Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38064712

RESUMEN

Anticancer nucleosides are effective against solid tumors and hematological malignancies, but typically are prone to nucleoside metabolism resistance mechanisms. Using a nucleoside-specific multiplexed high-throughput screening approach, we discovered 4'-ethynyl-2'-deoxycytidine (EdC) as a third-generation anticancer nucleoside prodrug with preferential activity against diffuse large B-cell lymphoma (DLBCL) and acute lymphoblastic leukemia (ALL). EdC requires deoxycytidine kinase (DCK) phosphorylation for its activity and induced replication fork arrest and accumulation of cells in S-phase, indicating it acts as a chain terminator. A 2.1Å co-crystal structure of DCK bound to EdC and UDP reveals how the rigid 4'-alkyne of EdC fits within the active site of DCK. Remarkably, EdC was resistant to cytidine deamination and SAMHD1 metabolism mechanisms and exhibited higher potency against ALL compared to FDA approved nelarabine. Finally, EdC was highly effective against DLBCL tumors and B-ALL in vivo. These data characterize EdC as a pre-clinical nucleoside prodrug candidate for DLBCL and ALL.

2.
Nat Commun ; 14(1): 3079, 2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37248212

RESUMEN

Cancer cells utilize the main de novo pathway and the alternative salvage pathway for deoxyribonucleotide biosynthesis to achieve adequate nucleotide pools. Deoxycytidine kinase is the rate-limiting enzyme of the salvage pathway and it has recently emerged as a target for anti-proliferative therapies for cancers where it is essential. Here, we present the development of a potent inhibitor applying an iterative multidisciplinary approach, which relies on computational design coupled with experimental evaluations. This strategy allows an acceleration of the hit-to-lead process by gradually implementing key chemical modifications to increase affinity and activity. Our lead compound, OR0642, is more than 1000 times more potent than its initial parent compound, masitinib, previously identified from a drug repositioning approach. OR0642 in combination with a physiological inhibitor of the de novo pathway doubled the survival rate in a human T-cell acute lymphoblastic leukemia patient-derived xenograft mouse model, demonstrating the proof-of-concept of this drug design strategy.


Asunto(s)
Reposicionamiento de Medicamentos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Ratones , Humanos , Animales , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Nucleótidos , Diseño de Fármacos , Modelos Animales de Enfermedad
3.
J Med Chem ; 65(7): 5660-5674, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35348328

RESUMEN

Differentially screening the Fr-PPIChem chemical library on the bromodomain and extra-terminal (BET) BRD4-BDII versus -BDI bromodomains led to the discovery of a BDII-selective tetrahydropyridothienopyrimidinone (THPTP)-based compound. Structure-activity relationship (SAR) and hit-to-lead approaches allowed us to develop CRCM5484, a potent inhibitor of BET proteins with a preferential and 475-fold selectivity for the second bromodomain of the BRD3 protein (BRD3-BDII) over its first bromodomain (BRD3-BDI). Its very low activity was demonstrated in various cell-based assays, corresponding with recent data describing other selective BDII compounds. However, screening on a drug sensitivity and resistance-profiling platform revealed its ability to modulate the anti-leukemic activity in combination with various FDA-approved and/or in-development drugs in a cell- and context-dependent differential manner. Altogether, the results confirm the originality of the THPTP molecular mode of action in the bromodomain (BD) cavity and its potential as a starting scaffold for the development of potent and selective bromodomain inhibitors.


Asunto(s)
Proteínas Nucleares , Factores de Transcripción , Proteínas de Ciclo Celular , Dominios Proteicos , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
4.
J Chem Inf Model ; 59(4): 1472-1485, 2019 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-30908019

RESUMEN

We recently reported an integrated fragment-based optimization strategy called DOTS (Diversity Oriented Target-focused Synthesis) that combines automated virtual screening (VS) with semirobotized organic synthesis coupled to in vitro evaluation. The molecular modeling part consists of hit-to-lead chemistry, based on the growing paradigm. Here, we have extended the applicability of the DOTS strategy by adding new functionalities, allowing a generic chemistry-driven linking approach with a particular emphasis on covalent drugs. Indeed, the covalent mode of action can be described as a specific case of linking, where suitable linkers are sought to fuse a bound organic compound with a nucleophilic protein side chain. The proof of concept is established using three retrospective study cases in which known noncovalent inhibitors have been converted to covalent inhibitors. Our method is able to automatically design reference covalent inhibitors (and/or analogs) from an initial activated substructure and predict their binding mode. More importantly, the reference compounds are ranked high among several hundred putative adducts, demonstrating the utility of the approach to design covalent inhibitors.


Asunto(s)
Simulación por Computador , Diseño de Fármacos , Bibliotecas de Moléculas Pequeñas/química , Modelos Moleculares , Conformación Molecular , Bibliotecas de Moléculas Pequeñas/farmacología
5.
Antiviral Res ; 162: 79-89, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30557576

RESUMEN

Arenaviridae is a viral family whose members are associated with rodent-transmitted infections to humans responsible of severe diseases. The current lack of a vaccine and limited therapeutic options make the development of efficacious drugs of high priority. The cap-snatching mechanism of transcription of Arenavirus performed by the endonuclease domain of the L-protein is unique and essential, so we developed a drug design program targeting the endonuclease activity of the prototypic Lymphocytic ChorioMeningitis Virus. Since the endonuclease activity is metal ion dependent, we designed a library of compounds bearing chelating motifs (diketo acids, polyphenols, and N-hydroxyisoquinoline-1,3-diones) able to block the catalytic center through the chelation of the critical metal ions, resulting in a functional impairment. We pre-screened 59 compounds by Differential Scanning Fluorimetry. Then, we characterized the binding affinity by Microscale Thermophoresis and evaluated selected compounds in in vitro and in cellula assays. We found several potent binders and inhibitors of the endonuclease activity. This study validates the proof of concept that the endonuclease domain of Arenavirus can be used as a target for anti-arena-viral drug discovery and that both diketo acids and N-hydroxyisoquinoline-1,3-diones can be considered further as potential metal-chelating pharmacophores.


Asunto(s)
Quelantes/farmacología , Endonucleasas/antagonistas & inhibidores , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Virus de la Coriomeningitis Linfocítica/enzimología , Proteínas Virales/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Virus de la Coriomeningitis Linfocítica/fisiología , Polifenoles/farmacología , Bibliotecas de Moléculas Pequeñas , Replicación Viral/efectos de los fármacos
6.
IUCrJ ; 5(Pt 2): 223-235, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29765612

RESUMEN

The Arenaviridae family, together with the Bunyaviridae and Orthomyxoviridae families, is one of the three negative-stranded RNA viral families that encode an endonuclease in their genome. The endonuclease domain is at the N-terminus of the L protein, a multifunctional protein that includes the RNA-dependent RNA polymerase. The synthesis of mRNA in arenaviruses is a process that is primed by capped nucleotides that are 'stolen' from the cellular mRNA by the endonuclease domain in cooperation with other domains of the L protein. This molecular mechanism has been demonstrated previously for the endonuclease of the prototype Lymphocytic choriomeningitis virus (LCMV). However, the mode of action of this enzyme is not fully understood as the original structure did not contain catalytic metal ions. The pivotal role played by the cap-snatching process in the life cycle of the virus and the highly conserved nature of the endonuclease domain make it a target of choice for the development of novel antiviral therapies. Here, the binding affinities of two diketo-acid (DKA) compounds (DPBA and L-742,001) for the endonuclease domain of LCMV were evaluated using biophysical methods. X-ray structures of the LCMV endonuclease domain with catalytic ions in complex with these two compounds were determined, and their efficacies were assessed in an in vitro endonuclease-activity assay. Based on these data and computational simulation, two new DKAs were synthesized. The LCMV endonuclease domain exhibits a good affinity for these DKAs, making them a good starting point for the design of arenavirus endonuclease inhibitors. In addition to providing the first example of an X-ray structure of an arenavirus endonuclease incorporating a ligand, this study provides a proof of concept that the design of optimized inhibitors against the arenavirus endonuclease is possible.

7.
Nat Commun ; 8(1): 1420, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29127277

RESUMEN

Masitinib, a highly selective protein kinase inhibitor, can sensitise gemcitabine-refractory cancer cell lines when used in combination with gemcitabine. Here we report a reverse proteomic approach that identifies the target responsible for this sensitisation: the deoxycytidine kinase (dCK). Masitinib, as well as other protein kinase inhibitors, such as imatinib, interact with dCK and provoke an unforeseen conformational-dependent activation of this nucleoside kinase, modulating phosphorylation of nucleoside analogue drugs. This phenomenon leads to an increase of prodrug phosphorylation of most of the chemotherapeutic drugs activated by this nucleoside kinase. The unforeseen dual activity of protein kinase inhibition/nucleoside kinase activation could be of great therapeutic benefit, through either reducing toxicity of therapeutic agents by maintaining effectiveness at lower doses or by counteracting drug resistance initiated via down modulation of dCK target.


Asunto(s)
Desoxicitidina Quinasa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Tiazoles/farmacología , Células A549 , Antineoplásicos/química , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Cristalografía por Rayos X , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina Quinasa/química , Diseño de Fármacos , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Humanos , Mesilato de Imatinib/química , Mesilato de Imatinib/farmacología , Modelos Biológicos , Modelos Moleculares , Fosforilación , Piperidinas , Polifarmacología , Inhibidores de Proteínas Quinasas/química , Proteómica , Piridinas , Tiazoles/química , Gemcitabina
8.
Antiviral Res ; 117: 122-31, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25766862

RESUMEN

The acyclic nucleosides thiophosphonates (9-[2-(thiophosphonomethoxy)ethyl]adenine (S-PMEA) and (R)-9-[2-(thiophosphonomethoxy)propyl]adenine (S-PMPA), exhibit antiviral activity against HIV-1, -2 and HBV. Their diphosphate forms S-PMEApp and S-PMPApp, synthesized as stereoisomeric mixture, are potent inhibitors of wild-type (WT) HIV-1 RT. Understanding HIV-1 RT stereoselectivity, however, awaits resolution of the diphosphate forms into defined stereoisomers. To this aim, thiophosphonate monophosphates S-PMEAp and S-PMPAp were synthesized and used in a stereocontrolled enzyme-catalyzed phosphoryl transfer reaction involving either nucleoside diphosphate kinase (NDPK) or creatine kinase (CK) to obtain thiophosphonate diphosphates as separated isomers. We then quantified substrate preference of recombinant WT HIV-1 RT toward pure stereoisomers using in vitro steady-state kinetic analyses. The crystal structure of a complex between Dictyostelium NDPK and S-PMPApp at 2.32Å allowed to determine the absolute configuration at the α-phosphorus atom in relation to the stereo-preference of studied enzymes. The RP isomer of S-PMPApp and S-PMEApp are the preferred substrate over SP for both NDPK and HIV-1 RT.


Asunto(s)
Adenina/análogos & derivados , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/farmacología , Transcriptasa Inversa del VIH/antagonistas & inhibidores , VIH-1/efectos de los fármacos , Adenina/síntesis química , Adenina/química , Adenina/farmacología , Fármacos Anti-VIH/química , Cromatografía Líquida de Alta Presión , Creatina Quinasa/metabolismo , Cristalización , Transcriptasa Inversa del VIH/química , Transcriptasa Inversa del VIH/metabolismo , VIH-1/enzimología , VIH-2/efectos de los fármacos , Concentración 50 Inhibidora , Cinética , Conformación Molecular , Nucleósido-Difosfato Quinasa/metabolismo , Fósforo/química , Estereoisomerismo
9.
Neoplasia ; 15(7): 826-39, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23814494

RESUMEN

Human melanoma is a significant clinical problem. As most melanoma patients relapse with lethal drug-resistant disease, understanding and preventing mechanism(s) of resistance is one of the highest priorities to improve melanoma therapy. Melanosomal sequestration and the cellular exportation of cytotoxic drugs have been proposed to be important melanoma-specific mechanisms that contribute to multidrug resistance in melanoma. Concretely, we found that treatment of melanoma with methotrexate (MTX) altered melanogenesis and accelerated the exportation of melanosomes; however, the cellular and molecular processes by which MTX is trapped into melanosomes and exported out of cells have not been elucidated. In this study, we identified myosin Va (MyoVa) as a possible mediator of these cellular processes. The results demonstrated that melanoma treatment with MTX leads to Akt2-dependent MyoVa phosphorylation, which enhances its ability to interact with melanosomes and accelerates their exportation. To understand the mechanism(s) by which MTX activates Akt2, we examined the effects of this drug on the activity of protein phosphatase 2A, an Akt inhibitor activated by the methylation of its catalytic subunit. Taken together, this study identified a novel trafficking pathway in melanoma that promotes tumor resistance through Akt2/MyoVa activation. Because of these findings, we explored several MTX combination therapies to increase the susceptibility of melanoma to this drug. By avoiding MTX exportation, we observed that the E2F1 apoptotic pathway is functional in melanoma, and its induction activates p73 and apoptosis protease-activating factor 1 following a p53-autonomous proapoptotic signaling event.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Melanoma/metabolismo , Metotrexato/farmacología , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Silenciador del Gen , Humanos , Melanoma/genética , Metilación/efectos de los fármacos , Ratones , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2C , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Molecules ; 18(7): 8319-41, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23863773

RESUMEN

Novel tea catechin derivatives have been synthesized, and a structure-activity study, related to the capacity of these and other polyphenols to bind dihydrofolate reductase (DHFR), has been performed. The data showed an effective binding between all molecules and the free enzyme, and the dissociation constants of the synthetic compounds and of the natural analogues were on the same order. Polyphenols with a catechin configuration were better DHFR inhibitors than those with an epicatechin configuration. Antiproliferative activity was also studied in cultured tumour cells, and the data showed that the activity of the novel derivatives was higher in catechin isomers. Derivatives with a hydroxyl group para on the ester-bonded gallate moiety presented a high in vitro binding to DHFR, but exhibited transport problems in cell culture due to ionization at physiologic pHs. The impact of the binding of catechins to serum albumin on their biological activity was also evaluated. The information provided in this study could be important for the design of novel medicinal active compounds derived from tea catechins. The data suggest that changes in their structure to avoid serum albumin interactions and to facilitate plasmatic membrane transport are essential for the intracellular functions of catechins.


Asunto(s)
Catequina/química , Catequina/farmacología , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacología , Polifenoles/farmacología , Té/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Catequina/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Flavonoides/análisis , Flavonoides/química , Ácido Fólico/química , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/síntesis química , Humanos , Polifenoles/síntesis química , Polifenoles/química , Albúmina Sérica/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo
11.
Cancer Cell ; 24(1): 105-19, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23792190

RESUMEN

Therapeutic resistance in melanoma and other cancers arises via irreversible genetic, and dynamic phenotypic, heterogeneity. Here, we use directed phenotype switching in melanoma to sensitize melanoma cells to lineage-specific therapy. We show that methotrexate (MTX) induces microphthalmia-associated transcription factor (MITF) expression to inhibit invasiveness and promote differentiation-associated expression of the melanocyte-specific Tyrosinase gene. Consequently, MTX sensitizes melanomas to a tyrosinase-processed antifolate prodrug 3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin (TMECG), that inhibits the essential enzyme DHFR with high affinity. The combination of MTX and TMECG leads to depletion of thymidine pools, double-strand DNA breaks, and highly efficient E2F1-mediated apoptosis in culture and in vivo. Importantly, this drug combination delivers an effective and tissue-restricted antimelanoma therapy in vitro and in vivo irrespective of BRAF, MEK, or p53 status.


Asunto(s)
Melanoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Catequina/análogos & derivados , Catequina/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Factor de Transcripción E2F1/fisiología , Humanos , Melanoma/patología , Metotrexato/farmacología , Factor de Transcripción Asociado a Microftalmía/genética , Fenotipo , Nucleótidos de Timina/metabolismo
12.
PLoS One ; 7(12): e52231, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251702

RESUMEN

BACKGROUND: Tumour suppressor genes are often transcriptionally silenced by promoter hypermethylation, and recent research has implicated alterations in chromatin structure as the mechanistic basis for this repression. In addition to DNA methylation, other epigenetic post-translational modifications that modulate the stability and binding of specific transcription factors to gene promoters have emerged as important mechanisms for controlling gene expression. The aim of this study was to analyse the implications of these mechanisms and their molecular connections in the reactivation of RASSF1A in breast cancer. METHODS: Compounds that modulate the intracellular concentration of adenosine, such as dipyridamole (DIPY), greatly increase the antiproliferative effects of 3-O-(3,4,5-trimethoxybenzoyl)-(-)-catechin (TMCG), a synthetic antifolate derived from the structure of tea catechins. Quantitative real-time PCR arrays and MALDI-TOF mass spectrometry indicated that this combination (TMCG/DIPY) induced apoptosis in breast cancer cells by modulating the methylation levels of DNA and proteins (such as E2F1), respectively. Chromatin immunoprecipitation (ChIP) assays were employed to confirm that this combination induced chromatin remodelling of the RASSF1A promoter and increased the occupancy of E2F1 at the promoter of this tumour suppressor gene. RESULTS: The TMCG/DIPY combination acted as an epigenetic treatment that reactivated RASSF1A expression and induced apoptosis in breast cancer cells. In addition to modulating DNA methylation and chromatin remodelling, this combination also induced demethylation of the E2F1 transcription factor. The ChIP assay showed enhancement of E2F1 occupancy at the unmethylated RASSF1A promoter after TMCG/DIPY treatment. Interestingly, inhibition of E2F1 demethylation using an irreversible inhibitor of lysine-specific demethylase 1 reduced both TMCG/DIPY-mediated RASSF1A expression and apoptosis in MDA-MB-231 cells, suggesting that DNA and protein demethylation may act together to control these molecular and cellular processes. CONCLUSIONS/SIGNIFICANCE: This study demonstrates that simultaneous targeting of DNA and E2F1 methylation is an effective epigenetic treatment that reactivates RASSF1A expression and induces apoptosis in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Metilación de ADN/efectos de los fármacos , ADN de Neoplasias/genética , Factor de Transcripción E2F1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Catequina/análogos & derivados , Catequina/farmacología , Línea Celular Tumoral , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Dipiridamol/farmacología , Sinergismo Farmacológico , Factor de Transcripción E2F1/genética , Femenino , Genes Supresores de Tumor , Humanos , Células MCF-7 , Ratones , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Supresoras de Tumor/genética
13.
Exp Cell Res ; 318(10): 1146-59, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22484375

RESUMEN

Melanoma, the most aggressive form of skin cancer, is notoriously resistant to all current modalities of cancer therapy, including to the drug methotrexate. Melanosomal sequestration and cellular exportation of methotrexate have been proposed to be important melanoma-specific mechanisms that contribute to the resistance of melanoma to methotrexate. In addition, other mechanisms of resistance that are present in most epithelial cancer cells are also operative in melanoma. This report elucidates how melanoma orchestrates these mechanisms to become extremely resistant to methotrexate, where both E2F1 and checkpoint kinase 1 (Chk1), two molecules with dual roles in survival/apoptosis, play prominent roles. The results indicated that MTX induced the depletion of dihydrofolate in melanoma cells, which stimulated the transcriptional activity of E2F1. The elevate expression of dihydrofolate reductase and thymidylate synthase, two E2F1-target genes involved in folate metabolism and required for G(1) progression, favored dTTP accumulation, which promoted DNA single strand breaks and the subsequent activation of Chk1. Under these conditions, melanoma cells are protected from apoptosis by arresting their cell cycle in S phase. Excess of dTTP could also inhibit E2F1-mediated apoptosis in melanoma cells.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Melanoma/metabolismo , Metotrexato/farmacología , Neoplasias Cutáneas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Factor de Transcripción E2F1/química , Factor de Transcripción E2F1/metabolismo , Activación Enzimática/efectos de los fármacos , Ácido Fólico/metabolismo , Humanos , Melanoma/patología , Ratones , Datos de Secuencia Molecular , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Puntos de Control de la Fase S del Ciclo Celular , Neoplasias Cutáneas/patología , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Nucleótidos de Timina/metabolismo , Transcripción Genética
14.
ChemMedChem ; 6(3): 440-9, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21302360

RESUMEN

Despite bioavailability issues, tea catechins have emerged as promising chemopreventive agents because of their efficacy in various animal models. We synthesized two catechin-derived compounds, 3-O-(3,4,5-trimethoxybenzoyl)-(-)-catechin (TMCG) and 3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin (TMECG), in an attempt to improve the stability and cellular absorption of tea polyphenols. The antiproliferative and pro-apoptotic activities of both compounds were analyzed with various cancer cell systems, and TMCG, which was easily synthesized in excellent yield, was more active than TMECG in both melanoma and non-melanoma cell lines. TMCG was also a better inhibitor of dihydrofolate reductase and was more efficiently oxidized by tyrosinase, potentially explaining the difference in activity between these epimers.


Asunto(s)
Antineoplásicos/química , Catequina/análogos & derivados , Antagonistas del Ácido Fólico/síntesis química , Melanoma/tratamiento farmacológico , Monofenol Monooxigenasa/química , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Antioxidantes/síntesis química , Antioxidantes/química , Antioxidantes/uso terapéutico , Catequina/síntesis química , Catequina/química , Catequina/uso terapéutico , Línea Celular Tumoral , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/uso terapéutico , Humanos , Concentración de Iones de Hidrógeno , Modelos Moleculares , Monofenol Monooxigenasa/metabolismo , Oxidación-Reducción , Estereoisomerismo , Té/química , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo
15.
Int J Mol Sci ; 10(12): 5398-5410, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-20054477

RESUMEN

Dihydrofolate reductase (DHFR) is the subject of intensive investigation since it appears to be the primary target enzyme for antifolate drugs. Fluorescence quenching experiments show that the ester bond-containing tea polyphenols (-)-epigallocatechin gallate (EGCG) and (-)-epicatechin gallate (ECG) are potent inhibitors of DHFR with dissociation constants (K(D))of 0.9 and 1.8 microM, respectively, while polyphenols lacking the ester bound gallate moiety [e.g., (-)-epigallocatechin (EGC) and (-)-epicatechin (EC)] did not bind to this enzyme. To avoid stability and bioavailability problems associated with tea catechins we synthesized a methylated derivative of ECG (3-O-(3,4,5-trimethoxybenzoyl)-(-)-epicatechin; TMECG), which effectively binds to DHFR (K(D) = 2.1 microM). In alkaline solution, TMECG generates a stable quinone methide product that strongly binds to the enzyme with a K(D) of 8.2 nM. Quercetin glucuronides also bind to DHFR but its effective binding was highly dependent of the sugar residue, with quercetin-3-xyloside being the stronger inhibitor of the enzyme with a K(D) of 0.6 microM. The finding that natural polyphenols are good inhibitors of human DHFR could explain the epidemiological data on their prophylactic effects for certain forms of cancer and open a possibility for the use of natural and synthetic polyphenols in cancer chemotherapy.


Asunto(s)
Antagonistas del Ácido Fólico/química , Polifenoles/química , Tetrahidrofolato Deshidrogenasa/química , Secuencia de Aminoácidos , Sitios de Unión , Catequina/análogos & derivados , Catequina/síntesis química , Catequina/química , Antagonistas del Ácido Fólico/farmacología , Glucurónidos/química , Glucurónidos/farmacología , Humanos , Indolquinonas/química , Indolquinonas/farmacología , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Polifenoles/farmacología , Unión Proteica , Quercetina/química , Tetrahidrofolato Deshidrogenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...