Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Eur J Med Chem ; 225: 113779, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34418785

RESUMEN

Starting from six potential hits identified in a virtual screening campaign directed to a cryptic pocket of BACE-1, at the edge of the catalytic cleft, we have synthesized and evaluated six hybrid compounds, designed to simultaneously reach BACE-1 secondary and catalytic sites and to exert additional activities of interest for Alzheimer's disease (AD). We have identified a lead compound with potent in vitro activity towards human BACE-1 and cholinesterases, moderate Aß42 and tau antiaggregating activity, and brain permeability, which is nontoxic in neuronal cells and zebrafish embryos at concentrations above those required for the in vitro activities. This compound completely restored short- and long-term memory in a mouse model of AD (SAMP8) relative to healthy control strain SAMR1, shifted APP processing towards the non-amyloidogenic pathway, reduced tau phosphorylation, and increased the levels of synaptic proteins PSD95 and synaptophysin, thereby emerging as a promising disease-modifying, cognition-enhancing anti-AD lead.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Aminoquinolinas/farmacología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/metabolismo , Aminoquinolinas/síntesis química , Aminoquinolinas/química , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Compuestos Heterocíclicos de 4 o más Anillos/síntesis química , Compuestos Heterocíclicos de 4 o más Anillos/química , Humanos , Simulación de Dinámica Molecular , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Proteínas tau/antagonistas & inhibidores , Proteínas tau/metabolismo
2.
Eur J Pharmacol ; 906: 174276, 2021 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-34174267

RESUMEN

Treatment of Parkinson's disease (PD) includes the use of monoamine oxidase-B (MAO-B) inhibitor drugs. In this work we have evaluated the possible gamma-decanolactone (GD) effect in vitro to inhibit the A and B isoforms of human monoamine oxidase (hMAO) enzyme and their citotoxicity in human hepatoma cell line (HepG2). Also, binding studies to A1, A2A A2B and A3 adenosine receptors were performed. A docking study of gamma-decanolactone has been carried out with the molecular targets of MAO-A and MAO-B isoforms. The physicochemical properties and ability to cross physiological barriers, as the blood brain barrier (BBB), was elucidated by computational studies. The in vivo assays, the rota-rod test, body temperature assessment and open field test were performed in reserpinized mice (1.5 mg/kg, i.p.; 18:00 before) to evaluate the effect of gamma-decanolactone (300 mg/kg), alone or associated with Levodopa plus Benserazide (LD + BZ, 100:25 mg/kg, i.p.). Gamma-decanolactone inhibited preferentially the MAO-B in a reversible manner, with an inhibitory concentration of 50% (IC50) 55.95 ± 9.06 µM. It was shown to be a safe drug since only at the highest concentration decreased the viability of HepG2 cells. It also does not bind to adenosine receptors investigated in this study. The molecular docking study show that the gamma-decanolactone ligand adopts a relatively compact conformation in the active site of hMAO-B, while we note an extended conformation of gamma-decanolactone ligand in the hMAO-A isoform. The physicochemical properties obtained, and the theoretical models utilized for the evaluation of ability to cross the BBB, predict a good gamma-decanolactone bioavailability and access to the central nervous system (CNS). In the in vivo studies, gamma-decanolactone partially reversed the ataxia of the reserpinized mice at 01:00 h and 01:30 h post-administration. Concomitant treatment of gamma-decanolactone with LD + BZ, at 01:30 h showed a potentiation of the reversibility of ataxia and facilitated the reversal of hypothermia caused by reserpine for all measured times (P <0.01 vs vehicle), except at 24:00 h, but not reversed the hypokinesia in the open field test. In summary, the results herein obtained and in conjunction with previous studies, suggest that gamma-decanolactone could be a drug with potential utility as antiparkinsonian drug.


Asunto(s)
Antiparkinsonianos/farmacología , Lactonas/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Antiparkinsonianos/química , Antiparkinsonianos/uso terapéutico , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Pruebas de Enzimas , Células Hep G2 , Humanos , Lactonas/uso terapéutico , Masculino , Ratones , Simulación del Acoplamiento Molecular , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/química , Inhibidores de la Monoaminooxidasa/uso terapéutico , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/patología , Permeabilidad , Receptores Purinérgicos P1/metabolismo , Proteínas Recombinantes/metabolismo , Reserpina/administración & dosificación , Reserpina/metabolismo , Reserpina/toxicidad , Relación Estructura-Actividad
3.
Angew Chem Int Ed Engl ; 60(33): 18022-18030, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-33904641

RESUMEN

Many diseases are polygenic and can only be treated efficiently with drugs that modulate multiple targets. However, rational design of compounds with multi-target profiles is rarely pursued because it is considered too difficult, in particular if the drug must enter the central nervous system. Here, a structure-based strategy to identify dual-target ligands of G-protein-coupled receptors is presented. We use this approach to design compounds that both antagonize the A2A adenosine receptor and activate the D2 dopamine receptor, which have excellent potential as antiparkinson drugs. Atomic resolution models of the receptors guided generation of a chemical library with compounds designed to occupy orthosteric and secondary binding pockets in both targets. Structure-based virtual screens identified ten compounds, of which three had affinity for both targets. One of these scaffolds was optimized to nanomolar dual-target activity and showed the predicted pharmacodynamic effect in a rat model of Parkinsonism.


Asunto(s)
Antiparkinsonianos/farmacología , Diseño de Fármacos , Receptor de Adenosina A2A/metabolismo , Receptores de Dopamina D2/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antiparkinsonianos/síntesis química , Antiparkinsonianos/química , Evaluación Preclínica de Medicamentos , Humanos , Ligandos , Estructura Molecular , Ratas , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química
4.
PLoS One ; 16(3): e0248139, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33690613

RESUMEN

This work tries to help overcome the lack of relevant translational screening assays, as a limitation for the identification of novel analgesics for neuropathic pain. Hyperexcitability and neurite shortening are common adverse effects of antiviral and antitumor drugs, leading to neuropathic pain. Now, as seen in the drug screening that we developed here, a high-content microscopy-based assay with immortalized dorsal root ganglia (DRG) neurons (differentiated F11 cells) allowed to identify drugs able to protect against the iatrogenic neurite shortening induced by the antitumor drug vincristine and the antiviral drug rilpivirine. We observed that vincristine and rilpivirine induced a significant reduction in the neurite length, which was reverted by α-lipoic acid. We had also evidenced protective effects of pregabalin and melatonin, acting through the α2δ-2 subunit of the voltage-dependent calcium channels and the MT1 receptor, respectively. Additionally, two hits originated from a previous primary screening aimed to detect inhibitors of hyperexcitability to inflammatory mediators in DRG neurons (nitrendipine and felodipine) also prevented neurite shortening in our model. In summary, in this work we developed a novel secondary assay for identifying hits with neuroprotective effect against iatrogenic neurite shortening, consistent with the anti-hyperexcitability action previously tested: highlighting nitrendipine and felodipine against iatrogenic damage in DRG neurons.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Neuritas/efectos de los fármacos , Analgésicos/farmacología , Línea Celular , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Humanos , Enfermedad Iatrogénica , Melatonina/farmacología , Neuralgia/tratamiento farmacológico , Neuritas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Pregabalina/farmacología , Rilpivirina/efectos adversos , Rilpivirina/farmacología , Ácido Tióctico/farmacología , Vincristina/efectos adversos , Vincristina/farmacología
5.
Biomed Pharmacother ; 121: 109601, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31739159

RESUMEN

BACKGROUND: Phosphoenolpyruvate carboxykinase (PEPCK) catalyzes the decarboxylation of oxaloacetate to phosphoenolpyruvate. The mitochondrial isozyme, PEPCK-M is highly expressed in cancer cells, where it plays a role in nutrient stress response. To date, pharmacological strategies to target this pathway have not been pursued. METHODS: A compound embodying a 3-alkyl-1,8-dibenzylxanthine nucleus (iPEPCK-2), was synthesized and successfully probed in silico on a PEPCK-M structural model. Potency and target engagement in vitro and in vivo were evaluated by kinetic and cellular thermal shift assays (CETSA). The compound and its target were validated in tumor growth models in vitro and in murine xenografts. RESULTS: Cross-inhibitory capacity and increased potency as compared to 3-MPA were confirmed in vitro and in vivo. Treatment with iPEPCK-2 inhibited cell growth and survival, especially in poor-nutrient environment, consistent with an impact on colony formation in soft agar. Finally, daily administration of the PEPCK-M inhibitor successfully inhibited tumor growth in two murine xenograft models as compared to vehicle, without weight loss, or any sign of apparent toxicity. CONCLUSION: We conclude that iPEPCK-2 is a compelling anticancer drug targeting PEPCK-M, a hallmark gene product involved in metabolic adaptations of the tumor.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Fosfoenolpiruvato Carboxiquinasa (ATP)/antagonistas & inhibidores , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Animales , Biomarcadores de Tumor/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Células HCT116 , Células HEK293 , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Estructura Secundaria de Proteína , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
Eur J Med Chem ; 182: 111624, 2019 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-31445234

RESUMEN

This work describes the rational discovery of novel chemotypes of p38α MAPK inhibitors using a funnel approach consisting of several computer-aided drug discovery methods and biological experiments. Among the identified hits, four compounds belonging to different chemical families showed IC50 values lower than 10 µM. In particular, the 1,4-benzodioxane derivative 5 turned out to be a potent and efficient p38α MAPK inhibitor having IC50 = 0.07 µM, and LEexp and LipE values of 0.38 and 4.8, respectively; noteworthy, the compound had also a promising kinase selectivity profile and the capability to suppress p38α MAPK effects in human immune cells. Overall, the collected findings highlight that the applied strategy has been successful in generating chemical novelty in the inhibitor kinase field, providing suitable chemical candidates for further inhibitor optimization.


Asunto(s)
Descubrimiento de Drogas , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Voluntarios Sanos , Humanos , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
7.
SLAS Discov ; 24(3): 398-413, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30616481

RESUMEN

Compound screening in biological assays and subsequent optimization of hits is indispensable for the development of new molecular research tools and drug candidates. To facilitate such discoveries, the European Research Infrastructure EU-OPENSCREEN was founded recently with the support of its member countries and the European Commission. Its distributed character harnesses complementary knowledge, expertise, and instrumentation in the discipline of chemical biology from 20 European partners, and its open working model ensures that academia and industry can readily access EU-OPENSCREEN's compound collection, equipment, and generated data. To demonstrate the power of this collaborative approach, this perspective article highlights recent projects from EU-OPENSCREEN partner institutions. These studies yielded (1) 2-aminoquinazolin-4(3 H)-ones as potential lead structures for new antimalarial drugs, (2) a novel lipodepsipeptide specifically inducing apoptosis in cells deficient for the pVHL tumor suppressor, (3) small-molecule-based ROCK inhibitors that induce definitive endoderm formation and can potentially be used for regenerative medicine, (4) potential pharmacological chaperones for inborn errors of metabolism and a familiar form of acute myeloid leukemia (AML), and (5) novel tankyrase inhibitors that entered a lead-to-candidate program. Collectively, these findings highlight the benefits of small-molecule screening, the plethora of assay designs, and the close connection between screening and medicinal chemistry within EU-OPENSCREEN.


Asunto(s)
Conducta Cooperativa , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Europa (Continente) , Ensayos Analíticos de Alto Rendimiento , Humanos , Relación Estructura-Actividad
8.
Molecules ; 23(5)2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29748476

RESUMEN

The identification of subtype-selective GPCR (G-protein coupled receptor) ligands is a challenging task. In this study, we developed a computational protocol to find compounds with 5-HT2BR versus 5-HT1BR selectivity. Our approach employs the hierarchical combination of machine learning methods, docking, and multiple scoring methods. First, we applied machine learning tools to filter a large database of druglike compounds by the new Neighbouring Substructures Fingerprint (NSFP). This two-dimensional fingerprint contains information on the connectivity of the substructural features of a compound. Preselected subsets of the database were then subjected to docking calculations. The main indicators of compounds' selectivity were their different interactions with the secondary binding pockets of both target proteins, while binding modes within the orthosteric binding pocket were preserved. The combined methodology of ligand-based and structure-based methods was validated prospectively, resulting in the identification of hits with nanomolar affinity and ten-fold to ten thousand-fold selectivities.


Asunto(s)
Evaluación Preclínica de Medicamentos , Aprendizaje Automático , Receptor de Serotonina 5-HT2B/metabolismo , Sitios de Unión , Humanos , Ligandos , Modelos Moleculares , Serotonina/química , Serotonina/metabolismo
9.
J Med Chem ; 61(12): 5269-5278, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29792714

RESUMEN

Modulation of multiple biological targets with a single drug can lead to synergistic therapeutic effects and has been demonstrated to be essential for efficient treatment of CNS disorders. However, rational design of compounds that interact with several targets is very challenging. Here, we demonstrate that structure-based virtual screening can guide the discovery of multi-target ligands of unrelated proteins relevant for Parkinson's disease. A library with 5.4 million molecules was docked to crystal structures of the A2A adenosine receptor (A2AAR) and monoamine oxidase B (MAO-B). Twenty-four compounds that were among the highest ranked for both binding sites were evaluated experimentally, resulting in the discovery of four dual-target ligands. The most potent compound was an A2AAR antagonist with nanomolar affinity ( Ki = 19 nM) and inhibited MAO-B with an IC50 of 100 nM. Optimization guided by the predicted binding modes led to the identification of a second potent dual-target scaffold. The two discovered scaffolds were shown to counteract 6-hydroxydopamine-induced neurotoxicity in dopaminergic neuronal-like SH-SY5Y cells. Structure-based screening can hence be used to identify ligands with specific polypharmacological profiles, providing new avenues for drug development against complex diseases.


Asunto(s)
Antiparkinsonianos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Simulación del Acoplamiento Molecular/métodos , Monoaminooxidasa/química , Receptor de Adenosina A2A/química , Antagonistas del Receptor de Adenosina A2/química , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Antiparkinsonianos/química , Sitios de Unión , Células CHO , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cricetulus , AMP Cíclico/metabolismo , Humanos , Ligandos , Terapia Molecular Dirigida , Monoaminooxidasa/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Receptor de Adenosina A2A/metabolismo , Relación Estructura-Actividad
10.
PLoS Pathog ; 12(7): e1005763, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27467575

RESUMEN

A major cause of the paucity of new starting points for drug discovery is the lack of interaction between academia and industry. Much of the global resource in biology is present in universities, whereas the focus of medicinal chemistry is still largely within industry. Open source drug discovery, with sharing of information, is clearly a first step towards overcoming this gap. But the interface could especially be bridged through a scale-up of open sharing of physical compounds, which would accelerate the finding of new starting points for drug discovery. The Medicines for Malaria Venture Malaria Box is a collection of over 400 compounds representing families of structures identified in phenotypic screens of pharmaceutical and academic libraries against the Plasmodium falciparum malaria parasite. The set has now been distributed to almost 200 research groups globally in the last two years, with the only stipulation that information from the screens is deposited in the public domain. This paper reports for the first time on 236 screens that have been carried out against the Malaria Box and compares these results with 55 assays that were previously published, in a format that allows a meta-analysis of the combined dataset. The combined biochemical and cellular assays presented here suggest mechanisms of action for 135 (34%) of the compounds active in killing multiple life-cycle stages of the malaria parasite, including asexual blood, liver, gametocyte, gametes and insect ookinete stages. In addition, many compounds demonstrated activity against other pathogens, showing hits in assays with 16 protozoa, 7 helminths, 9 bacterial and mycobacterial species, the dengue fever mosquito vector, and the NCI60 human cancer cell line panel of 60 human tumor cell lines. Toxicological, pharmacokinetic and metabolic properties were collected on all the compounds, assisting in the selection of the most promising candidates for murine proof-of-concept experiments and medicinal chemistry programs. The data for all of these assays are presented and analyzed to show how outstanding leads for many indications can be selected. These results reveal the immense potential for translating the dispersed expertise in biological assays involving human pathogens into drug discovery starting points, by providing open access to new families of molecules, and emphasize how a small additional investment made to help acquire and distribute compounds, and sharing the data, can catalyze drug discovery for dozens of different indications. Another lesson is that when multiple screens from different groups are run on the same library, results can be integrated quickly to select the most valuable starting points for subsequent medicinal chemistry efforts.


Asunto(s)
Antimaláricos/uso terapéutico , Conjuntos de Datos como Asunto , Descubrimiento de Drogas/métodos , Malaria/tratamiento farmacológico , Enfermedades Desatendidas/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Humanos , Bibliotecas de Moléculas Pequeñas
11.
J Biomol Screen ; 21(8): 816-23, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27095818

RESUMEN

G protein-coupled receptors (GPCRs) exist as collections of conformations in equilibrium, and the efficacy of drugs has been proposed to be associated with their absolute and relative affinities for these different conformations. The serotonin 2A (5-HT2A) receptor regulates multiple physiological functions, is involved in the pathophysiology of schizophrenia, and serves as an important target of atypical antipsychotic drugs. This receptor was one of the first GPCRs for which the functional selectivity phenomenon was observed, with its various ligands exerting differential effects on the phospholipase A2 (PLA2) and phospholipase C (PLC) signaling pathways. We aimed to develop a multiplex functional assay in 96-well plates for the simultaneous measurement of the PLA2 and PLC pathways coupled to 5-HT2A receptors; this approach enables the detection of either functional selectivity or cooperativity phenomena in early drug screening stages. The suitability of the method for running screening campaigns was tested using the Prestwick Chemical Library, and 22 confirmed hits with activities of more than 90% were identified; 11 of these hits produced statistically significant differences between the two effector pathways. Thus, we have developed a miniaturized multiplex assay in 96-well plates to measure functional selectivity for 5-HT2A receptors in the early stages of the drug discovery process.


Asunto(s)
Antipsicóticos/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento/métodos , Receptores de Serotonina 5-HT2/metabolismo , Esquizofrenia/tratamiento farmacológico , Antagonistas del Receptor de Serotonina 5-HT2/aislamiento & purificación , Antipsicóticos/uso terapéutico , Sitios de Unión , Evaluación Preclínica de Medicamentos , Humanos , Ligandos , Fosfolipasas A2/metabolismo , Esquizofrenia/metabolismo , Antagonistas del Receptor de Serotonina 5-HT2/metabolismo , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo
12.
J Chem Inf Model ; 55(1): 135-48, 2015 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-25483751

RESUMEN

Alzheimer's disease is a neurodegenerative pathology with unmet clinical needs. A highly desirable approach to this syndrome would be to find a single lead that could bind to some or all of the selected biomolecules that participate in the amyloid cascade, the most accepted route for Alzheimer disease genesis. In order to circumvent the challenge posed by the sizable differences in the binding sites of the molecular targets, we propose a computer-assisted protocol based on a pharmacophore and a set of required interactions with the targets that allows for the automated screening of candidates. We used a combination of docking and molecular dynamics protocols in order to discard nonbinders, optimize the best candidates, and provide a rationale for their potential as inhibitors. To provide a proof of concept, we proceeded to screen the literature and databases, a task that allowed us to identify a set of carbazole-containing compounds that initially showed affinity only for the cholinergic targets in our experimental assays. Two cycles of design based on our protocol led to a new set of analogues that were synthesized and assayed. The assay results revealed that the designed inhibitors had improved affinities for BACE-1 by more than 3 orders of magnitude and also displayed amyloid aggregation inhibition and affinity for AChE and BuChE, a result that led us to a group of multitarget amyloid cascade inhibitors that also could have a positive effect at the cholinergic level.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Diseño Asistido por Computadora , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Secretasas de la Proteína Precursora del Amiloide/química , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/química , Sitios de Unión , Carbazoles/química , Carbazoles/farmacología , Técnicas de Química Sintética , Humanos , Indoles/química , Indoles/farmacología , Ligandos , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo
13.
PLoS One ; 9(12): e115460, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25522365

RESUMEN

Integration of open access, curated, high-quality information from multiple disciplines in the Life and Biomedical Sciences provides a holistic understanding of the domain. Additionally, the effective linking of diverse data sources can unearth hidden relationships and guide potential research strategies. However, given the lack of consistency between descriptors and identifiers used in different resources and the absence of a simple mechanism to link them, gathering and combining relevant, comprehensive information from diverse databases remains a challenge. The Open Pharmacological Concepts Triple Store (Open PHACTS) is an Innovative Medicines Initiative project that uses semantic web technology approaches to enable scientists to easily access and process data from multiple sources to solve real-world drug discovery problems. The project draws together sources of publicly-available pharmacological, physicochemical and biomolecular data, represents it in a stable infrastructure and provides well-defined information exploration and retrieval methods. Here, we highlight the utility of this platform in conjunction with workflow tools to solve pharmacological research questions that require interoperability between target, compound, and pathway data. Use cases presented herein cover 1) the comprehensive identification of chemical matter for a dopamine receptor drug discovery program 2) the identification of compounds active against all targets in the Epidermal growth factor receptor (ErbB) signaling pathway that have a relevance to disease and 3) the evaluation of established targets in the Vitamin D metabolism pathway to aid novel Vitamin D analogue design. The example workflows presented illustrate how the Open PHACTS Discovery Platform can be used to exploit existing knowledge and generate new hypotheses in the process of drug discovery.


Asunto(s)
Bases de Datos como Asunto , Descubrimiento de Drogas/organización & administración , Programas Informáticos , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/estadística & datos numéricos
14.
J Med Chem ; 57(20): 8590-607, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25264825

RESUMEN

A forward chemical genetic approach was followed to discover new targets and lead compounds for Parkinson's disease (PD) treatment. By analysis of the cell protection produced by some small molecules, a diphenyl sulfide compound was revealed to be a new phosphodiesterase 7 (PDE7) inhibitor and identified as a new hit. This result allows us to confirm the utility of PDE7 inhibitors as a potential pharmacological treatment of PD. On the basis of these data, a diverse family of diphenyl sulfides has been developed and pharmacologically evaluated in the present work. Moreover, to gain insight into the safety of PDE7 inhibitors for human chronic treatment, we evaluated the new compounds in a surrogate emesis model, showing nonemetic effects.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Anestesia/efectos adversos , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Química Sintética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/química , Evaluación Preclínica de Medicamentos/métodos , Humanos , Concentración 50 Inhibidora , Masculino , Ratones Endogámicos , Modelos Moleculares , Inhibidores de Fosfodiesterasa/síntesis química , Ratas , Relación Estructura-Actividad , Sulfuros/química , Sulfuros/farmacología , Vómitos/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA