Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Chem Biol ; 18(12): 1341-1350, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36229685

RESUMEN

Patients with castration-resistant prostate cancer inevitably acquire resistance to antiandrogen therapies in part because of androgen receptor (AR) mutations or splice variants enabling restored AR signaling. Here we show that ligand-activated AR can form transcriptionally active condensates. Both structured and unstructured regions of AR contribute to the effective phase separation of AR and disordered N-terminal domain plays a predominant role. AR liquid-liquid phase separation behaviors faithfully report transcriptional activity and antiandrogen efficacy. Antiandrogens can promote phase separation and transcriptional activity of AR-resistant mutants in a ligand-independent manner. We conducted a phase-separation-based phenotypic screen and identified ET516 that specifically disrupts AR condensates, effectively suppresses AR transcriptional activity and inhibits the proliferation and tumor growth of prostate cancer cells expressing AR-resistant mutants. Our results demonstrate liquid-liquid phase separation as an emerging mechanism underlying drug resistance and show that targeting phase separation may provide a feasible approach for drug discovery.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Masculino , Humanos , Receptores Androgénicos/genética , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Ligandos , Resistencia a Antineoplásicos , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología
2.
J Virol ; 87(13): 7348-56, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23616646

RESUMEN

The 2A proteinase (2A(pro)) is an enterovirally encoded cysteine protease that plays essential roles in both the processing of viral precursor polyprotein and the hijacking of host cell translation and other processes in the virus life cycle. Crystallographic studies of 2A(pro) from enterovirus 71 (EV71) and its interaction with the substrate are reported here. EV71 2A(pro) was comprised of an N-terminal domain of a four-stranded antiparallel ß sheet and a C-terminal domain of a six-stranded antiparallel ß barrel with a tightly bound zinc atom. Unlike in other 2A(pro) structures, there is an open cleft across the surface of the protein in an open conformation. As demonstrated by the crystallographic studies and modeling of the complex structure, the open cleft could be fitted with the substrate. On comparison 2A(pro) of EV71 to those of the human rhinovirus 2 and coxsackievirus B4, the open conformation could be closed with a hinge motion in the bII2 and cII ß strands. This was supported by molecular dynamic simulation. The structural variation among different 2A(pro) structures indicates a conformational flexibility in the substrate-binding cleft. The open structure provides an accessible framework for the design and development of therapeutics against the viral target.


Asunto(s)
Cisteína Endopeptidasas/química , Enterovirus Humano A/enzimología , Modelos Moleculares , Conformación Proteica , Secuencia de Aminoácidos , Cristalografía por Rayos X , Cisteína Endopeptidasas/genética , Escherichia coli , Transferencia Resonante de Energía de Fluorescencia , Vectores Genéticos/genética , Datos de Secuencia Molecular , Mutagénesis , Reacción en Cadena de la Polimerasa , Alineación de Secuencia
3.
J Med Chem ; 65(7): 5317-5333, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35352560

RESUMEN

Polycomb Repressive Complex 2 (PRC2) plays an important role in transcriptional regulation during animal development and in cell differentiation, and alteration of PRC2 activity has been associated with cancer. On a molecular level, PRC2 catalyzes methylation of histone H3 lysine 27 (H3K27), resulting in mono-, di-, or trimethylated forms of H3K27, of which the trimethylated form H3K27me3 leads to transcriptional repression of polycomb target genes. Previously, we have shown that binding of the low-molecular-weight compound EED226 to the H3K27me3 binding pocket of the regulatory subunit EED can effectively inhibit PRC2 activity in cells and reduce tumor growth in mouse xenograft models. Here, we report the stepwise optimization of the tool compound EED226 toward the potent and selective EED inhibitor MAK683 (compound 22) and its subsequent preclinical characterization. Based on a balanced PK/PD profile, efficacy, and mitigated risk of forming reactive metabolites, MAK683 has been selected for clinical development.


Asunto(s)
Histonas , Neoplasias , Animales , Inhibidores Enzimáticos , Histonas/metabolismo , Humanos , Metilación , Ratones , Neoplasias/tratamiento farmacológico , Complejo Represivo Polycomb 2
4.
BMC Pharmacol ; 10: 10, 2010 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-20735854

RESUMEN

BACKGROUND: Infarct-induced left ventricular (LV) remodeling is a deleterious consequence after acute myocardial infarction (MI) which may further advance to congestive heart failure. Therefore, new therapeutic strategies to attenuate the effects of LV remodeling are urgently needed. Salvianolic acid B (SalB) from Salviae mitiorrhizae, which has been widely used in China for the treatment of cardiovascular diseases, is a potential candidate for therapeutic intervention of LV remodeling targeting matrix metalloproteinase-9 (MMP-9). RESULTS: Molecular modeling and LIGPLOT analysis revealed in silico docking of SalB at the catalytic site of MMP-9. Following this lead, we expressed truncated MMP-9 which contains only the catalytic domain, and used this active protein for in-gel gelatin zymography, enzymatic analysis, and SalB binding by Biacore. Data generated from these assays indicated that SalB functioned as a competitive inhibitor of MMP-9. In our rat model for cardiac remodeling, western blot, echocardiography, hemodynamic measurement and histopathological detection were used to detect the effects and mechanism of SalB on cardio-protection. Our results showed that in MI rat, SalB selectively inhibited MMP-9 activities without affecting MMP-9 expression while no effect of SalB was seen on MMP-2. Moreover, SalB treatment in MI rat could efficiently increase left ventricle wall thickness, improve heart contractility, and decrease heart fibrosis. CONCLUSIONS: As a competitive inhibitor of MMP-9, SalB presents significant effects on preventing LV structural damage and preserving cardiac function. Further studies to develop SalB and its analogues for their potential for cardioprotection in clinic are warranted.


Asunto(s)
Benzofuranos/farmacología , Benzofuranos/uso terapéutico , Inhibidores de la Metaloproteinasa de la Matriz , Infarto del Miocardio/tratamiento farmacológico , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Animales , Benzofuranos/química , Benzofuranos/metabolismo , Sitios de Unión , Simulación por Computador , Regulación hacia Abajo/efectos de los fármacos , Diseño de Fármacos , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/química , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Conformación Molecular , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Distribución Aleatoria , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
5.
BMC Bioinformatics ; 9: 104, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18282303

RESUMEN

BACKGROUND: Target identification is important for modern drug discovery. With the advances in the development of molecular docking, potential binding proteins may be discovered by docking a small molecule to a repository of proteins with three-dimensional (3D) structures. To complete this task, a reverse docking program and a drug target database with 3D structures are necessary. To this end, we have developed a web server tool, TarFisDock (Target Fishing Docking) http://www.dddc.ac.cn/tarfisdock, which has been used widely by others. Recently, we have constructed a protein target database, Potential Drug Target Database (PDTD), and have integrated PDTD with TarFisDock. This combination aims to assist target identification and validation. DESCRIPTION: PDTD is a web-accessible protein database for in silico target identification. It currently contains >1100 protein entries with 3D structures presented in the Protein Data Bank. The data are extracted from the literatures and several online databases such as TTD, DrugBank and Thomson Pharma. The database covers diverse information of >830 known or potential drug targets, including protein and active sites structures in both PDB and mol2 formats, related diseases, biological functions as well as associated regulating (signaling) pathways. Each target is categorized by both nosology and biochemical function. PDTD supports keyword search function, such as PDB ID, target name, and disease name. Data set generated by PDTD can be viewed with the plug-in of molecular visualization tools and also can be downloaded freely. Remarkably, PDTD is specially designed for target identification. In conjunction with TarFisDock, PDTD can be used to identify binding proteins for small molecules. The results can be downloaded in the form of mol2 file with the binding pose of the probe compound and a list of potential binding targets according to their ranking scores. CONCLUSION: PDTD serves as a comprehensive and unique repository of drug targets. Integrated with TarFisDock, PDTD is a useful resource to identify binding proteins for active compounds or existing drugs. Its potential applications include in silico drug target identification, virtual screening, and the discovery of the secondary effects of an old drug (i.e. new pharmacological usage) or an existing target (i.e. new pharmacological or toxic relevance), thus it may be a valuable platform for the pharmaceutical researchers. PDTD is available online at http://www.dddc.ac.cn/pdtd/.


Asunto(s)
Bases de Datos de Proteínas , Sistemas de Liberación de Medicamentos/métodos , Diseño de Fármacos , Internet , Proteínas/química , Proteínas/ultraestructura , Interfaz Usuario-Computador , Sitios de Unión , Sistemas de Administración de Bases de Datos , Almacenamiento y Recuperación de la Información/métodos , Unión Proteica , Conformación Proteica , Análisis de Secuencia de Proteína/métodos
6.
Nucleic Acids Res ; 34(Web Server issue): W219-24, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16844997

RESUMEN

TarFisDock is a web-based tool for automating the procedure of searching for small molecule-protein interactions over a large repertoire of protein structures. It offers PDTD (potential drug target database), a target database containing 698 protein structures covering 15 therapeutic areas and a reverse ligand-protein docking program. In contrast to conventional ligand-protein docking, reverse ligand-protein docking aims to seek potential protein targets by screening an appropriate protein database. The input file of this web server is the small molecule to be tested, in standard mol2 format; TarFisDock then searches for possible binding proteins for the given small molecule by use of a docking approach. The ligand-protein interaction energy terms of the program DOCK are adopted for ranking the proteins. To test the reliability of the TarFisDock server, we searched the PDTD for putative binding proteins for vitamin E and 4H-tamoxifen. The top 2 and 10% candidates of vitamin E binding proteins identified by TarFisDock respectively cover 30 and 50% of reported targets verified or implicated by experiments; and 30 and 50% of experimentally confirmed targets for 4H-tamoxifen appear amongst the top 2 and 5% of the TarFisDock predicted candidates, respectively. Therefore, TarFisDock may be a useful tool for target identification, mechanism study of old drugs and probes discovered from natural products. TarFisDock and PDTD are available at http://www.dddc.ac.cn/tarfisdock/.


Asunto(s)
Diseño de Fármacos , Proteínas/química , Programas Informáticos , Bases de Datos de Proteínas , Internet , Ligandos , Proteínas/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/química , Tamoxifeno/metabolismo , Interfaz Usuario-Computador , Vitamina E/química , Vitamina E/metabolismo
7.
PLoS One ; 12(1): e0169855, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28072869

RESUMEN

Polycomb repressive complex 2 (PRC2), a histone H3 lysine 27 methyltransferase, plays a key role in gene regulation and is a known epigenetics drug target for cancer therapy. The WD40 domain-containing protein EED is the regulatory subunit of PRC2. It binds to the tri-methylated lysine 27 of the histone H3 (H3K27me3), and through which stimulates the activity of PRC2 allosterically. Recently, we disclosed a novel PRC2 inhibitor EED226 which binds to the K27me3-pocket on EED and showed strong antitumor activity in xenograft mice model. Here, we further report the identification and validation of four other EED binders along with EED162, the parental compound of EED226. The crystal structures for all these five compounds in complex with EED revealed a common deep pocket induced by the binding of this diverse set of compounds. This pocket was created after significant conformational rearrangement of the aromatic cage residues (Y365, Y148 and F97) in the H3K27me3 binding pocket of EED, the width of which was delineated by the side chains of these rearranged residues. In addition, all five compounds interact with the Arg367 at the bottom of the pocket. Each compound also displays unique features in its interaction with EED, suggesting the dynamics of the H3K27me3 pocket in accommodating the binding of different compounds. Our results provide structural insights for rational design of novel EED binder for the inhibition of PRC2 complex activity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Simulación del Acoplamiento Molecular , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Sulfonas/farmacología , Triazoles/farmacología , Animales , Sitios de Unión , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Ratones , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/metabolismo , Relación Estructura-Actividad Cuantitativa , Sulfonas/química , Triazoles/química
8.
J Med Chem ; 60(6): 2215-2226, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28092155

RESUMEN

Overexpression and somatic heterozygous mutations of EZH2, the catalytic subunit of polycomb repressive complex 2 (PRC2), are associated with several tumor types. EZH2 inhibitor, EPZ-6438 (tazemetostat), demonstrated clinical efficacy in patients with acceptable safety profile as monotherapy. EED, another subunit of PRC2 complex, is essential for its histone methyltransferase activity through direct binding to trimethylated lysine 27 on histone 3 (H3K27Me3). Herein we disclose the discovery of a first-in-class potent, selective, and orally bioavailable EED inhibitor compound 43 (EED226). Guided by X-ray crystallography, compound 43 was discovered by fragmentation and regrowth of compound 7, a PRC2 HTS hit that directly binds EED. The ensuing scaffold hopping followed by multiparameter optimization led to the discovery of 43. Compound 43 induces robust and sustained tumor regression in EZH2MUT preclinical DLBCL model. For the first time we demonstrate that specific and direct inhibition of EED can be effective as an anticancer strategy.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Sulfonas/química , Sulfonas/farmacología , Triazoles/química , Triazoles/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Perros , Femenino , Haplorrinos , Histonas/metabolismo , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/metabolismo , Lisina/metabolismo , Masculino , Metilación/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/metabolismo , Ratas , Sulfonas/farmacocinética , Sulfonas/uso terapéutico , Triazoles/farmacocinética , Triazoles/uso terapéutico
9.
ACS Med Chem Lett ; 7(10): 896-901, 2016 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-27774125

RESUMEN

A series of trisubstituted hydroxylactams was identified as potent enzymatic and cellular inhibitors of human lactate dehydrogenase A. Utilizing structure-based design and physical property optimization, multiple inhibitors were discovered with <10 µM lactate IC50 in a MiaPaca2 cell line. Optimization of the series led to 29, a potent cell active molecule (MiaPaca2 IC50 = 0.67 µM) that also possessed good exposure when dosed orally to mice.

10.
J Med Chem ; 55(16): 7037-53, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22862148

RESUMEN

LBVS of 12480 in-house compounds, followed by HTRF assay, resulted in one nonsteroidal compound (11) with antagonistic activity against FXR (69.01 ± 11.75 µM). On the basis of 11, 26 new derivatives (12a-z) were designed and synthesized accordingly. Five derivatives (12f-g, 12p, 12u, and 12y) showed better antagonistic activities against FXR than compound 11. Remarkably, the most potent derivative, 12u (8.96 ± 3.62 µM), showed antagonistic capability approximately 10 times and 8-fold higher than that of the control (GS) and the starting compound 11, respectively. 12u was further confirmed to have high binding affinity with FXRαLBD, FXR specificity over six other nuclear receptors, and potent antagonistic activity against FXR in two cell testing platforms. 12u strongly suppressed the regulating effects of CDCA on FXR target genes. The therapeutic potential of 12u was identified by lowering the contents of triglyceride and cholesterol in human hepatoma HepG2 cells and in the cholesterol-fed C57BL/6 mices.


Asunto(s)
Anticolesterolemiantes/síntesis química , Compuestos de Bencilideno/síntesis química , Pirazoles/síntesis química , Pirazolonas/síntesis química , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Animales , Anticolesterolemiantes/química , Anticolesterolemiantes/farmacología , Compuestos de Bencilideno/química , Compuestos de Bencilideno/farmacología , Disponibilidad Biológica , Ácido Quenodesoxicólico/farmacología , Colesterol/metabolismo , Bases de Datos de Compuestos Químicos , Células HEK293 , Células Hep G2 , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Pirazoles/química , Pirazoles/farmacología , Pirazolonas/química , Pirazolonas/farmacología , Ratas , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad , Transcripción Genética , Activación Transcripcional , Triglicéridos/metabolismo
11.
12.
Biochem Pharmacol ; 75(6): 1381-92, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18164690

RESUMEN

Deoxyelephantopin (ESD) was reported to potentiate apoptosis, inhibit invasion and abolish osteoclastogenesis but no target protein was disclosed. Here, we discovered that ESD could significantly inhibit the proliferation of different cancer cells and induce apoptosis and cell cycle arrest at G(2)/M phase in HeLa cell. Moreover, biochemical and biophysical assays revealed that ESD acted as a specific partial agonist against PPARgamma. Molecular docking with site-directed mutagenesis analyses indicated that ESD functioned as a partial agonist of PPARgamma by adopting a distinct binding mode to PPARgamma compared with rosiglitazone. The PPARgamma knockdown results indicated that the inhibition of ESD against the cancer cell proliferation is more possibly through PPARgamma-independent pathway and our findings might supply potent binding features for ESD/PPARgamma interaction at atomic level, and shed light on the potential acting target information for this natural compound.


Asunto(s)
Antineoplásicos/farmacología , Lactonas/farmacología , PPAR gamma/agonistas , Sesquiterpenos/farmacología , Animales , Antineoplásicos/metabolismo , Apoptosis , Células COS , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Células HeLa , Humanos , Lactonas/metabolismo , Modelos Moleculares , PPAR gamma/metabolismo , Sesquiterpenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA