Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 523(3): 795-801, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-31954521

RESUMEN

The DEAD-box family of RNA helicases plays essential roles in both transcriptional and translational mRNA degradation; they unwind short double-stranded RNA by breaking the RNA-RNA interactions. Two DEAD-box RNA helicases, eukaryotic translation initiation factor 4A3 (eIF4A3) and DEAD-box helicase 3 (DDX3X), show high homology in the ATP-binding region and are considered key molecules for cancer progression. Several small molecules that target eIF4A3 and DDX3X have been reported to inhibit cancer cell growth; however, more potent compounds are required for cancer therapeutics, and there is a critical need for high-throughput assays to screen for RNA helicase inhibitors. In this study, we developed novel fluorescence resonance energy transfer-based high-throughput RNA helicase assays for eIF4A3 and DDX3X. Using these assays, we identified several eIF4A3 allosteric inhibitors whose inhibitory effect on eIF4A3 ATPase showed a strong correlation with inhibitory effect on helicase activity. From 102 compounds that exhibited eIF4A3 ATPase inhibition, we identified a selective DDX3X inhibitor, C1, which showed stronger inhibition of DDX3X than of eIF4A3. Small-molecule helicase inhibitors can be valuable for clarifying the molecular machinery of DEAD-box RNA helicases. The high-throughput quantitative assays established here should facilitate the evaluation of the helicase inhibitory activity of compounds.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , ARN Helicasas DEAD-box/metabolismo , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Pruebas de Enzimas/métodos , Factor 4A Eucariótico de Iniciación/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Bibliotecas de Moléculas Pequeñas/química
2.
J Med Chem ; 61(17): 7710-7728, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30067358

RESUMEN

Cyclin-dependent kinase 12 (CDK12) plays a key role in the coordination of transcription with elongation and mRNA processing. CDK12 mutations found in tumors and CDK12 inhibition sensitize cancer cells to DNA-damaging reagents and DNA-repair inhibitors. This suggests that CDK12 inhibitors are potential therapeutics for cancer that may cause synthetic lethality. Here, we report the discovery of 3-benzyl-1-( trans-4-((5-cyanopyridin-2-yl)amino)cyclohexyl)-1-arylurea derivatives as novel and selective CDK12 inhibitors. Structure-activity relationship studies of a HTS hit, structure-based drug design, and conformation-oriented design using the Cambridge Structural Database afforded the optimized compound 2, which exhibited not only potent CDK12 (and CDK13) inhibitory activity and excellent selectivity but also good physicochemical properties. Furthermore, 2 inhibited the phosphorylation of Ser2 in the C-terminal domain of RNA polymerase II and induced growth inhibition in SK-BR-3 cells. Therefore, 2 represents an excellent chemical probe for functional studies of CDK12 and could be a promising lead compound for drug discovery.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Supervivencia Celular , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Inhibidores Enzimáticos/química , Femenino , Humanos , Fosforilación , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
3.
Bioorg Med Chem ; 26(12): 3261-3286, 2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-29754833

RESUMEN

We previously identified 2-tert-butyl-4-[(3-methoxypropyl)amino]-N-(2-methylpropyl)-N-[(3S,5R)-5-(morpholin-4-ylcarbonyl)piperidin-3-yl]pyrimidine-5-carboxamide 3 as a potent renin inhibitor. Since 3 showed unacceptably low bioavailability (BA) in rats, structural modification, using SBDD and focused on physicochemical properties was conducted to improve its PK profile while maintaining renin inhibitory activity. Conversion of the amino group attached at the 4-position of pyrimidine to methylene group improved PK profile and decreased renin inhibitory activity. New central cores with carbon side chains were explored to improve potency. We had designed a series of 5-membered azoles and fused heterocycles that interacted with the lipophilic S3 pocket. In the course of modification, renin inhibitory activity was enhanced by the formation of an additional hydrogen bonding with the hydroxyl group of Thr77. Consequently, a series of novel benzimidazole derivatives were discovered as potent and orally bioavailable renin inhibitors. Among those, compound 13 exhibited more than five-fold of plasma renin inhibition than aliskiren in cynomolgus monkeys at dose ratio.


Asunto(s)
Bencimidazoles/química , Piperidinas/química , Inhibidores de Proteasas/síntesis química , Renina/antagonistas & inhibidores , Administración Oral , Animales , Bencimidazoles/metabolismo , Bencimidazoles/farmacocinética , Sitios de Unión , Disponibilidad Biológica , Cristalografía por Rayos X , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Piperidinas/metabolismo , Piperidinas/farmacocinética , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacocinética , Estructura Terciaria de Proteína , Ratas , Renina/metabolismo , Relación Estructura-Actividad
4.
Oncotarget ; 9(26): 18480-18493, 2018 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-29719619

RESUMEN

Protein arginine methyltransferase (PRMT) 4 (also known as coactivator-associated arginine methyltransferase 1; CARM1) is involved in a variety of biological processes and is considered as a candidate oncogene owing to its overexpression in several types of cancer. Selective PRMT4 inhibitors are useful tools for clarifying the molecular events regulated by PRMT4 and for validating PRMT4 as a therapeutic target. Here, we report the discovery of TP-064, a potent, selective, and cell-active chemical probe of human PRMT4 and its co-crystal structure with PRMT4. TP-064 inhibited the methyltransferase activity of PRMT4 with high potency (half-maximal inhibitory concentration, IC50 < 10 nM) and selectivity over other PRMT family proteins, and reduced arginine dimethylation of the PRMT4 substrates BRG1-associated factor 155 (BAF155; IC50= 340 ± 30 nM) and Mediator complex subunit 12 (MED12; IC50 = 43 ± 10 nM). TP-064 treatment inhibited the proliferation of a subset of multiple myeloma cell lines, with affected cells arrested in G1 phase of the cell cycle. TP-064 and its negative control (TP-064N) will be valuable tools to further investigate the biology of PRMT4 and the therapeutic potential of PRMT4 inhibition.

5.
J Biol Chem ; 293(18): 6776-6790, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29545311

RESUMEN

Aberrant expression of proteins often underlies many diseases, including cancer. A recently developed approach in drug development is small molecule-mediated, selective degradation of dysregulated proteins. We have devised a protein-knockdown system that utilizes chimeric molecules termed specific and nongenetic IAP-dependent protein erasers (SNIPERs) to induce ubiquitylation and proteasomal degradation of various target proteins. SNIPER(ER)-87 consists of an inhibitor of apoptosis protein (IAP) ligand LCL161 derivative that is conjugated to the estrogen receptor α (ERα) ligand 4-hydroxytamoxifen by a PEG linker, and we have previously reported that this SNIPER efficiently degrades the ERα protein. Here, we report that derivatization of the IAP ligand module yields SNIPER(ER)s with superior protein-knockdown activity. These improved SNIPER(ER)s exhibited higher binding affinities to IAPs and induced more potent degradation of ERα than does SNIPER(ER)-87. Further, they induced simultaneous degradation of cellular inhibitor of apoptosis protein 1 (cIAP1) and delayed degradation of X-linked IAP (XIAP). Notably, these reengineered SNIPER(ER)s efficiently induced apoptosis in MCF-7 human breast cancer cells that require IAPs for continued cellular survival. We found that one of these molecules, SNIPER(ER)-110, inhibits the growth of MCF-7 tumor xenografts in mice more potently than the previously characterized SNIPER(ER)-87. Mechanistic analysis revealed that our novel SNIPER(ER)s preferentially recruit XIAP, rather than cIAP1, to degrade ERα. Our results suggest that derivatized IAP ligands could facilitate further development of SNIPERs with potent protein-knockdown and cytocidal activities against cancer cells requiring IAPs for survival.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Animales , Antineoplásicos/farmacología , Regulación hacia Abajo , Humanos , Ligandos , Células MCF-7 , Ratones , Unión Proteica , Proteolisis , Tiazoles/farmacología , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Biochemistry ; 57(8): 1369-1379, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29293322

RESUMEN

B-cell lymphoma 6 (BCL6) is the most frequently involved oncogene in diffuse large B-cell lymphomas (DLBCLs). BCL6 shows potent transcriptional repressor activity through interactions with its corepressors, such as BCL6 corepressor (BCOR). The inhibition of the protein-protein interaction (PPI) between BCL6 and its corepressors suppresses the growth of BCL6-dependent DLBCLs, thus making BCL6 an attractive drug target for lymphoma treatment. However, potent small-molecule PPI inhibitor identification remains challenging because of the lack of deep cavities at PPI interfaces. This article reports the discovery of a potent, cell-active small-molecule BCL6 inhibitor, BCL6-i (8), that operates through irreversible inhibition. First, we synthesized irreversible lead compound 4, which targets Cys53 in a cavity on the BCL6-BTB domain dimer by introducing an irreversible warhead to high-throughput screening hit compound 1. Further chemical optimization of 4 based on kinact/KI evaluation produced BCL6-i with a kinact/KI value of 1.9 × 104 M-1 s-1, corresponding to a 670-fold improvement in potency compared to that of 4. By exploiting the property of irreversible inhibition, engagement of BCL6-i to intracellular BCL6 was confirmed. BCL6-i showed intracellular PPI inhibitory activity between BCL6 and its corepressors, thus resulting in BCL6-dependent DLBCL cell growth inhibition. BCL6-i is a cell-active chemical probe with the most potent BCL6 inhibitory activity reported to date. The discovery process of BCL6-i illustrates the utility of irreversible inhibition for identifying potent chemical probes for intractable target proteins.


Asunto(s)
Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Línea Celular Tumoral , Cisteína/análisis , Cisteína/metabolismo , Descubrimiento de Drogas , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/metabolismo , Modelos Moleculares , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6/química , Bibliotecas de Moléculas Pequeñas/química
7.
J Med Chem ; 61(2): 543-575, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-28594553

RESUMEN

Targeted protein degradation using small molecules is a novel strategy for drug development. We have developed hybrid molecules named specific and nongenetic inhibitor of apoptosis protein [IAP]-dependent protein erasers (SNIPERs) that recruit IAP ubiquitin ligases to degrade target proteins. Here, we show novel SNIPERs capable of inducing proteasomal degradation of the androgen receptor (AR). Through derivatization of the SNIPER(AR) molecule at the AR ligand and IAP ligand and linker, we developed 42a (SNIPER(AR)-51), which shows effective protein knockdown activity against AR. Consistent with the degradation of the AR protein, 42a inhibits AR-mediated gene expression and proliferation of androgen-dependent prostate cancer cells. In addition, 42a efficiently induces caspase activation and apoptosis in prostate cancer cells, which was not observed in the cells treated with AR antagonists. These results suggest that SNIPER(AR)s could be leads for an anticancer drug against prostate cancers that exhibit AR-dependent proliferation.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Proteolisis/efectos de los fármacos , Receptores Androgénicos/metabolismo , Células A549 , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ligandos , Masculino , Neoplasias de la Próstata/patología , Relación Estructura-Actividad
8.
ACS Med Chem Lett ; 8(10): 1077-1082, 2017 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-29057054

RESUMEN

Starting from our previous eIF4A3-selective inhibitor 1a, a novel series of (piperazine-1-carbonyl)pyridin-2(1H)-one derivatives was designed, synthesized, and evaluated for identification of orally bioavailable probe molecules. Compounds 1o and 1q showed improved physicochemical and ADMET profiles, while maintaining potent and subtype-selective eIF4A3 inhibitory potency. In accord with their promising PK profiles and results from initial in vivo PD studies, compounds 1o and 1q showed antitumor efficacy with T/C values of 54% and 29%, respectively, without severe body weight loss. Thus, our novel series of compounds represents promising probe molecules for the in vivo pharmacological study of selective eIF4A3 inhibition.

9.
Bioorg Med Chem ; 25(17): 4876-4886, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28760529

RESUMEN

B-cell lymphoma 6 (BCL6) is a transcriptional repressor that can form complexes with corepressors via protein-protein interactions (PPIs). The complexes of BCL6 and corepressors play an important role in the formation of germinal centers (GCs), and differentiation and proliferation of lymphocytes. Therefore, BCL6-corepressor interaction inhibitors would be drug candidates for managing autoimmune diseases and cancer. Starting from high-throughput screening hits 1a and 2a, we identified a novel BCL6-corepressor interaction inhibitor 8c (cell-free enzyme-linked immunosorbent assay [ELISA] IC50=0.10µM, cell-based mammalian two-hybrid [M2H] assay IC50=0.72µM) by utilizing structure-based drug design (SBDD) based on an X-ray crystal structure of 1a bound to BCL6. Compound 8c also showed a good pharmacokinetic profile, which was acceptable for both in vitro and in vivo studies.


Asunto(s)
Diseño de Fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Aminas/química , Aminas/metabolismo , Aminas/farmacocinética , Sitios de Unión , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Semivida , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Simulación de Dinámica Molecular , Unión Proteica , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Técnicas del Sistema de Dos Híbridos
10.
J Med Chem ; 60(10): 4358-4368, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28471657

RESUMEN

B-cell lymphoma 6 (BCL6) is a transcriptional factor that expresses in lymphocytes and regulates the differentiation and proliferation of lymphocytes. Therefore, BCL6 is a therapeutic target for autoimmune diseases and cancer treatment. This report presents the discovery of BCL6-corepressor interaction inhibitors by using a biophysics-driven fragment-based approach. Using the surface plasmon resonance (SPR)-based fragment screening, we successfully identified fragment 1 (SPR KD = 1200 µM, ligand efficiency (LE) = 0.28), a competitive binder to the natural ligand BCoR peptide. Moreover, we elaborated 1 into the more potent compound 7 (SPR KD = 0.078 µM, LE = 0.37, cell-free protein-protein interaction (PPI) IC50 = 0.48 µM (ELISA), cellular PPI IC50 = 8.6 µM (M2H)) by a structure-based design and structural integration with a second high-throughput screening hit.


Asunto(s)
Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Descubrimiento de Drogas , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Proteínas Proto-Oncogénicas c-bcl-6/química , Resonancia por Plasmón de Superficie
11.
Cancer Sci ; 108(8): 1657-1666, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28556300

RESUMEN

Chromosomal translocation occurs in some cancer cells, which results in the expression of aberrant oncogenic fusion proteins that include BCR-ABL in chronic myelogenous leukemia (CML). Inhibitors of ABL tyrosine kinase, such as imatinib and dasatinib, exhibit remarkable therapeutic effects, although emergence of drug resistance hampers the therapy during long-term treatment. An alternative approach to treat CML is to downregulate the BCR-ABL protein. We have devised a protein knockdown system by hybrid molecules named Specific and Non-genetic inhibitor of apoptosis protein [IAP]-dependent Protein Erasers (SNIPER), which is designed to induce IAP-mediated ubiquitylation and proteasomal degradation of target proteins, and a couple of SNIPER(ABL) against BCR-ABL protein have been developed recently. In this study, we tested various combinations of ABL inhibitors and IAP ligands, and the linker was optimized for protein knockdown activity of SNIPER(ABL). The resulting SNIPER(ABL)-39, in which dasatinib is conjugated to an IAP ligand LCL161 derivative by polyethylene glycol (PEG) × 3 linker, shows a potent activity to degrade the BCR-ABL protein. Mechanistic analysis suggested that both cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP) play a role in the degradation of BCR-ABL protein. Consistent with the degradation of BCR-ABL protein, the SNIPER(ABL)-39 inhibited the phosphorylation of signal transducer and activator of transcription 5 (STAT5) and Crk like proto-oncogene (CrkL), and suppressed the growth of BCR-ABL-positive CML cells. These results suggest that SNIPER(ABL)-39 could be a candidate for a degradation-based novel anti-cancer drug against BCR-ABL-positive CML.


Asunto(s)
Dasatinib/farmacología , Proteínas de Fusión bcr-abl/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Tiazoles/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dasatinib/química , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células K562 , Ligandos , Fosforilación/efectos de los fármacos , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Proteolisis , Proto-Oncogenes Mas , Ubiquitinación/efectos de los fármacos
12.
ACS Chem Biol ; 12(7): 1760-1768, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28440616

RESUMEN

Eukaryotic initiation factor 4A-3 (eIF4A3) is an Asp-Glu-Ala-Asp (DEAD) box-family adenosine triphosphate (ATP)-dependent RNA helicase. Subtypes eIF4A1 and eIF4A2 are required for translation initiation, but eIF4A3 participates in the exon junction complex (EJC) and functions in RNA metabolism including nonsense-mediated RNA decay (NMD). No small molecules for NMD inhibition via selective inhibition of eIF4A3 have been discovered. Here, we identified allosteric eIF4A3 inhibitors from a high-throughput screening campaign. Chemical optimization of the lead compounds based on ATPase activity yielded compound 2, which exhibited noncompetitive inhibition with ATP or RNA and high selectivity for eIF4A3 over other helicases. The optimized compounds suppressed the helicase activity of eIF4A3 in an ATPase-dependent manner. Hydrogen/deuterium exchange mass spectrometry demonstrated that the deuterium-incorporation pattern of compound 2 overlapped with that of an allosteric pan-eIF4A inhibitor, hippuristanol, suggesting that compound 2 binds to an allosteric region on eIF4A3. We examined NMD activity using a luciferase-based cellular reporter system and a quantitative real-time polymerase chain-reaction-based cellular system to monitor levels of endogenous NMD substrates. NMD suppression by the compounds correlated positively with their ATPase-inhibitory activity. In conclusion, we developed a novel eIF4A3 inhibitor that targets the EJC. The optimized chemical probes represent useful tools for understanding the functions of eIF4A3 in RNA homeostasis.


Asunto(s)
ADN Helicasas/química , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Degradación de ARNm Mediada por Codón sin Sentido/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Regulación Alostérica , Secuencia de Aminoácidos , Unión Competitiva , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Concentración 50 Inhibidora , Alineación de Secuencia , Bibliotecas de Moléculas Pequeñas/farmacología , Esteroles/química , Esteroles/farmacología
13.
J Med Chem ; 60(8): 3335-3351, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28358513

RESUMEN

Eukaryotic initiation factor 4A3 (eIF4A3), a member of the DEAD-box RNA helicase family, is one of the core components of the exon junction complex (EJC). The EJC is known to be involved in a variety of RNA metabolic processes typified by nonsense-mediated RNA decay (NMD). In order to identify molecular probes to investigate the functions and therapeutic relevance of eIF4A3, a search for selective eIF4A3 inhibitors was conducted. Through the chemical optimization of 1,4-diacylpiperazine derivatives identified via high-throughput screening (HTS), we discovered the first reported selective eIF4A3 inhibitor 53a exhibiting cellular NMD inhibitory activity. A surface plasmon resonance (SPR) biosensing assay ascertained the direct binding of 53a and its analog 52a to eIF4A3 and revealed that the binding occurs at a non-ATP binding site. Compounds 52a and 53a represent novel molecular probes for further study of eIF4A3, the EJC, and NMD.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Piperazinas/farmacología , Adenosina Trifosfatasas/antagonistas & inhibidores , Descubrimiento de Drogas , Células HEK293 , Humanos , Espectrometría de Masas , Piperazinas/química , Espectroscopía de Protones por Resonancia Magnética , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie
14.
Bioorg Med Chem ; 25(7): 2200-2209, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28283335

RESUMEN

Eukaryotic initiation factor 4A3 (eIF4A3), an ATP-dependent RNA helicase, is a core component of exon junction complex (EJC). EJC has a variety of roles in RNA metabolism such as translation, surveillance, and localization of spliced RNA. It is worthwhile to identify selective eIF4A3 inhibitors with a view to investigating the functions of eIF4A3 and EJC further to clarify the roles of the ATPase and helicase activities in cells. Our chemical optimization of hit compound 2 culminated in the discovery of ATP-competitive eIF4A3 inhibitor 18 with submicromolar ATPase inhibitory activity and excellent selectivity over other helicases. Hence, compound 18 could be a valuable chemical probe to elucidate the detailed functions of eIF4A3 and EJC.


Asunto(s)
Adenosina Trifosfato/metabolismo , ARN Helicasas DEAD-box/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Historia Medieval , Espectroscopía de Protones por Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray
15.
J Biol Chem ; 292(11): 4556-4570, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28154167

RESUMEN

Many diseases, especially cancers, result from aberrant or overexpression of pathogenic proteins. Specific inhibitors against these proteins have shown remarkable therapeutic effects, but these are limited mainly to enzymes. An alternative approach that may have utility in drug development relies on selective degradation of pathogenic proteins via small chimeric molecules linking an E3 ubiquitin ligase to the targeted protein for proteasomal degradation. To this end, we recently developed a protein knockdown system based on hybrid small molecule SNIPERs (Specific and Nongenetic IAP-dependent Protein Erasers) that recruit inhibitor of the apoptosis protein (IAP) ubiquitin ligases to specifically degrade targeted proteins. Here, we extend our previous study to show a proof of concept of the SNIPER technology in vivo By incorporating a high affinity IAP ligand, we developed a novel SNIPER against estrogen receptor α (ERα), SNIPER(ER)-87, that has a potent protein knockdown activity. The SNIPER(ER) reduced ERα levels in tumor xenografts and suppressed the growth of ERα-positive breast tumors in mice. Mechanistically, it preferentially recruits X-linked IAP (XIAP) rather than cellular IAP1, to degrade ERα via the ubiquitin-proteasome pathway. With this IAP ligand, potent SNIPERs against other pathogenic proteins, BCR-ABL, bromodomain-containing protein 4 (BRD4), and phosphodiesterase-4 (PDE4) could also be developed. These results indicate that forced ubiquitylation by SNIPERs is a useful method to achieve efficient protein knockdown with potential therapeutic activities and could also be applied to study the role of ubiquitylation in many cellular processes.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Receptor alfa de Estrógeno/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteolisis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Animales , Antineoplásicos/farmacología , Mama/efectos de los fármacos , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Descubrimiento de Drogas , Receptor alfa de Estrógeno/antagonistas & inhibidores , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Ligandos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Complejo de la Endopetidasa Proteasomal/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitinación/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
16.
Biochem Biophys Res Commun ; 482(2): 310-316, 2017 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-27856253

RESUMEN

B cell lymphoma 6 (BCL6) is a transcriptional repressor that interacts with its corepressors BcoR and SMRT. Since this protein-protein interaction (PPI) induces activation and differentiation of B lymphocytes, BCL6 has been an attractive drug target for potential autoimmune disease treatments. Here we report a novel BCL6 inhibitory peptide, F1324 (Ac-LWYTDIRMSWRVP-OH), which we discovered using phage display technology; we also discuss this peptide's structure-activity relationship (SAR). For BCL6(5-129) binding, KD and IC50 values of F1324 were 0.57 nM and 1 nM according to the results of an SPR analysis and cell-free ELISA assay, respectively. In contrast, BcoR(Arg498-514Pro) and SMRT(Leu1422-Arg1438) exhibited relatively weak micromole-order binding to BCL6. Furthermore, Fusion protein AcGFP-F1324 transiently expressed in HEK293T cells inhibited intracellular PPI in cell-based M2H assay. By examination of the truncation and fragmentation of F1324, the C-terminal sequence WRVP, which is similar to the BcoR(509-512) sequence WVVP, was identified as being critical for BCL6 binding. In addition, subsequent single-crystal X-ray diffraction analysis of F1324/BCL6(5-129) complex revealed that the high affinity of F1324 was caused by effective interaction of its side chains while its main chain structure was similar to that of BcoR(Arg498-514Pro). To our knowledge, F1324 is the strongest BCL6-binding peptide yet reported.


Asunto(s)
Inhibidores Enzimáticos/química , Péptidos/química , Proteínas Proto-Oncogénicas c-bcl-6/química , Proteínas Proto-Oncogénicas c-bcl-6/ultraestructura , Sitios de Unión , Activación Enzimática , Unión Proteica , Relación Estructura-Actividad
17.
Bioorg Med Chem ; 24(22): 5771-5780, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27687967

RESUMEN

The action of the aspartyl protease renin is the rate-limiting initial step of the renin-angiotensin-aldosterone system. Therefore, renin is a particularly promising target for blood pressure as well as onset and progression of cardiovascular and renal diseases. New pyrimidine derivatives 5-14 were designed in an attempt to enhance the renin inhibitory activity of compound 3 identified by our previous fragment-based drug design approach. Introduction of a basic amine essential for interaction with the two aspartic acids in the catalytic site and optimization of the S1/S3 binding elements including an induced-fit structural change of Leu114 ('Leu-in' to 'Leu-out') by a rational structure-based drug design approach led to the discovery of N-(piperidin-3-yl)pyrimidine-5-carboxamide 14, a 65,000-fold more potent renin inhibitor than compound 3. Surprisingly, this remarkable enhancement in the inhibitory activity of compound 14 has been achieved by the overall addition of only seven heavy atoms to compound 3. Compound 14 demonstrated excellent selectivity over other aspartyl proteases and moderate oral bioavailability in rats.


Asunto(s)
Diseño de Fármacos , Piperidinas/farmacología , Inhibidores de Proteasas/farmacología , Pirimidinas/farmacología , Renina/antagonistas & inhibidores , Animales , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Pirimidinas/síntesis química , Pirimidinas/química , Ratas , Ratas Sprague-Dawley , Renina/sangre , Relación Estructura-Actividad
18.
ACS Med Chem Lett ; 7(10): 933-938, 2016 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-27774132

RESUMEN

The aspartic proteinase renin is an attractive target for the treatment of hypertension and cardiovascular/renal disease such as chronic kidney disease and heart failure. We introduced an S1' site binder into the lead compound 1 guided by structure-based drug design (SBDD), and further optimization of physicochemical properties led to the discovery of benzimidazole derivative 10 (1-(4-methoxybutyl)-N-(2-methylpropyl)-N-[(3S,5R)-5-(morpholin-4-yl)carbonylpiperidin-3-yl]-1H-benzimidazole-2-carboxamide hydrochloride, TAK-272) as a highly potent and orally active renin inhibitor. Compound 10 demonstrated good oral bioavailability (BA) and long-lasting efficacy in rats. Compound 10 is currently in clinical trials.

19.
Bioorg Med Chem ; 24(22): 6066-6074, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27720325

RESUMEN

A novel approach was conducted for fragment-based lead discovery and applied to renin inhibitors. The biochemical screening of a fragment library against renin provided the hit fragment which showed a characteristic interaction pattern with the target protein. The hit fragment bound only to the S1, S3, and S3SP (S3 subpocket) sites without any interactions with the catalytic aspartate residues (Asp32 and Asp215 (pepsin numbering)). Prior to making chemical modifications to the hit fragment, we first identified its essential binding sites by utilizing the hit fragment's substructures. Second, we created a new and smaller scaffold, which better occupied the identified essential S3 and S3SP sites, by utilizing library synthesis with high-throughput chemistry. We then revisited the S1 site and efficiently explored a good building block attaching to the scaffold with library synthesis. In the library syntheses, the binding modes of each pivotal compound were determined and confirmed by X-ray crystallography and the library was strategically designed by structure-based computational approach not only to obtain a more active compound but also to obtain informative Structure Activity Relationship (SAR). As a result, we obtained a lead compound offering synthetic accessibility as well as the improved in vitro ADMET profiles. The fragments and compounds possessing a characteristic interaction pattern provided new structural insights into renin's active site and the potential to create a new generation of renin inhibitors. In addition, we demonstrated our FBDD strategy integrating highly sensitive biochemical assay, X-ray crystallography, and high-throughput synthesis and in silico library design aimed at fragment morphing at the initial stage was effective to elucidate a pocket profile and a promising lead compound.


Asunto(s)
Descubrimiento de Drogas , Inhibidores de Proteasas/farmacología , Renina/antagonistas & inhibidores , Animales , Células CHO , Supervivencia Celular/efectos de los fármacos , Cricetulus , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Renina/metabolismo , Relación Estructura-Actividad
20.
Mol Endocrinol ; 28(2): 249-59, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24422633

RESUMEN

Therapies based on conventional nuclear receptor ligands are extremely powerful, yet their broad and long-term use is often hindered by undesired side effects that are often part of the receptor's biological function. Selective control of nuclear receptors such as the glucocorticoid receptor (GR) using conventional ligands has proven particularly challenging. Because they act solely in an allosteric manner, conventional ligands are constrained to act via cofactors that can intrinsically partner with the receptor. Furthermore, effective means to rationally encode a bias for specific coregulators are generally lacking. Using the (GR) as a framework, we demonstrate here a versatile approach, based on bifunctional ligands, that extends the regulatory repertoire of GR in a deliberate and controlled manner. By linking the macrolide FK506 to a conventional agonist (dexamethasone) or antagonist (RU-486), we demonstrate that it is possible to bridge the intact receptor to either positively or negatively acting coregulatory proteins bearing an FK506 binding protein domain. Using this strategy, we show that extrinsic recruitment of a strong activation function can enhance the efficacy of the full agonist dexamethasone and reverse the antagonist character of RU-486 at an endogenous locus. Notably, the extrinsic recruitment of histone deacetylase-1 reduces the ability of GR to activate transcription from a canonical GR response element while preserving ligand-mediated repression of nuclear factor-κB. By providing novel ways for the receptor to engage specific coregulators, this unique ligand design approach has the potential to yield both novel tools for GR study and more selective therapeutics.


Asunto(s)
Receptores de Glucocorticoides/fisiología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Dexametasona/farmacología , Células HEK293 , Histona Desacetilasa 1/metabolismo , Humanos , Ligandos , Mifepristona/farmacología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Unión Proteica , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/antagonistas & inhibidores , Tacrolimus/farmacología , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...