significantly enhanced the sensitivity of ABCG2-overexpressing cells to its substrates. WHI-P154 moderately sensitized ABCB1-overexpressing KB-C2 cells to its substrates whereas showed no sensitizing effect on ABCC1-, ABCC2 or ABCC10-mediated drug resistance. Moreover, WHI-P154 produced a significant increase in the intracellular accumulation of [³H]-mitoxantrone in ABCG2-overexpressing cells. The expression levels nor the localization of the ABCG2 protein was altered after treatment of ABCG2-overexpressing cells with WHI-P154. Further studies indicated that WHI-P154 enhanced the ATPase activity of ABCG2 at low concentrations (<10 µM). Additionally, a docking model predicted the binding conformation of WHI-P154 within the transmembrane region of homology-modeled human ABCG2 transporter. Collectively, these findings highlighted WHI-P154 could significantly reverse ABCG2-mediated multidrug drug resistance by directly blocking the efflux function.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Quinazolinas/farmacología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Western Blotting , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos
15.
Chembiochem ; 15(1): 157-69, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24288265

RESUMEN

Multidrug resistance caused by ATP binding cassette transporter P-glycoprotein (P-gp) through extrusion of anticancer drugs from the cells is a major cause of failure in cancer chemotherapy. Previously, selenazole-containing cyclic peptides were reported as P-gp inhibitors and were also used for co-crystallization with mouse P-gp, which has 87 % homology to human P-gp. It has been reported that human P-gp can simultaneously accommodate two to three moderately sized molecules at the drug binding pocket. Our in silico analysis, based on the homology model of human P-gp, spurred our efforts to investigate the optimal size of (S)-valine-derived thiazole units that can be accommodated at the drug binding pocket. Towards this goal, we synthesized varying lengths of linear and cyclic derivatives of (S)-valine-derived thiazole units to investigate the optimal size, lipophilicity, and structural form (linear or cyclic) of valine-derived thiazole peptides that can be accommodated in the P-gp binding pocket and affects its activity, previously an unexplored concept. Among these oligomers, lipophilic linear (13) and cyclic trimer (17) derivatives of QZ59S-SSS were found to be the most and equally potent inhibitors of human P-gp (IC50 =1.5 µM). As the cyclic trimer and linear trimer compounds are equipotent, future studies should focus on noncyclic counterparts of cyclic peptides maintaining linear trimer length. A binding model of the linear trimer 13 within the drug binding site on the homology model of human P-gp represents an opportunity for future optimization, specifically replacing valine and thiazole groups in the noncyclic form.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Diseño de Fármacos , Oligopéptidos/síntesis química , Péptidos Cíclicos/química , Tiazoles/química , Valina/análogos & derivados , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Sitios de Unión , Transporte Biológico/efectos de los fármacos , Compuestos de Boro/química , Células HeLa , Humanos , Ratones , Simulación del Acoplamiento Molecular , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Péptidos Cíclicos/metabolismo , Péptidos Cíclicos/farmacología , Peptidomiméticos , Fotoblanqueo/efectos de los fármacos , Unión Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Tiazoles/metabolismo , Valina/química , Valina/metabolismo
16.
Mol Pharm ; 11(7): 2313-22, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-24865254

RESUMEN

Nilotinib (Tasigna) is a tyrosine kinase inhibitor approved by the FDA to treat chronic phase chronic myeloid leukemia patients. It is also a transport substrate of the ATP-binding cassette (ABC) drug efflux transporters ABCB1 (P-glycoprotein, P-gp) and ABCG2 (BCRP), which may have an effect on the pharmacokinetics and toxicity of this drug. The goal of this study was to identify pharmacophoric features of nilotinib in order to potentially develop specific inhibitors of BCR-ABL kinase with minimal interactions with ABC drug transporters. Three-dimensional pharmacophore modeling and quantitative structure-activity relationship (QSAR) studies were carried out on a series of nilotinib analogues to identify chemical features that contribute to inhibitory activity of nilotinib against BCR-ABL kinase activity, P-gp, and ABCG2. Twenty-five derivatives of nilotinib were synthesized and were then tested to measure their activity to inhibit BCR-ABL kinase and to inhibit the function of ABC drug transporters. A set of in vitro experiments including kinase activity and cell-based transport assays and photolabeling of P-gp and ABCG2 with a transport substrate, [(125)I]-iodoarylazido-prazosin (IAAP), were carried out in isolated membranes to evaluate the potency of the derivatives to inhibit the function of ABC drug transporters and BCR-ABL kinase. Sixteen, fourteen, and ten compounds were selected as QSAR data sets, respectively, to generate PHASE v3.1 pharmacophore models for BCR-ABL kinase, ABCG2, and P-gp inhibitors. The IC50 values of these derivatives against P-gp, ABCG2, or BCR-ABL kinase were used to generate pharmacophore features required for optimal interactions with these targets. A seven-point pharmacophore (AADDRRR) for BCR-ABL kinase inhibitory activity, a six-point pharmacophore (ADHRRR) for ABCG2 inhibitory activity, and a seven-point pharmacophore (AADDRRR) for P-gp inhibitory activity were generated. The derived models clearly demonstrate high predictive power for test sets of BCR-ABL, ABCG2, and P-gp inhibitors. In aggregate, these results should aid in the development of specific inhibitors of BCR-ABL kinase that exhibit no or minimal interaction with ABC drug transporters.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/química , Pirimidinas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Células MCF-7 , Relación Estructura-Actividad Cuantitativa
17.
Mar Drugs ; 12(7): 3818-37, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24979269

RESUMEN

ATP binding cassette (ABC) transporters, such as P-gp, BCRP and MRP1, can increase efflux of clinical chemotherapeutic agents and lead to multi-drug resistance (MDR) in cancer cells. While the discovery and development of clinically useful inhibitors has proved elusive to date, this molecular target nevertheless remains a promising strategy for addressing and potentially overcoming MDR. In a search for new classes of inhibitor, we used fluorescent accumulation and efflux assays supported by cell flow cytometry and MDR reversal assays, against a panel of sensitive and MDR human cancer cell lines, to evaluate the marine sponge co-metabolites 1-12 as inhibitors of P-gp, BCRP or MRP1 initiated MDR. These studies identified and characterized lamellarin O (11) as a selective inhibitor of BCRP mediated drug efflux. A structure-activity relationship analysis inclusive of the natural products 1-12 and the synthetic analogues 13-19, supported by in silico docking studies, revealed key structural requirements for the lamellarin O (11) BCRP inhibitory pharmacophore.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Cumarinas/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Isoquinolinas/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Poríferos/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Australia , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos , Fluoresceínas/metabolismo , Humanos , Mitoxantrona/metabolismo , Simulación del Acoplamiento Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Proteínas de Neoplasias/fisiología , Relación Estructura-Actividad
18.
bioRxiv ; 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38826291

RESUMEN

PARP1 and PARP2 recognize DNA breaks immediately upon their formation, generate a burst of local PARylation to signal their location, and are co-targeted by all current FDA-approved forms of PARP inhibitors (PARPi) used in the cancer clinic. Recent evidence indicates that the same PARPi molecules impact PARP2 differently from PARP1, raising the possibility that allosteric activation may also differ. We find that unlike for PARP1, destabilization of the autoinhibitory domain of PARP2 is insufficient for DNA damage-induced catalytic activation. Rather, PARP2 activation requires further unfolding of an active site α-helix absent in PARP1. Only one clinical PARPi, Olaparib, stabilizes the PARP2 active site α-helix, representing a structural feature with the potential to discriminate small molecule inhibitors. Collectively, our findings reveal unanticipated differences in local structure and changes in activation-coupled backbone dynamics between PARP1 and PARP2.

19.
Int J Cancer ; 132(1): 224-35, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22623106

RESUMEN

Saracatinib, a highly selective, dual Src/Abl kinase inhibitor, is currently in a Phase II clinical trial for the treatment of ovarian cancer. In our study, we investigated the effect of saracatinib on the reversal of multidrug resistance (MDR) induced by ATP-binding cassette (ABC) transporters in vitro and in vivo. Our results showed that saracatinib significantly enhanced the cytotoxicity of ABCB1 substrate drugs in ABCB1 overexpressing HeLa/v200, MCF-7/adr and HEK293/ABCB1 cells, an effect that was stronger than that of gefitinib, whereas it had no effect on the cytotoxicity of the substrates in ABCC1 overexpressing HL-60/adr cells and its parental sensitive cells. Additionally, saracatinib significantly increased the doxorubicin (Dox) and Rho 123 accumulation in HeLa/v200 and MCF-7/adr cells, whereas it had no effect on HeLa and MCF-7 cells. Furthermore, saracatinib stimulated the ATPase activity and inhibited photolabeling of ABCB1 with [(125)I]-iodoarylazidoprazosin in a concentration-dependent manner. In addition, the homology modeling predicted the binding conformation of saracatinib within the large hydrophobic drug-binding cavity of human ABCB1. However, neither the expression level of ABCB1 nor the phosphorylation level of Akt was altered at the reversal concentrations of saracatinib. Importantly, saracatinib significantly enhanced the effect of paclitaxel against ABCB1-overexpressing HeLa/v200 cancer cell xenografts in nude mice. In conclusion, saracatinib reverses ABCB1-mediated MDR in vitro and in vivo by directly inhibiting ABCB1 transport function, without altering ABCB1 expression or AKT phosphorylation. These findings may be helpful to attenuate the effect of MDR by combining saracatinib with other chemotherapeutic drugs in the clinic.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Benzodioxoles/farmacología , Quinazolinas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP , Adenosina Trifosfatasas/metabolismo , Animales , Línea Celular Tumoral , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Gefitinib , Células HEK293 , Células HL-60 , Células HeLa , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Paclitaxel/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinasas Asociadas a rho/metabolismo
20.
Bioorg Med Chem Lett ; 23(20): 5523-7, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24012180

RESUMEN

A focused library of rhodanine compounds containing novel substituents at the C5-position was synthesized and tested in vitro against a panel of clinically relevant MRSA strains. The present SAR study was based on our lead compound 1 (MIC=1.95 µg/mL), with a focus on identifying optimal C5-arylidene substituents. In order to obtain this objective, we condensed several unique aromatic aldehydes with phenylalanine-derived rhodanine intermediates to obtain C5-substituted target rhodanine compounds for evaluation as anti-MRSA compounds. These efforts produced three compounds with significant efficacy: 23, 32 and 44, with MIC values ranging from 0.98 to 1.95 µg/mL against all tested MRSA strains as compared to the reference antibiotics penicillin G (MIC=15.60-250.0 µg/mL) and ciprofloxacin (MIC=7.80-62.50 µg/mL) and comparable to that of vancomycin (MIC=0.48 µg/mL). In addition, compounds 24, 28, 37, 41, 46 and 48 (MIC=1.95-3.90 µg/mL) were efficacious against all MRSA strains. The majority of the synthesized compounds had bactericidal activity at concentrations only two to fourfold higher than their MIC. Overall, the results suggest that compounds 23, 32 and 44 may be of potential use in the treatment of MRSA infections.


Asunto(s)
Antibacterianos/síntesis química , Fenilalanina/química , Rodanina/química , Antibacterianos/química , Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Rodanina/síntesis química , Rodanina/farmacología , Relación Estructura-Actividad , Vancomicina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA