Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Commun ; 9(1): 2732, 2018 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-30013058

RESUMEN

Genetic aberrations driving pro-oncogenic and pro-metastatic activity remain an elusive target in the quest of precision oncology. To identify such drivers, we use an animal model of KRAS-mutant lung adenocarcinoma to perform an in vivo functional screen of 217 genetic aberrations selected from lung cancer genomics datasets. We identify 28 genes whose expression promoted tumor metastasis to the lung in mice. We employ two tools for examining the KRAS-dependence of genes identified from our screen: 1) a human lung cell model containing a regulatable mutant KRAS allele and 2) a lentiviral system permitting co-expression of DNA-barcoded cDNAs with Cre recombinase to activate a mutant KRAS allele in the lungs of mice. Mechanistic evaluation of one gene, GATAD2B, illuminates its role as a dual activity gene, promoting both pro-tumorigenic and pro-metastatic activities in KRAS-mutant lung cancer through interaction with c-MYC and hyperactivation of the c-MYC pathway.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Factores de Transcripción GATA/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma del Pulmón/mortalidad , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/terapia , Animales , Línea Celular Tumoral , Femenino , Factores de Transcripción GATA/antagonistas & inhibidores , Factores de Transcripción GATA/metabolismo , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Integrasas/genética , Integrasas/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras , Transducción de Señal , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Bioinformatics ; 34(7): 1183-1191, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29186335

RESUMEN

Motivation: As cancer genomics initiatives move toward comprehensive identification of genetic alterations in cancer, attention is now turning to understanding how interactions among these genes lead to the acquisition of tumor hallmarks. Emerging pharmacological and clinical data suggest a highly promising role of cancer-specific protein-protein interactions (PPIs) as druggable cancer targets. However, large-scale experimental identification of cancer-related PPIs remains challenging, and currently available resources to explore oncogenic PPI networks are limited. Results: Recently, we have developed a PPI high-throughput screening platform to detect PPIs between cancer-associated proteins in the context of cancer cells. Here, we present the OncoPPi Portal, an interactive web resource that allows investigators to access, manipulate and interpret a high-quality cancer-focused network of PPIs experimentally detected in cancer cell lines. To facilitate prioritization of PPIs for further biological studies, this resource combines network connectivity analysis, mutual exclusivity analysis of genomic alterations, cellular co-localization of interacting proteins and domain-domain interactions. Estimates of PPI essentiality allow users to evaluate the functional impact of PPI disruption on cancer cell proliferation. Furthermore, connecting the OncoPPi network with the approved drugs and compounds in clinical trials enables discovery of new tumor dependencies to inform strategies to interrogate undruggable targets like tumor suppressors. The OncoPPi Portal serves as a resource for the cancer research community to facilitate discovery of cancer targets and therapeutic development. Availability and implementation: The OncoPPi Portal is available at http://oncoppi.emory.edu. Contact: andrey.ivanov@emory.edu or hfu@emory.edu.


Asunto(s)
Nube Computacional , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Mapeo de Interacción de Proteínas/métodos , Humanos , Internet
4.
Nat Commun ; 8: 14356, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28205554

RESUMEN

As genomics advances reveal the cancer gene landscape, a daunting task is to understand how these genes contribute to dysregulated oncogenic pathways. Integration of cancer genes into networks offers opportunities to reveal protein-protein interactions (PPIs) with functional and therapeutic significance. Here, we report the generation of a cancer-focused PPI network, termed OncoPPi, and identification of >260 cancer-associated PPIs not in other large-scale interactomes. PPI hubs reveal new regulatory mechanisms for cancer genes like MYC, STK11, RASSF1 and CDK4. As example, the NSD3 (WHSC1L1)-MYC interaction suggests a new mechanism for NSD3/BRD4 chromatin complex regulation of MYC-driven tumours. Association of undruggable tumour suppressors with drug targets informs therapeutic options. Based on OncoPPi-derived STK11-CDK4 connectivity, we observe enhanced sensitivity of STK11-silenced lung cancer cells to the FDA-approved CDK4 inhibitor palbociclib. OncoPPi is a focused PPI resource that links cancer genes into a signalling network for discovery of PPI targets and network-implicated tumour vulnerabilities for therapeutic interrogation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/efectos de los fármacos , Redes Reguladoras de Genes/genética , Oncogenes/efectos de los fármacos , Oncogenes/genética , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas de Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Bases de Datos de Proteínas , Genes Supresores de Tumor/efectos de los fármacos , Genes myc/genética , Genómica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Terapia Molecular Dirigida , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oncogenes/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , Mapeo de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
Mol Pharmacol ; 91(4): 339-347, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28087810

RESUMEN

The c-Myc (MYC) transcription factor is a major cancer driver and a well-validated therapeutic target. However, directly targeting MYC has been challenging. Thus, identifying proteins that interact with and regulate MYC may provide alternative strategies to inhibit its oncogenic activity. In this study, we report the development of a NanoLuc-based protein-fragment complementation assay (NanoPCA) and mapping of the MYC protein interaction hub in live mammalian cells. The NanoPCA system was configured to enable detection of protein-protein interactions (PPI) at the endogenous level, as shown with PRAS40 dimerization, and detection of weak interactions, such as PINCH1-NCK2. Importantly, NanoPCA allows the study of PPI dynamics with reversible interactions. To demonstrate its utility for large-scale PPI detection in mammalian intracellular environment, we have used NanoPCA to examine MYC interaction with 83 cancer-associated proteins in live cancer cell lines. Our new MYC PPI data confirmed known MYC-interacting proteins, such as MAX, GSK3A, and SMARCA4, and revealed a panel of novel MYC interaction partners, such as RAC-α serine/threonine-protein kinase (AKT)1, liver kinase B (LKB)1, and Yes-associated protein (YAP)1. The MYC interactions with AKT1, LKB1, and YAP1 were confirmed by coimmunoprecipitation of endogenous proteins. Importantly, AKT1, LKB1, and YAP1 were able to activate MYC in a transcriptional reporter assay. Thus, these vital growth control proteins may represent promising MYC regulators, suggesting new mechanisms that couple energetic and metabolic pathways and developmental signaling to MYC-regulated cellular programs.


Asunto(s)
Bioensayo , Luciferasas/metabolismo , Nanopartículas/química , Fosfoproteínas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Línea Celular Tumoral , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Unión Proteica , Reproducibilidad de los Resultados
6.
PLoS One ; 12(1): e0170339, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28118365

RESUMEN

Protein-protein interactions (PPIs) mediate the transmission and regulation of oncogenic signals that are essential to cellular proliferation and survival, and thus represent potential targets for anti-cancer therapeutic discovery. Despite their significance, there is no method to experimentally disrupt and interrogate the essentiality of individual endogenous PPIs. The ability to computationally predict or infer PPI essentiality would help prioritize PPIs for drug discovery and help advance understanding of cancer biology. Here we introduce a computational method (MEDICI) to predict PPI essentiality by combining gene knockdown studies with network models of protein interaction pathways in an analytic framework. Our method uses network topology to model how gene silencing can disrupt PPIs, relating the unknown essentialities of individual PPIs to experimentally observed protein essentialities. This model is then deconvolved to recover the unknown essentialities of individual PPIs. We demonstrate the validity of our approach via prediction of sensitivities to compounds based on PPI essentiality and differences in essentiality based on genetic mutations. We further show that lung cancer patients have improved overall survival when specific PPIs are no longer present, suggesting that these PPIs may be potentially new targets for therapeutic development. Software is freely available at https://github.com/cooperlab/MEDICI. Datasets are available at https://ctd2.nci.nih.gov/dataPortal.


Asunto(s)
Antineoplásicos/farmacología , Minería de Datos/métodos , Descubrimiento de Drogas , Proteínas de Neoplasias/metabolismo , Programas Informáticos , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Análisis por Conglomerados , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Terapia Molecular Dirigida , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Redes Neurales de la Computación , Mapeo de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
7.
J Mol Cell Biol ; 8(3): 271-81, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26578655

RESUMEN

Large-scale genomics studies have generated vast resources for in-depth understanding of vital biological and pathological processes. A rising challenge is to leverage such enormous information to rapidly decipher the intricate protein-protein interactions (PPIs) for functional characterization and therapeutic interventions. While a number of powerful technologies have been employed to detect PPIs, a singular PPI biosensor platform with both high sensitivity and robustness in a mammalian cell environment remains to be established. Here we describe the development and integration of a highly sensitive NanoLuc luciferase-based bioluminescence resonance energy transfer technology, termed BRET(n), which enables ultra-high-throughput (uHTS) PPI detection in live cells with streamlined co-expression of biosensors in a miniaturized format. We further demonstrate the application of BRET(n) in uHTS format in chemical biology research, including the discovery of chemical probes that disrupt PRAS40 dimerization and pathway connectivity profiling among core members of the Hippo signaling pathway. Such hippo pathway profiling not only confirmed previously reported PPIs, but also revealed two novel interactions, suggesting new mechanisms for regulation of Hippo signaling. Our BRET(n) biosensor platform with uHTS capability is expected to accelerate systematic PPI network mapping and PPI modulator-based drug discovery.


Asunto(s)
Técnicas Biosensibles/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Mapeo de Interacción de Proteínas/métodos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fluorescencia , Células HEK293 , Humanos , Imidazoles/farmacología , Luciferasas/metabolismo , Miniaturización , Piperazinas/farmacología , Multimerización de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína p53 Supresora de Tumor/metabolismo
8.
Trends Pharmacol Sci ; 34(7): 393-400, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23725674

RESUMEN

The emergence and convergence of cancer genomics, targeted therapies, and network oncology have significantly expanded the landscape of protein-protein interaction (PPI) networks in cancer for therapeutic discovery. Extensive biological and clinical investigations have led to the identification of protein interaction hubs and nodes that are critical for the acquisition and maintenance of characteristics of cancer essential for cell transformation. Such cancer-enabling PPIs have become promising therapeutic targets. With technological advances in PPI modulator discovery and validation of PPI-targeting agents in clinical settings, targeting of PPI interfaces as an anticancer strategy has become a reality. Future research directed at genomics-based PPI target discovery, PPI interface characterization, PPI-focused chemical library design, and patient-genomic subpopulation-driven clinical studies is expected to accelerate the development of the next generation of PPI-based anticancer agents for personalized precision medicine. Here we briefly review prominent PPIs that mediate cancer-acquired properties, highlight recognized challenges and promising clinical results in targeting PPIs, and outline emerging opportunities.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Terapia Molecular Dirigida , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Mapeo de Interacción de Proteínas , Animales , Antineoplásicos/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Estructura Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
9.
Handb Exp Pharmacol ; (193): 123-59, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19639281

RESUMEN

A(3) adenosine receptor (A(3)AR) ligands have been modified to optimize their interaction with the A(3)AR. Most of these modifications have been made to the N(6) and C2 positions of adenine as well as the ribose moiety, and using a combination of these substitutions leads to the most efficacious, selective, and potent ligands. A(3)AR agonists such as IB-MECA and Cl-IB-MECA are now advancing into Phase II clinical trials for treatments targeting diseases such as cancer, arthritis, and psoriasis. Also, a wide number of compounds exerting high potency and selectivity in antagonizing the human (h)A(3)AR have been discovered. These molecules are generally characterized by a notable structural diversity, taking into account that aromatic nitrogen-containing monocyclic (thiazoles and thiadiazoles), bicyclic (isoquinoline, quinozalines, (aza)adenines), tricyclic systems (pyrazoloquinolines, triazoloquinoxalines, pyrazolotriazolopyrimidines, triazolopurines, tricyclic xanthines) and nucleoside derivatives have been identified as potent and selective A(3)AR antagonists. Probably due to the "enigmatic" physiological role of A(3)AR, whose activation may produce opposite effects (for example, concerning tissue protection in inflammatory and cancer cells) and may produce effects that are species dependent, only a few molecules have reached preclinical investigation. Indeed, the most advanced A(3)AR antagonists remain in preclinical testing. Among the antagonists described above, compound OT-7999 is expected to enter clinical trials for the treatment of glaucoma, while several thiazole derivatives are in development as antiallergic, antiasthmatic and/or antiinflammatory drugs.


Asunto(s)
Agonistas del Receptor de Adenosina A3 , Antagonistas del Receptor de Adenosina A3 , Animales , Química Farmacéutica , Humanos , Relación Estructura-Actividad
10.
J Med Chem ; 52(23): 7580-92, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19499950

RESUMEN

(N)-Methanocarba nucleosides containing bicyclo[3.1.0]hexane replacement of the ribose ring previously demonstrated selectivity as A(3) adenosine receptor (AR) agonists (5'-uronamides) or antagonists (5'-truncated). Here, these two series were modified in parallel at the adenine C2 position. N(6)-3-Chlorobenzyl-5'-N-methyluronamides derivatives with functionalized 2-alkynyl chains of varying length terminating in a reactive carboxylate, ester, or amine group were full, potent human A(3)AR agonists. Flexibility of chain substitution allowed the conjugation with a fluorescent cyanine dye (Cy5) and biotin, resulting in binding K(i) values of 17 and 36 nM, respectively. The distal end of the chain was predicted by homology modeling to bind at the A(3)AR extracellular regions. Corresponding l-nucleosides were nearly inactive in AR binding. In the 5'-truncated nucleoside series, 2-Cl analogues were more potent at A(3)AR than 2-H and 2-F, functional efficacy in adenylate cyclase inhibition varied, and introduction of a 2-alkynyl chain greatly reduced affinity. SAR parallels between the two series lost stringency at distal positions. The most potent and selective novel compounds were amine congener 15 (K(i) = 2.1 nM) and truncated partial agonist 22 (K(i) = 4.9 nM).


Asunto(s)
Agonistas del Receptor de Adenosina A3 , Antagonistas del Receptor de Adenosina A3 , Compuestos Bicíclicos con Puentes/química , Nucleósidos/química , Nucleósidos/farmacología , Amidas/química , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Ligandos , Modelos Moleculares , Conformación Molecular , Nucleósidos/síntesis química , Nucleósidos/metabolismo , Receptor de Adenosina A3/química , Receptor de Adenosina A3/metabolismo , Estereoisomerismo , Especificidad por Sustrato
11.
J Med Chem ; 52(10): 3284-92, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19402631

RESUMEN

Homology modeling of the human A(2A) adenosine receptor (AR) based on bovine rhodopsin predicted a protein structure that was very similar to the recently determined crystallographic structure. The discrepancy between the experimentally observed orientation of the antagonist and those obtained by previous antagonist docking is related to the loop structure of rhodopsin being carried over to the model of the A(2A) AR and was rectified when the beta(2)-adrenergic receptor was used as a template for homology modeling. Docking of the triazolotriazine antagonist ligand ZM241385 1 was greatly improved by including water molecules of the X-ray structure or by using a constraint from mutagenesis. Automatic agonists docking to both a new homology modeled receptor and the A(2A) AR crystallographic structure produced similar results. Heterocyclic nitrogen atoms closely corresponded when the docked adenine moiety of agonists and 1 were overlaid. The cumulative mutagenesis data, which support the proposed mode of agonist docking, can be reexamined in light of the crystallographic structure. Thus, homology modeling of GPCRs remains a useful technique in probing the structure of the protein and predicting modes of ligand docking.


Asunto(s)
Simulación por Computador , Modelos Moleculares , Receptor de Adenosina A2A/química , Receptores Acoplados a Proteínas G/química , Agonistas del Receptor de Adenosina A2 , Antagonistas del Receptor de Adenosina A2 , Animales , Bovinos , Cristalografía por Rayos X , Humanos , Mutagénesis , Unión Proteica , Conformación Proteica , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Rodopsina/agonistas , Rodopsina/antagonistas & inhibidores , Rodopsina/química , Homología de Secuencia de Aminoácido
12.
Bioorg Med Chem Lett ; 18(18): 5091-4, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18715782

RESUMEN

Several adamantane-based taxol mimetics were synthesized and found to be cytotoxic at micromolar concentrations and to cause tubulin aggregation. The extent of the aggregation is maximal for N-benzoyl-(2R,3S)-phenylisoseryloxyadamantane (5) and is very sensitive to the structural modifications. A hybrid compound (15), combining adamantane-based taxol mimetic with colchicine was synthesized and found to possess both microtubule depolymerizing and microtubule bundling activities in A549 human lung carcinoma cells.


Asunto(s)
Adamantano , Antineoplásicos Fitogénicos , Tubulina (Proteína)/metabolismo , Adamantano/análogos & derivados , Adamantano/síntesis química , Adamantano/química , Adamantano/farmacología , Animales , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Encéfalo/metabolismo , Bovinos , Colchicina/farmacología , Técnicas Químicas Combinatorias , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Microtúbulos/metabolismo , Imitación Molecular , Paclitaxel/farmacología , Relación Estructura-Actividad , Tubulina (Proteína)/química
13.
Bioorg Med Chem Lett ; 18(15): 4312-5, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18639453

RESUMEN

The theoretical possibility of bivalent binding of a dendrimer, covalently appended with multiple copies of a small ligand, to a homodimer of a G protein-coupled receptor was investigated with a molecular modeling approach. A molecular model was constructed of a third generation (G3) poly(amidoamine) (PAMAM) dendrimer condensed with multiple copies of the potent A(2A) adenosine receptor agonist CGS21680. The dendrimer was bound to an A(2A) adenosine receptor homodimer. Two units of the nucleoside CGS21680 could occupy the A(2A) receptor homodimer simultaneously. The binding mode of CGS21680 moieties linked to the PAMAM dendrimer and docked to the A(2A) receptor was found to be similar to the binding mode of a monomeric CGS21680 ligand.


Asunto(s)
Agonistas del Receptor de Adenosina A2 , Adenosina/análogos & derivados , Dendrímeros , Modelos Moleculares , Fenetilaminas/química , Poliaminas/química , Receptores Acoplados a Proteínas G/metabolismo , Adenosina/sangre , Adenosina/química , Humanos , Ligandos , Conformación Molecular , Estructura Molecular , Fenetilaminas/sangre
14.
J Med Chem ; 51(7): 2088-99, 2008 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-18321038

RESUMEN

The binding modes at the A 2B adenosine receptor (AR) of 72 derivatives of adenosine and its 5'- N-methyluronamide with diverse substitutions at the 2 and N (6) positions were studied using a molecular modeling approach. The compounds in their receptor-docked conformations were used to build CoMFA and CoMSIA quantitative structure-activity relationship models. Various parameters, including different types of atomic charges, were examined. The best statistical parameters were obtained with a joint CoMFA and CoMSIA model: R (2) = 0.960, Q (2) = 0.676, SEE = 0.175, F = 158, and R (2) test = 0.782 for an independent test set containing 18 compounds. On the basis of the modeling results, four novel adenosine analogues, having elongated or bulky substitutions at N (6) position and/or 2 position, were synthesized and evaluated biologically. All of the proposed compounds were potent, full agonists at the A 2B AR in adenylate cyclase studies. Thus, in support of the modeling, bulky substitutions at both positions did not prevent A 2B AR activation, which predicts separate regions for docking of these moieties.


Asunto(s)
Agonistas del Receptor de Adenosina A2 , Adenosina/análogos & derivados , Adenosina/farmacología , Simulación por Computador , Diseño de Fármacos , Relación Estructura-Actividad Cuantitativa , Adenosina/química , Sitios de Unión , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformación Molecular , Receptor de Adenosina A2B/química , Electricidad Estática , Estereoisomerismo
15.
Biochemistry ; 46(25): 7437-48, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17542617

RESUMEN

His272 (7.43) in the seventh transmembrane domain (TM7) of the human A3 adenosine receptor (AR) interacts with the 3' position of nucleosides, based on selective affinity enhancement at a H272E mutant A3 AR (neoceptor) of 3'-ureido, but not 3'-OH, adenosine analogues. Here, mutation of the analogous H278 of the human A1 AR to Ala, Asp, Glu, or Leu enhanced the affinity of novel 2'- and 3'-ureido adenosine analogues, such as 10 (N6-cyclopentyl-3'-ureido-3'-deoxyadenosine), by >100-fold, while decreasing the affinity or potency of adenosine and other 3'-OH adenosine analogues. His278 mutant receptors produced a similar enhancement regardless of the charge character of the substituted residue, implicating steric rather than electrostatic factors in the gain of function, a hypothesis supported by rhodopsin-based molecular modeling. It was also demonstrated that this interaction was orientationally specific; i.e., mutations at the neighboring Thr277 did not enhance the affinity for a series of 2'- and 3'-ureido nucleosides. Additionally, H-bonding groups placed on substituents at the N6 or 5' position demonstrated no enhancement in the mutant receptors. These reengineered human A1 ARs revealed orthogonality similar to that of the A3 but not the A2A AR, in which mutation of the corresponding residue, His278, to Asp did not enhance nucleoside affinity. Functionally, the H278D A1 AR was detectable only in a measure of membrane potential and not in calcium mobilization. This neoceptor approach should be useful for the validation of molecular modeling and the dissection of promiscuous GPCR signaling.


Asunto(s)
Ingeniería Genética , Nucleósidos/química , Nucleósidos/farmacología , Receptor de Adenosina A1/metabolismo , Agonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A1 , Animales , Sitios de Unión , Células CHO , Calcio/análisis , Línea Celular , Cricetinae , Cricetulus , AMP Cíclico/biosíntesis , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Potenciales de la Membrana , Modelos Moleculares , Estructura Molecular , Mutación , Nucleósidos/síntesis química , Estructura Secundaria de Proteína , Ensayo de Unión Radioligante , Receptor de Adenosina A1/química , Receptor de Adenosina A1/genética , Relación Estructura-Actividad , Transfección
16.
J Med Chem ; 50(8): 1810-27, 2007 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-17378544

RESUMEN

2, N6, and 5'-substituted adenosine derivatives were synthesized via alkylation of 2-oxypurine nucleosides leading to 2-arylalkylether derivatives. 2-(3-(Indolyl)ethyloxy)adenosine 17 was examined in both binding and cAMP assays and found to be a potent agonist of the human A2BAR. Simplification, altered connectivity, and mimicking of the indole ring of 17 failed to maintain A2BAR potency. Introduction of N6-ethyl or N6-guanidino substitution, shown to favor A2BAR potency, failed to enhance potency in the 2-(3-(indolyl)ethyloxy)adenosine series. Indole 5' '- or 6' '-halo substitution was favored at the A2BAR, but a 5'-N-ethylcarboxyamide did not further enhance potency. 2-(3' '-(6' '-Bromoindolyl)ethyloxy)adenosine 28 displayed an A2BAR EC50 value of 128 nM, that is, more potent than the parent 17 (299 nM) and similar to 5'-N-ethylcarboxamidoadenosine (140 nM). Compound 28 was a full agonist at A2B and A2AARs and a low efficacy partial agonist at A1 and A3ARs. Thus, we have identified and optimized 2-(2-arylethyl)oxo moieties in AR agonists that enhance A2BAR potency and selectivity.


Asunto(s)
Agonistas del Receptor de Adenosina A2 , Adenosina/análogos & derivados , Adenosina/síntesis química , Adenosina/farmacología , Animales , Sitios de Unión , Unión Competitiva , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/biosíntesis , Humanos , Modelos Moleculares , Estereoisomerismo , Relación Estructura-Actividad
17.
J Med Chem ; 50(6): 1166-76, 2007 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-17302398

RESUMEN

A rhodopsin-based homology model of the nucleotide-activated human P2Y2 receptor, including loops, termini, and phospholipids, was optimized with the Monte Carlo multiple minimum conformational search routine. Docked uridine 5'-triphosphate (UTP) formed a nucleobase pi-pi complex with conserved Phe3.32. Selectivity-enhancing 2'-amino-2'-deoxy substitution interacted through pi-hydrogen-bonding with aromatic Phe6.51 and Tyr3.33. A "sequential ligand composition" approach for docking the flexible dinucleotide agonist Up4U demonstrated a shift of conserved cationic Arg3.29 from the UTP gamma position to the delta position of Up4U and Up4 ribose. Synthesized nucleotides were tested as agonists at human P2Y receptors expressed in 1321N1 astrocytoma cells. 2'-Amino and 2-thio modifications were synergized to enhance potency and selectivity; compound 8 (EC50 = 8 nM) was 300-fold P2Y2-selective versus P2Y4. 2'-Amine acetylation reduced potency, and trifluoroacetylation produced intermediate potency. 5-Amino nucleobase substitution did not enhance P2Y2 potency through a predicted hydrophilic interaction possibly because of destabilization of the receptor-favored Northern conformation of ribose. This detailed view of P2Y2 receptor recognition suggests mutations for model validation.


Asunto(s)
Modelos Moleculares , Agonistas del Receptor Purinérgico P2 , Receptores Purinérgicos P2/química , Uridina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Sitios de Unión , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Método de Montecarlo , Conformación Proteica , Relación Estructura-Actividad Cuantitativa , Receptores Purinérgicos P2Y2 , Uridina Trifosfato/síntesis química , Uridina Trifosfato/química , Uridina Trifosfato/farmacología
18.
J Mol Graph Model ; 25(5): 740-54, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17095272

RESUMEN

Molecular models of all known subtypes (A1, A2A, A2B, and A3) of the human adenosine receptors were built in homology with bovine rhodopsin. These models include the transmembrane domain as well as all extracellular and intracellular hydrophilic loops and terminal domains. The molecular docking of adenosine and 46 selected derivatives was performed for each receptor subtype. A binding mode common for all studied agonists was proposed, and possible explanations for differences in the ligand activities were suggested.


Asunto(s)
Agonistas del Receptor Purinérgico P1 , Receptores Purinérgicos P1/química , Adenosina/análogos & derivados , Adenosina/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Bovinos , Gráficos por Computador , Simulación por Computador , Humanos , Técnicas In Vitro , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Receptores Purinérgicos P1/clasificación , Receptores Purinérgicos P1/genética , Rodopsina/química , Rodopsina/genética , Homología de Secuencia de Aminoácido
19.
Biochem Pharmacol ; 71(4): 540-9, 2006 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-16359641

RESUMEN

With the long-term goal of developing receptor subtype-selective high affinity agonists for the uracil nucleotide-activated P2Y receptors we have carried out a series of structure activity and molecular modeling studies of the human P2Y2 and P2Y4 receptors. UTP analogues with substitutions in the 2'-position of the ribose moiety retained capacity to activate both P2Y2 and P2Y4 receptors. Certain of these analogues were equieffective for activation of both receptors whereas 2'-amino-2'-deoxy-UTP exhibited higher potency for the P2Y2 receptor and 2'-azido-UTP exhibited higher potency for the P2Y4 receptor. 4-Thio substitution of the uracil base resulted in a UTP analogue with increased potency relative to UTP for activation of both the P2Y2 and P2Y4 receptors. In contrast, 2-thio substitution and halo- or alkyl substitution in the 5-position of the uracil base resulted in molecules that were 3-30-fold more potent at the P2Y2 receptor than P2Y4 receptor. 6-Aza-UTP was a P2Y2 receptor agonist that exhibited no activity at the P2Y4 receptor. Stereoisomers of UTPalphaS and 2'-deoxy-UTPalphaS were more potent at the P2Y2 than P2Y4 receptor, and the R-configuration was favored at both receptors. Molecular docking studies revealed that the binding mode of UTP is similar for both the P2Y2 and P2Y4 receptor binding pockets with the most prominent dissimilarities of the two receptors located in the second transmembrane domain (V90 in the P2Y2 receptor and I92 in the P2Y4 receptor) and the second extracellular loop (T182 in the P2Y2 receptor and L184 in the P2Y4 receptor). In summary, this work reveals substitutions in UTP that differentially affect agonist activity at P2Y2 versus P2Y4 receptors and in combination with molecular modeling studies should lead to chemical synthesis of new receptor subtype-selective drugs.


Asunto(s)
Modelos Moleculares , Pentosafosfatos/farmacología , Agonistas del Receptor Purinérgico P2 , Relación Estructura-Actividad , Uridina Trifosfato/farmacología , Unión Competitiva , Humanos , Conformación Molecular , Pentosafosfatos/síntesis química , Pentosafosfatos/química , Receptores Purinérgicos P2 , Receptores Purinérgicos P2Y2 , Estereoisomerismo , Células Tumorales Cultivadas , Uridina Trifosfato/síntesis química , Uridina Trifosfato/química
20.
J Med Chem ; 48(22): 6813-20, 2005 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16250640

RESUMEN

A molecular model of the human A(2B) adenosine receptor containing seven transmembrane alpha helices connected by three intracellular and three extracellular hydrophilic loops had been constructed. A molecular docking of seven structurally diverse xanthine antagonists of the A(2B) receptor was performed, and the differences in their binding modes were investigated. The 1 ns molecular dynamics (MD) simulations of several obtained ligand-receptor complexes inserted into the phospholipid bilayer were carried out. The conformational changes of the A(2B) receptor occurring during MD simulations were explored, and the stable binding modes of the studied antagonists were determined. According to the models presented in this work, the involvement of the His251, Asn282, Ser92, Thr89, and some aromatic residues in ligand recognition was determined. The obtained binding modes of the A(2B) antagonists demonstrate good agreement with the site-directed mutagenesis data.


Asunto(s)
Antagonistas del Receptor de Adenosina A2 , Modelos Moleculares , Receptor de Adenosina A2B/química , Secuencia de Aminoácidos , Sitios de Unión , Simulación por Computador , Humanos , Membrana Dobles de Lípidos/química , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosfolípidos/química , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA