Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nutrients ; 16(12)2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38931171

RESUMEN

Taurine, a non-proteogenic amino acid and commonly used nutritional supplement, can protect various tissues from degeneration associated with the action of the DNA-damaging chemotherapeutic agent cisplatin. Whether and how taurine protects human ovarian cancer (OC) cells from DNA damage caused by cisplatin is not well understood. We found that OC ascites-derived cells contained significantly more intracellular taurine than cell culture-modeled OC. In culture, elevation of intracellular taurine concentration to OC ascites-cell-associated levels suppressed proliferation of various OC cell lines and patient-derived organoids, reduced glycolysis, and induced cell protection from cisplatin. Taurine cell protection was associated with decreased DNA damage in response to cisplatin. A combination of RNA sequencing, reverse-phase protein arrays, live-cell microscopy, flow cytometry, and biochemical validation experiments provided evidence for taurine-mediated induction of mutant or wild-type p53 binding to DNA, activation of p53 effectors involved in negative regulation of the cell cycle (p21), and glycolysis (TIGAR). Paradoxically, taurine's suppression of cell proliferation was associated with activation of pro-mitogenic signal transduction including ERK, mTOR, and increased mRNA expression of major DNA damage-sensing molecules such as DNAPK, ATM and ATR. While inhibition of ERK or p53 did not interfere with taurine's ability to protect cells from cisplatin, suppression of mTOR with Torin2, a clinically relevant inhibitor that also targets DNAPK and ATM/ATR, broke taurine's cell protection. Our studies implicate that elevation of intracellular taurine could suppress cell growth and metabolism, and activate cell protective mechanisms involving mTOR and DNA damage-sensing signal transducti.


Asunto(s)
Cisplatino , Daño del ADN , Neoplasias Ováricas , Serina-Treonina Quinasas TOR , Taurina , Proteína p53 Supresora de Tumor , Taurina/farmacología , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Femenino , Neoplasias Ováricas/metabolismo , Daño del ADN/efectos de los fármacos , Cisplatino/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Glucólisis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Antineoplásicos/farmacología
3.
Stem Cell Reports ; 19(2): 299-313, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38215755

RESUMEN

Successful allogeneic human pluripotent stem cell (hPSC)-derived therapies must overcome immunological rejection by the recipient. To build reagents to define these barriers, we genetically ablated ß2M, TAP1, CIITA, CD74, MICA, and MICB to limit expression of HLA-I, HLA-II, and natural killer (NK) cell activating ligands in hPSCs. Transplantation of these cells that also expressed covalent single chain trimers of Qa1 and H2-Kb to inhibit NK cells and CD55, Crry, and CD59 to inhibit complement deposition led to persistent teratomas in wild-type mice. Transplantation of HLA-deficient hPSCs into mice genetically deficient in complement and depleted of NK cells also led to persistent teratomas. Thus, T cell, NK cell, and complement evasion are necessary to prevent immunological rejection of hPSCs and their progeny. These cells and versions expressing human orthologs of immune evasion factors can be used to define cell type-specific immune barriers and conduct preclinical testing in immunocompetent mouse models.


Asunto(s)
Células Madre Pluripotentes , Teratoma , Humanos , Animales , Ratones , Células Asesinas Naturales , Línea Celular , Linfocitos T , Proteínas del Sistema Complemento
4.
bioRxiv ; 2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37425790

RESUMEN

Allogeneic human pluripotent stem cell (hPSC)-derived cells and tissues for therapeutic transplantation must necessarily overcome immunological rejection by the recipient. To define these barriers and to create cells capable of evading rejection for preclinical testing in immunocompetent mouse models, we genetically ablated ß2m, Tap1, Ciita, Cd74, Mica, and Micb to limit expression of HLA-I, HLA-II, and natural killer cell activating ligands in hPSCs. Though these and even unedited hPSCs readily formed teratomas in cord blood-humanized immunodeficient mice, grafts were rapidly rejected by immunocompetent wild-type mice. Transplantation of these cells that also expressed covalent single chain trimers of Qa1 and H2-Kb to inhibit natural killer cells and CD55, Crry, and CD59 to inhibit complement deposition led to persistent teratomas in wild-type mice. Expression of additional inhibitory factors such as CD24, CD47, and/or PD-L1 had no discernible impact on teratoma growth or persistence. Transplantation of HLA-deficient hPSCs into mice genetically deficient in complement and depleted of natural killer cells also led to persistent teratomas. Thus, T cell, NK cell, and complement evasion are necessary to prevent immunological rejection of hPSCs and their progeny. These cells and versions expressing human orthologs of immune evasion factors can be used to refine tissue- and cell type-specific immune barriers, and to conduct preclinical testing in immunocompetent mouse models.

5.
bioRxiv ; 2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-36909636

RESUMEN

Loss of treatment-induced ovarian carcinoma (OC) growth suppression poses a major clinical challenge because it leads to disease recurrence. Therefore, there is a compelling need for well- -tolerated approaches that can support tumor growth-suppression after therapy is stopped. We have profiled ascites as OC tumor microenvironments to search for potential non-toxic soluble components that would activate tumor suppressor pathways in OC cells. Our investigations revealed that low levels of taurine, a non-proteogenic sulfonic amino acid, were present within OC ascites. Taurine supplementation, beyond levels found in ascites, induced growth suppression without causing cytotoxicity in various OC cells, including chemotherapy-resistant cell clones and patient-derived organoids representing primary or chemotherapy recovered disease. Inhibition of proliferation by taurine was linked to increased mutant or wild-type p53 proteins binding to DNA, induction of p21, and independently of p53, TIGAR expression. Taurine-induced activation of p21 and TIGAR was associated with suppression of cell-cycle progression, glycolysis, and mitochondrial respiration. Expression of p21 or TIGAR in OC cells mimicked taurine-induced growth suppression. Our studies support the potential therapeutic value of taurine supplementation in OC.

6.
Nat Commun ; 13(1): 1590, 2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-35338121

RESUMEN

Drug discovery for diseases such as Parkinson's disease are impeded by the lack of screenable cellular phenotypes. We present an unbiased phenotypic profiling platform that combines automated cell culture, high-content imaging, Cell Painting, and deep learning. We applied this platform to primary fibroblasts from 91 Parkinson's disease patients and matched healthy controls, creating the largest publicly available Cell Painting image dataset to date at 48 terabytes. We use fixed weights from a convolutional deep neural network trained on ImageNet to generate deep embeddings from each image and train machine learning models to detect morphological disease phenotypes. Our platform's robustness and sensitivity allow the detection of individual-specific variation with high fidelity across batches and plate layouts. Lastly, our models confidently separate LRRK2 and sporadic Parkinson's disease lines from healthy controls (receiver operating characteristic area under curve 0.79 (0.08 standard deviation)), supporting the capacity of this platform for complex disease modeling and drug screening applications.


Asunto(s)
Aprendizaje Profundo , Enfermedad de Parkinson , Fibroblastos , Humanos , Aprendizaje Automático , Redes Neurales de la Computación
7.
J Med Chem ; 63(5): 2411-2425, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32101422

RESUMEN

The measurement of receptor occupancy (RO) using positron emission tomography (PET) has been instrumental in guiding discovery and development of CNS directed therapeutics. We and others have investigated muscarinic acetylcholine receptor 4 (M4) positive allosteric modulators (PAMs) for the treatment of symptoms associated with neuropsychiatric disorders. In this article, we describe the synthesis, in vitro, and in vivo characterization of a series of central pyridine-related M4 PAMs that can be conveniently radiolabeled with carbon-11 as PET tracers for the in vivo imaging of an allosteric binding site of the M4 receptor. We first demonstrated its feasibility by mapping the receptor distribution in mouse brain and confirming that a lead molecule 1 binds selectively to the receptor only in the presence of the orthosteric agonist carbachol. Through a competitive binding affinity assay and a number of physiochemical properties filters, several related compounds were identified as candidates for in vivo evaluation. These candidates were then radiolabeled with 11C and studied in vivo in rhesus monkeys. This research eventually led to the discovery of the clinical radiotracer candidate [11C]MK-6884.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Piridinas/farmacología , Receptor Muscarínico M4/agonistas , Animales , Células CHO , Radioisótopos de Carbono/química , Radioisótopos de Carbono/farmacología , Cricetulus , Humanos , Macaca mulatta , Agonistas Muscarínicos/química , Enfermedades Neurodegenerativas/diagnóstico por imagen , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Tomografía de Emisión de Positrones , Piridinas/química , Receptor Muscarínico M4/metabolismo
8.
Anal Chem ; 88(3): 1864-70, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26727249

RESUMEN

The process of bringing a drug to market involves many steps, including the preclinical stage, where various properties of the drug candidate molecule are determined. These properties, which include drug absorption, distribution, metabolism, and excretion, are often displayed in a pharmacokinetic (PK) profile. While PK profiles are determined in animal models, in vitro systems that model in vivo processes are available, although each possesses shortcomings. Here, we present a 3D-printed, diffusion-based, and dynamic in vitro PK device. The device contains six flow channels, each with integrated porous membrane-based insert wells. The pores of these membranes enable drugs to freely diffuse back and forth between the flow channels and the inserts, thus enabling both loading and clearance portions of a standard PK curve to be generated. The device is designed to work with 96-well plate technology and consumes single-digit milliliter volumes to generate multiple PK profiles, simultaneously. Generation of PK profiles by use of the device was initially performed with fluorescein as a test molecule. Effects of such parameters as flow rate, loading time, volume in the insert well, and initial concentration of the test molecule were investigated. A prediction model was generated from this data, enabling the user to predict the concentration of the test molecule at any point along the PK profile within a coefficient of variation of ∼ 5%. Depletion of the analyte from the well was characterized and was determined to follow first-order rate kinetics, indicated by statistically equivalent (p > 0.05) depletion half-lives that were independent of the starting concentration. A PK curve for an approved antibiotic, levofloxacin, was generated to show utility beyond the fluorescein test molecule.


Asunto(s)
Difusión , Evaluación Preclínica de Medicamentos/instrumentación , Levofloxacino/farmacocinética , Técnicas Analíticas Microfluídicas , Impresión Tridimensional , Animales , Antibacterianos/farmacocinética , Cinética , Técnicas Analíticas Microfluídicas/instrumentación , Modelos Animales , Impresión Tridimensional/instrumentación
9.
J Biomol Screen ; 19(1): 108-18, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23989454

RESUMEN

The health and disease-related biology of the CXCR4 chemokine receptor presents the challenge of finding a small molecule that can bind CXCR4 and block T-cell tropic human immunodeficiency virus type 1 (HIV-1) cell entry, while preserving the ability of CXCR4 to respond to its native ligand, CXCL12. HIV entry into the host cell involves the interaction of the viral envelope glycoprotein gp120 binding to CD4, followed by a rearrangement in gp120, and subsequent interaction with the chemokine receptor CXCR4 or CCR5. These initial events can be re-created in a cell fusion assay that represents a surrogate system, mimicking the early stages of viral entry via these host cell receptors. In the current study, a T-tropic HIV cell fusion assay was established using U2OS cells expressing the envelope glycoprotein gp160 from the T-tropic HIV NL4-3 and HeLa cells expressing CD4 and CXCR4. Detection of the cell fusion event was based on a Gal4/VP16-activated ß-lactamase signal and was measured by automated microscopy or laser scanning plate cytometry. Changes in morphology associated with cell fusion were combined with ß-lactamase activity to generate results with robust assay statistics in both 384-well and 1536-well plates. Compounds were subsequently characterized by CXCR4 signaling assays to eliminate functional antagonists and allow the identification of a function-sparing HIV entry inhibitor.


Asunto(s)
Inhibidores de Fusión de VIH/farmacología , Pruebas de Sensibilidad Microbiana/métodos , Fenotipo , Receptores CXCR4/metabolismo , Internalización del Virus/efectos de los fármacos , Línea Celular , Expresión Génica , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Humanos , Receptores CXCR4/genética , Proteínas Recombinantes de Fusión , Reproducibilidad de los Resultados
10.
ACS Chem Biol ; 8(11): 2442-51, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23957438

RESUMEN

Modern medicine is founded on the discovery of penicillin and subsequent small molecules that inhibit bacterial peptidoglycan (PG) and cell wall synthesis. However, the discovery of new chemically and mechanistically distinct classes of PG inhibitors has become exceedingly rare, prompting speculation that intracellular enzymes involved in PG precursor synthesis are not 'druggable' targets. Here, we describe a ß-lactam potentiation screen to identify small molecules that augment the activity of ß-lactams against methicillin-resistant Staphylococcus aureus (MRSA) and mechanistically characterize a compound resulting from this screen, which we have named murgocil. We provide extensive genetic, biochemical, and structural modeling data demonstrating both in vitro and in whole cells that murgocil specifically inhibits the intracellular membrane-associated glycosyltransferase, MurG, which synthesizes the lipid II PG substrate that penicillin binding proteins (PBPs) polymerize and cross-link into the cell wall. Further, we demonstrate that the chemical synergy and cidality achieved between murgocil and the ß-lactam imipenem is mediated through MurG dependent localization of PBP2 to the division septum. Collectively, these data validate our approach to rationally identify new target-specific bioactive ß-lactam potentiation agents and demonstrate that murgocil now serves as a highly selective and potent chemical probe to assist our understanding of PG biosynthesis and cell wall biogenesis across Staphylococcal species.


Asunto(s)
Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , Peptidoglicano Glicosiltransferasa/metabolismo , Pirazoles/farmacología , Staphylococcus aureus/efectos de los fármacos , Esteroles/farmacología , Simulación por Computador , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/farmacología , Humanos , Microscopía Fluorescente , Modelos Moleculares , Pirazoles/química , Staphylococcus aureus/enzimología , Esteroles/química
11.
Cancer Chemother Pharmacol ; 68(4): 923-33, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21298383

RESUMEN

PURPOSE: Aurora kinases are required for orderly progression of cells through mitosis, and inhibition of these kinases by siRNA or small molecule inhibitors results in cell death. We previously reported the synthesis of SCH 1473759, a novel sub-nanomolar Aurora A/B inhibitor. METHODS: We utilized SCH 1473759 and a panel of tumor cell lines and xenograft models to gain knowledge about optimal dosing schedule and chemotherapeutic combinations for Aurora A/B inhibitors. RESULTS: SCH 1473759 was active against a large panel of tumor cell lines from different tissue origin and genetic backgrounds. Asynchronous cells required 24-h exposure to SCH 1473759 for maximal induction of >4 N DNA content and inhibition of cell growth. However, following taxane- or KSP inhibitor-induced mitotic arrest, less than 4-h exposure induced >4 N DNA content. This finding correlated with the ability of SCH 1473759 to accelerate exit from mitosis in response to taxane- and KSP inhibitor-induced arrest. We tested various dosing schedules in vivo and demonstrated SCH 1473759 dose- and schedule-dependent anti-tumor activity in four human tumor xenograft models. Further, the efficacy was enhanced in combination with taxanes and found to be most efficacious when SCH 1473759 was dosed 12-h post-taxane treatment. CONCLUSIONS: SCH 1473759 demonstrated potent mechanism-based activity, and activity was shown to be enhanced in combination with taxanes and KSP inhibitors. This information may be useful for optimizing the clinical efficacy of Aurora inhibitors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Imidazoles/farmacología , Cinesinas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazinas/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Aurora Quinasa A , Aurora Quinasas , Línea Celular Tumoral , Esquema de Medicación , Femenino , Humanos , Imidazoles/administración & dosificación , Masculino , Ratones , Ratones Desnudos , Neoplasias/patología , Pirazinas/administración & dosificación , Taxoides/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Curr Opin Drug Discov Devel ; 13(4): 389-402, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20597025

RESUMEN

Thermal shift assays are one of the label-free technologies gaining popularity in drug discovery and academic research. This review summarizes the various detection formats used in these assays, and provides an in-depth presentation of fluorescence-based thermal shift assays. The dual applications of stability profiling and affinity ranking are highlighted, and basic experimental protocol and data analysis algorithms are recommended. The advantages and limitations of the technology are also discussed to provide guidance for meaningful data interpretation. In addition, an emerging paradigm is described that incorporates thermal shift assays with other label-free biosensors and conventional IC(50) assays to derive a mechanism of action-informed SAR in lead identification and optimization during drug discovery.


Asunto(s)
Bioensayo/métodos , Fluorescencia , Temperatura , Algoritmos , Animales , Humanos , Preparaciones Farmacéuticas
13.
Expert Opin Drug Discov ; 5(11): 1023-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22827742

RESUMEN

Binding kinetics (BK), an often overlooked key aspect of the broader concept of drug mechanism of action (MOA), is increasingly recognized as a springboard from pharmacokinetics (PK) to pharmacodynamics, and as a critical differentiator and predictor for drug efficacy and safety. Just as greater attention to PK issues has helped reduce the attrition of drugs tested in clinical trials, the emerging paradigm shift from primarily affinity/potency-emphasized to a more holistic BK-perceptive and MOA-informed approach is expected to further enhance the success of drug discovery and development. This perspective attempts to envision what this new approach looks like when proper emphasis is placed on BK and MOA in designing better and best in class drugs.


Asunto(s)
Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Farmacología/métodos , Animales , Ensayos Clínicos como Asunto , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Cinética
14.
J Biomol Screen ; 15(1): 52-61, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20019290

RESUMEN

A high-throughput mass spectrometry assay to measure the catalytic activity of UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase, LpxC, is described. This reaction is essential in the biosynthesis of lipopolysaccharide (LPS) of gram-negative bacteria and is an attractive target for the development of new antibacterial agents. The assay uses the RapidFire mass spectrometry platform to measure the native LpxC substrate and the reaction product and thereby generates a ratiometric readout with minimal artifacts due to detection interference. The assay was robust in a high-throughput screen of a library of more than 700,000 compounds arrayed as orthogonal mixtures, with a median Z' factor of 0.74. Selected novel inhibitors from the screening campaign were confirmed as binding to LpxC by biophysical measurements using a thermal stability shift assay. Some inhibitors showed whole-cell antimicrobial activity against a sensitive strain of Escherichia coli with reduced LpxC activity (strain D22; minimum inhibitory concentrations ranging from 0.625-20 microg/mL). The results show that mass spectrometry-based screening is a valuable high-throughput screening tool for detecting inhibitors of enzymatic targets involving difficult to detect reactions.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Antibacterianos/análisis , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Espectrometría de Masas/métodos , Antibacterianos/química , Antibacterianos/farmacología , Dimetilsulfóxido/farmacología , Inhibidores Enzimáticos/química , Estabilidad de Enzimas/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Fluorescencia , Pruebas de Sensibilidad Microbiana , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Especificidad por Sustrato/efectos de los fármacos , Temperatura
15.
World J Gastroenterol ; 15(44): 5549-57, 2009 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-19938193

RESUMEN

AIM: To investigate the effect of short-chain fatty acids (SCFAs) on production of prostaglandin E(2) (PGE(2)), cytokines and chemokines in human monocytes. METHODS: Human neutrophils and monocytes were isolated from human whole blood by using 1-Step Polymorph and RosetteSep Human Monocyte Enrichment Cocktail, respectively. Human GPR41 and GPR43 mRNA expression was examined by quantitative real-time polymerase chain reaction. The calcium flux assay was used to examine the biological activities of SCFAs in human neutrophils and monocytes. The effect of SCFAs on human monocytes and peripheral blood mononuclear cells (PBMC) was studied by measuring PGE(2), cytokines and chemokines in the supernatant. The effect of SCFAs in vivo was examined by intraplantar injection into rat paws. RESULTS: Human GPR43 is highly expressed in human neutrophils and monocytes. SCFAs induce robust calcium flux in human neutrophils, but not in human monocytes. In this study, we show that SCFAs can induce human monocyte release of PGE(2) and that this effect can be enhanced in the presence of lipopolysaccharide (LPS). In addition, we demonstrate that PGE(2) production induced by SCFA was inhibited by pertussis toxin, suggesting the involvement of a receptor-mediated mechanism. Furthermore, SCFAs can specifically inhibit constitutive monocyte chemotactic protein-1 (MCP-1) production and LPS-induced interleukin-10 (IL-10) production in human monocytes without affecting the secretion of other cytokines and chemokines examined. Similar activities were observed in human PBMC for the release of PGE(2), MCP-1 and IL-10 after SCFA treatment. In addition, SCFAs inhibit LPS-induced production of tumor necrosis factor-alpha and interferon-gamma in human PBMC. Finally, we show that SCFAs and LPS can induce PGE(2) production in vivo by intraplantar injection into rat paws (P < 0.01). CONCLUSION: SCFAs can have distinct antiinflammatory activities due to their regulation of PGE(2), cytokine and chemokine release from human immune cells.


Asunto(s)
Antiinflamatorios/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Ácidos Grasos Volátiles/metabolismo , Animales , Antiinflamatorios/farmacología , Calcio/metabolismo , Quimiocinas/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Humanos , Interleucina-10/metabolismo , Lipopolisacáridos/metabolismo , Masculino , Monocitos/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Curr Opin Drug Discov Devel ; 12(4): 488-96, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19562645

RESUMEN

The importance of kinetics in drug-target interactions, and particularly the residence time of a drug with its target, is increasingly recognized to play a pivotal role in determining both the efficacy and toxicity of a drug. Drug residence time can often be demonstrated to be a key differentiating factor between drugs that act upon a common target. Drug-target residence time can result in either favorable or unfavorable outcomes, and the use of such information could lead to the more efficient design of best-in-class drugs. This review highlights several key concepts and observations related to drug-target residence time, and suggests the use of a kinetics-perceptive and energetics-informed approach to address the challenges facing current drug discovery efforts.


Asunto(s)
Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas , Farmacocinética , Relación Dosis-Respuesta a Droga , Modelos Biológicos , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Unión Proteica , Relación Estructura-Actividad , Factores de Tiempo
17.
J Biomol Screen ; 14(1): 49-58, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19171920

RESUMEN

The authors have characterized a set of cannabinoid CB(2) receptor ligands, including triaryl bis sulfone inverse agonists, in a cell-based receptor/beta-arrestin interaction assay (DiscoveRx PathHunter). The results were compared with results using a competitive ligand binding assay, and with effects on forskolin-stimulated cAMP levels (PerkinElmer LANCE). The authors show good correlation between the 3 assay systems tested, with the beta-arrestin protein complementation assay exhibiting a more robust signal than the cAMP assay for cannabinoid CB(2) agonists. Further assay validation shows that DiscoveRx PathHunter HEK293 CB(2) beta-arrestin assay can be carried out from cryopreserved cell suspensions, eliminating variations caused by the need for multiple cell pools during live cell screening campaigns. These results, and the authors' results evaluating a test set of random library compounds, validate the use of ligand-induced interaction between the human cannabinoid CB(2) receptor and beta-arrestin as an appropriate and valuable screening platform for compounds specific for the cannabinoid CB(2) receptor.


Asunto(s)
Arrestinas/análisis , Arrestinas/metabolismo , Receptor Cannabinoide CB2/metabolismo , Animales , Línea Celular , Cricetinae , Humanos , Ligandos , Receptor Cannabinoide CB2/genética , beta-Arrestinas
18.
J Med Chem ; 51(3): 581-8, 2008 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-18198821

RESUMEN

Melanin-concentrating hormone receptor 1 (MCH-R1) is a G-protein-coupled receptor (GPCR) and a target for the development of therapeutics for obesity. The structure-based development of MCH-R1 and other GPCR antagonists is hampered by the lack of an available experimentally determined atomic structure. A ligand-steered homology modeling approach has been developed (where information about existing ligands is used explicitly to shape and optimize the binding site) followed by docking-based virtual screening. Top scoring compounds identified virtually were tested experimentally in an MCH-R1 competitive binding assay, and six novel chemotypes as low micromolar affinity antagonist "hits" were identified. This success rate is more than a 10-fold improvement over random high-throughput screening, which supports our ligand-steered method. Clearly, the ligand-steered homology modeling method reduces the uncertainty of structure modeling for difficult targets like GPCRs.


Asunto(s)
Ligandos , Modelos Moleculares , Receptores de la Hormona Hipofisaria/antagonistas & inhibidores , Receptores de la Hormona Hipofisaria/química , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/química , Animales , Sitios de Unión , Unión Competitiva , Células CHO , Bovinos , Cricetinae , Cricetulus , Bases de Datos Factuales , Humanos , Receptores de la Hormona Hipofisaria/metabolismo , Receptores de Somatostatina/metabolismo , Rodopsina/química , Homología de Secuencia de Aminoácido , Procesos Estocásticos , Relación Estructura-Actividad , Termodinámica
19.
Genomics ; 90(5): 629-35, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17869477

RESUMEN

Mice lacking GPR103A expression display osteopenia. Analysis of mouse quantitative trait loci literature associated with bone mineral density suggested GPR103A ligand P518/Qrfp (chromosome 2qB) as a candidate osteoporosis gene. Promoter and coding regions of mouse P518/Qrfp were sequenced from genomic DNA obtained from the osteoporosis-prone strain SAMP6 and control strains SAMR1, A/J, AKR/J, BALB/c, C3H/HeJ, C57BL/6J, and DBA/2J. Four single-nucleotide polymorphisms (SNPs) were identified in only SAMP6 genomic DNA, g.-1773 T-->C, g.110 A-->G (N37S), g.188 G-->A (R63K), and g.135 T-->C (H45H). The promoter SNP generated a novel neuron-restrictive silencing factor binding site, a repressor that decreases gene expression in nonneuronal tissues. TaqMan analysis demonstrated fivefold lower P518/Qrfp liver expression in SAMP6 versus SAMR1 or C57BL/6J control strains. Tissue distribution of human, mouse, and rat P518/Qrfp and its receptors showed expression in bone and spinal cord. A direct role for P518/Qrfp function in maintaining bone mineral density is suggested.


Asunto(s)
Enfermedades Óseas Metabólicas/genética , Sistemas de Lectura Abierta/genética , Péptidos/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Carácter Cuantitativo Heredable , Receptores Acoplados a Proteínas G/genética , Secuencia de Aminoácidos , Animales , Densidad Ósea , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ligandos , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Ratas , Homología de Secuencia de Aminoácido , Distribución Tisular
20.
Brain Res Mol Brain Res ; 142(1): 58-64, 2005 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-16246453

RESUMEN

The mas-related gene (Mrg) family is a large family of G-protein-coupled receptors which are variable in number depending on species. The so-called sensory-neuron-specific receptors (SNSRs) make up a subset of the Mrg family, and several of these have been implicated in nociceptive processes. To verify their specific localization in sensory ganglia, we have determined the expression patterns of two of them, rMrgA and rMrgC, in a panel of rat tissues. The quantitative PCR results in the rat tissue panel indicate that, while several non-neuronal tissues contain significant levels of mRNA for both receptors, these two receptors are most highly expressed in dorsal root ganglia and trigeminal ganglia. Given this, we have examined the effects of spinal nerve ligation (SNL) on the expression of these genes. Peripheral neuropathy induced by ligation of spinal nerves at L5 and L6 resulted in a pronounced mechanical allodynia. These behavioral changes in tactile sensitivity were accompanied by significant decreases (10- to 100-fold) in the mRNA expression of both rMrgA and rMrgC exclusively in the L5 and L6 dorsal root ganglia ipsilateral to the SNL. In situ hybridization studies demonstrated that this decrease did not result from neuronal loss but rather from a reduction in the hybridization signals for rMrgC over small-to-medium diameter L5 and L6 dorsal root ganglia neurons. While the functional implications of the altered regulation of rMrgA and rMrgC in neuropathic pain models remain unclear, the results suggest that therapeutics targeting these receptors may have limited utility.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Lateralidad Funcional , Ganglios Espinales/citología , Hibridación in Situ/métodos , Ligadura/métodos , Masculino , Proteínas del Tejido Nervioso/genética , Neuralgia/etiología , Neuralgia/genética , Neuronas Aferentes/metabolismo , Dimensión del Dolor/métodos , Umbral del Dolor/fisiología , Enfermedades del Sistema Nervioso Periférico/complicaciones , Enfermedades del Sistema Nervioso Periférico/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...