Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Biol Chem ; 296: 100443, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33617882

RESUMEN

Polymorphic variation of immune system proteins can drive variability of individual immune responses. Endoplasmic reticulum aminopeptidase 1 (ERAP1) generates antigenic peptides for presentation by major histocompatibility complex class I molecules. Coding SNPs in ERAP1 have been associated with predisposition to inflammatory rheumatic disease and shown to affect functional properties of the enzyme, but the interplay between combinations of these SNPs as they exist in allotypes has not been thoroughly explored. We used phased genotype data to estimate ERAP1 allotype frequency in 2504 individuals across five major human populations, generated highly pure recombinant enzymes corresponding to the ten most common ERAP1 allotypes, and systematically characterized their in vitro enzymatic properties. We find that ERAP1 allotypes possess a wide range of enzymatic activities, up to 60-fold, whose ranking is substrate dependent. Strikingly, allotype 10, previously associated with Behçet's disease, is consistently a low-activity outlier, suggesting that a significant percentage of individuals carry a subactive ERAP1 gene. Enzymatic analysis revealed that ERAP1 allotypes can differ in both catalytic efficiency and substrate affinity, differences that can change intermediate accumulation in multistep trimming reactions. Alterations in efficacy of an allosteric inhibitor that targets the regulatory site suggest that allotypic variation influences the communication between the regulatory and the active site. Our work defines the wide landscape of ERAP1 activity in human populations and demonstrates how common allotypes can induce substrate-dependent variability in antigen processing, thus contributing, in synergy with major histocompatibility complex haplotypes, to immune response variability and predisposition to chronic inflammatory conditions.


Asunto(s)
Aminopeptidasas/inmunología , Aminopeptidasas/metabolismo , Antígenos de Histocompatibilidad Menor/inmunología , Antígenos de Histocompatibilidad Menor/metabolismo , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Aminopeptidasas/genética , Presentación de Antígeno/inmunología , Antígenos/genética , Antígenos/inmunología , Bases de Datos Genéticas , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Genotipo , Haplotipos/genética , Haplotipos/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Antígenos de Histocompatibilidad Menor/genética , Péptidos/metabolismo , Polimorfismo de Nucleótido Simple
2.
J Biol Chem ; 297(2): 100928, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34274316

RESUMEN

B-cell lymphoma 6 (BCL6) is a zinc finger transcriptional repressor possessing a BTB-POZ (BR-C, ttk, and bab for BTB; pox virus and zinc finger for POZ) domain, which is required for homodimerization and association with corepressors. BCL6 has multiple roles in normal immunity, autoimmunity, and some types of lymphoma. Mice bearing disrupted BCL6 loci demonstrate suppressed high-affinity antibody responses to T-dependent antigens. The corepressor binding groove in the BTB-POZ domain is a potential target for small compound-mediated therapy. Several inhibitors targeting this binding groove have been described, but these compounds have limited or absent in vivo activity. Biophysical studies of a novel compound, GSK137, showed an in vitro pIC50 of 8 and a cellular pIC50 of 7.3 for blocking binding of a peptide derived from the corepressor silencing mediator for retinoid or thyroid hormone receptors to the BCL6 BTB-POZ domain. The compound has good solubility (128 µg/ml) and permeability (86 nM/s). GSK137 caused little change in cell viability or proliferation in four BCL6-expressing B-cell lymphoma lines, although there was modest dose-dependent accumulation of G1 phase cells. Pharmacokinetic studies in mice showed a profile compatible with achieving good levels of target engagement. GSK137, administered orally, suppressed immunoglobulin G responses and reduced numbers of germinal centers and germinal center B cells following immunization of mice with the hapten trinitrophenol. Overall, we report a novel small-molecule BCL6 inhibitor with in vivo activity that inhibits the T-dependent antigen immune response.


Asunto(s)
Proteínas Proto-Oncogénicas c-bcl-6 , Animales , Linfocitos B/metabolismo , Humanos , Ratones , Transcripción Genética , Dedos de Zinc
3.
Bioorg Med Chem ; 27(8): 1456-1478, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30858025

RESUMEN

With the goal of discovering more selective anti-inflammatory drugs, than COX inhibitors, to attenuate prostaglandin signaling, a fragment-based screen of hematopoietic prostaglandin D synthase was performed. The 76 crystallographic hits were sorted into similar groups, with the 3-cyano-quinoline 1a (FP IC50 = 220,000 nM, LE = 0.43) being a potent member of the 6,6-fused heterocyclic cluster. Employing SAR insights gained from structural comparisons of other H-PGDS fragment binding mode clusters, the initial hit 1a was converted into the 70-fold more potent quinoline 1d (IC50 = 3,100 nM, LE = 0.49). A systematic substitution of the amine moiety of 1d, utilizing structural information and array chemistry, with modifications to improve inhibitor stability, resulted in the identification of the 300-fold more active H-PGDS inhibitor tool compound 1bv (IC50 = 9.9 nM, LE = 0.42). This selective inhibitor exhibited good murine pharmacokinetics, dose-dependently attenuated PGD2 production in a mast cell degranulation assay and should be suitable to further explore H-PGDS biology.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Lipocalinas/antagonistas & inhibidores , Quinolinas/química , Quinolinas/farmacología , Animales , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacocinética , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/química , Lipocalinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Quinolinas/farmacocinética
4.
SLAS Discov ; 28(1): 3-11, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36414185

RESUMEN

MALDI-TOF MS is a powerful analytical technique that provides a fast and label-free readout for in vitro assays in the high-throughput screening (HTS) environment. Here, we describe the development of a novel, HTS compatible, MALDI-TOF MS-based drug discovery assay for the endoplasmic reticulum aminopeptidase 1 (ERAP1), an important target in immuno-oncology and auto-immune diseases. A MALDI-TOF MS assay was developed beginning with an already established ERAP1 RapidFire MS (RF MS) assay, where the peptide YTAFTIPSI is trimmed into the product TAFTIPSI. We noted low ionisation efficiency of these peptides in MALDI-TOF MS and hence incorporated arginine residues into the peptide sequences to improve ionisation. The optimal assay conditions were established with these new basic assay peptides on the MALDI-TOF MS platform and validated with known ERAP1 inhibitors. Assay stability, reproducibility and robustness was demonstrated on the MALDI-TOF MS platform. From a set of 699 confirmed ERAP1 binders, identified in a prior affinity selection mass spectrometry (ASMS) screen, active compounds were determined at single concentration and in a dose-response format with the new MALDI-TOF MS setup. Furthermore, to allow for platform performance comparison, the same compound set was tested on the established RF MS setup, as the new basic peptides showed fragmentation in ESI-MS. The two platforms showed a comparable performance, but the MALDI-TOF MS platform had several advantages, such as shorter sample cycle times, reduced reagent consumption, and a lower tight-binding limit.


Asunto(s)
Aminopeptidasas , Ensayos Analíticos de Alto Rendimiento , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Reproducibilidad de los Resultados , Ensayos Analíticos de Alto Rendimiento/métodos , Péptidos
5.
Transcult Psychiatry ; : 13634615211065869, 2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-35040741

RESUMEN

Evidence suggests that faith communities can support psychological wellbeing but can also potentially diminish wellbeing through stigma, imposed spiritualization, and marginalization. In particular, for evangelical Christianity, whose theological praxis typically accentuates literalist spiritual onto-etiologies, including the belief that mental distress can be treated solely through spiritual intervention (prayer, fasting, and deliverance), there may be negative implications for Christians with mental distress. The current qualitative survey examined the responses of 293 self-identified evangelical Christians, concerning their experiences of mental distress in relation to their church community. An inductive thematic analysis revealed five themes: 1) Tensions between Faith and Suffering; 2) Cautions about a Reductive Spiritualization; 3) Feeling Othered and Disconnected; 4) Faith as Alleviating Distress; and 5) Inviting an Integrationist Position. Findings reveal stigma and the totalizing spiritualization of mental distress can be experienced as both dismissive and invalidating and can problematize secular help-seeking. This lends support to previous research which has suggested that evangelical Christian communities tend to link mental distress to spiritual deficiencies, which can hold potentially negative consequences for their wellbeing. Nevertheless, a degree of complexity and nuance emerged whereby spiritual explanations and interventions were also experienced as sometimes helpful in alleviating suffering. Overall, findings suggest evangelical communities are increasingly adopting integrationist understandings of mental distress, whereby spiritual narratives are assimilated alongside the biopsychosocial. We argue that church communities and psychotherapeutic practitioners should support movement from a position of dichotomizing psychological suffering (e.g., spiritual vs. biopsychosocial) towards a spiritually syntonic frame, which contextualizes distress in terms of the whole person. Considerations for psychotherapeutic practice and further research are made.

6.
EMBO Mol Med ; 13(3): e13167, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33512066

RESUMEN

Severe α1 -antitrypsin deficiency results from the Z allele (Glu342Lys) that causes the accumulation of homopolymers of mutant α1 -antitrypsin within the endoplasmic reticulum of hepatocytes in association with liver disease. We have used a DNA-encoded chemical library to undertake a high-throughput screen to identify small molecules that bind to, and stabilise Z α1 -antitrypsin. The lead compound blocks Z α1 -antitrypsin polymerisation in vitro, reduces intracellular polymerisation and increases the secretion of Z α1 -antitrypsin threefold in an iPSC model of disease. Crystallographic and biophysical analyses demonstrate that GSK716 and related molecules bind to a cryptic binding pocket, negate the local effects of the Z mutation and stabilise the bound state against progression along the polymerisation pathway. Oral dosing of transgenic mice at 100 mg/kg three times a day for 20 days increased the secretion of Z α1 -antitrypsin into the plasma by sevenfold. There was no observable clearance of hepatic inclusions with respect to controls over the same time period. This study provides proof of principle that "mutation ameliorating" small molecules can block the aberrant polymerisation that underlies Z α1 -antitrypsin deficiency.


Asunto(s)
Deficiencia de alfa 1-Antitripsina , alfa 1-Antitripsina , Animales , Retículo Endoplásmico , Hepatocitos , Ratones , alfa 1-Antitripsina/genética
8.
Biochem J ; 419(1): 65-73, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19061480

RESUMEN

Identification of small-molecule inhibitors by high-throughput screening necessitates the development of robust, reproducible and cost-effective assays. The assay approach adopted may utilize isolated proteins or whole cells containing the target of interest. To enable protein-based assays, the baculovirus expression system is commonly used for generation and isolation of recombinant proteins. We have applied the baculovirus system into a cell-based assay format using NIK [NF-kappaB (nuclear factor kappaB)-inducing kinase] as a paradigm. We illustrate the use of the insect-cell-based assay in monitoring the activity of NIK against its physiological downstream substrate IkappaB (inhibitor of NF-kappaB) kinase-1. The assay was robust, yielding a signal/background ratio of 2:1 and an average Z' value of >0.65 when used to screen a focused compound set. Using secondary assays to validate a selection of the hits, we identified a compound that (i) was non-cytotoxic, (ii) interacted directly with NIK, and (iii) inhibited lymphotoxin-induced NF-kappaB p52 translocation to the nucleus. The insect cell assay represents a novel approach to monitoring kinase inhibition, with major advantages over other cell-based systems including ease of use, amenability to scale-up, protein expression levels and the flexibility to express a number of proteins by infecting with numerous baculoviruses.


Asunto(s)
Bioensayo/métodos , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Western Blotting , Línea Celular , Humanos , Quinasa I-kappa B/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Fosforilación , Spodoptera , Quinasa de Factor Nuclear kappa B
9.
J Med Chem ; 63(6): 3348-3358, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32109056

RESUMEN

ER aminopeptidase 1 (ERAP1) is an intracellular enzyme that generates antigenic peptides and is an emerging target for cancer immunotherapy and the control of autoimmunity. ERAP1 inhibitors described previously target the active site and are limited in selectivity, minimizing their clinical potential. To address this, we targeted the regulatory site of ERAP1 using a high-throughput screen and discovered a small molecule hit that is highly selective for ERAP1. (4aR,5S,6R,8S,8aR)-5-(2-(Furan-3-yl)ethyl)-8-hydroxy-5,6,8a-trimethyl-3,4,4a,5,6,7,8,8a-octahydronaphthalene-1-carboxylic acid is a natural product found in Dodonaea viscosa that constitutes a submicromolar, highly selective, and cell-active modulator of ERAP1. Although the compound activates hydrolysis of small model substrates, it is a competitive inhibitor for physiologically relevant longer peptides. Crystallographic analysis confirmed that the compound targets the regulatory site of the enzyme that normally binds the C-terminus of the peptide substrate. Our findings constitute a novel starting point for the development of selective ERAP1 modulators that have potential for further clinical development.


Asunto(s)
Aminopeptidasas/antagonistas & inhibidores , Presentación de Antígeno/efectos de los fármacos , Diterpenos de Tipo Clerodano/farmacología , Epítopos/metabolismo , Péptidos/metabolismo , Inhibidores de Proteasas/farmacología , Sitio Alostérico , Aminopeptidasas/química , Aminopeptidasas/metabolismo , Animales , Dominio Catalítico , Cristalografía por Rayos X , Diterpenos de Tipo Clerodano/química , Diterpenos de Tipo Clerodano/metabolismo , Activadores de Enzimas/química , Activadores de Enzimas/metabolismo , Activadores de Enzimas/farmacología , Epítopos/química , Células HeLa , Humanos , Ratones , Antígenos de Histocompatibilidad Menor/química , Antígenos de Histocompatibilidad Menor/metabolismo , Péptidos/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Unión Proteica , Proteolisis/efectos de los fármacos
10.
JCI Insight ; 3(16)2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30135318

RESUMEN

Inhibiting MAPK14 (p38α) diminishes cardiac damage in myocardial ischemia. During myocardial ischemia, p38α interacts with TAB1, a scaffold protein, which promotes p38α autoactivation; active p38α (pp38α) then transphosphorylates TAB1. Previously, we solved the X-ray structure of the p38α-TAB1 (residues 384-412) complex. Here, we further characterize the interaction by solving the structure of the pp38α-TAB1 (residues 1-438) complex in the active state. Based on this information, we created a global knock-in (KI) mouse with substitution of 4 residues on TAB1 that we show are required for docking onto p38α. Whereas ablating p38α or TAB1 resulted in early embryonal lethality, the TAB1-KI mice were viable and had no appreciable alteration in their lymphocyte repertoire or myocardial transcriptional profile; nonetheless, following in vivo regional myocardial ischemia, infarction volume was significantly reduced and the transphosphorylation of TAB1 was disabled. Unexpectedly, the activation of myocardial p38α during ischemia was only mildly attenuated in TAB1-KI hearts. We also identified a group of fragments able to disrupt the interaction between p38α and TAB1. We conclude that the interaction between the 2 proteins can be targeted with small molecules. The data reveal that it is possible to selectively inhibit signaling downstream of p38α to attenuate ischemic injury.


Asunto(s)
Adamantano/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Adamantano/análogos & derivados , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/genética , Mutación , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/etiología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Dominios y Motivos de Interacción de Proteínas/genética , Daño por Reperfusión/complicaciones
11.
J Med Chem ; 60(8): 3383-3404, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28398044

RESUMEN

Recently, we reported a novel role for KMO in the pathogenesis of acute pancreatitis (AP). A number of inhibitors of kynurenine 3-monooxygenase (KMO) have previously been described as potential treatments for neurodegenerative conditions and particularly for Huntington's disease. However, the inhibitors reported to date have insufficient aqueous solubility relative to their cellular potency to be compatible with the intravenous (iv) dosing route required in AP. We have identified and optimized a novel series of high affinity KMO inhibitors with favorable physicochemical properties. The leading example is exquisitely selective, has low clearance in two species, prevents lung and kidney damage in a rat model of acute pancreatitis, and is progressing into preclinical development.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Pancreatitis/tratamiento farmacológico , Enfermedad Aguda , Animales , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratas
12.
Nat Commun ; 8: 15827, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28604669

RESUMEN

Kynurenine-3-monooxygenase (KMO) is a key FAD-dependent enzyme of tryptophan metabolism. In animal models, KMO inhibition has shown benefit in neurodegenerative diseases such as Huntington's and Alzheimer's. Most recently it has been identified as a target for acute pancreatitis multiple organ dysfunction syndrome (AP-MODS); a devastating inflammatory condition with a mortality rate in excess of 20%. Here we report and dissect the molecular mechanism of action of three classes of KMO inhibitors with differentiated binding modes and kinetics. Two novel inhibitor classes trap the catalytic flavin in a previously unobserved tilting conformation. This correlates with picomolar affinities, increased residence times and an absence of the peroxide production seen with previous substrate site inhibitors. These structural and mechanistic insights culminated in GSK065(C1) and GSK366(C2), molecules suitable for preclinical evaluation. Moreover, revising the repertoire of flavin dynamics in this enzyme class offers exciting new opportunities for inhibitor design.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Insuficiencia Multiorgánica/metabolismo , Pancreatitis/metabolismo , Animales , Inhibidores Enzimáticos/química , Escherichia coli/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Modelos Moleculares , Dominios Proteicos , Células Sf9
13.
Nat Med ; 22(2): 202-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26752518

RESUMEN

Acute pancreatitis (AP) is a common and devastating inflammatory condition of the pancreas that is considered to be a paradigm of sterile inflammation leading to systemic multiple organ dysfunction syndrome (MODS) and death. Acute mortality from AP-MODS exceeds 20% (ref. 3), and the lifespans of those who survive the initial episode are typically shorter than those of the general population. There are no specific therapies available to protect individuals from AP-MODS. Here we show that kynurenine-3-monooxygenase (KMO), a key enzyme of tryptophan metabolism, is central to the pathogenesis of AP-MODS. We created a mouse strain that is deficient for Kmo (encoding KMO) and that has a robust biochemical phenotype that protects against extrapancreatic tissue injury to the lung, kidney and liver in experimental AP-MODS. A medicinal chemistry strategy based on modifications of the kynurenine substrate led to the discovery of the oxazolidinone GSK180 as a potent and specific inhibitor of KMO. The binding mode of the inhibitor in the active site was confirmed by X-ray co-crystallography at 3.2 Å resolution. Treatment with GSK180 resulted in rapid changes in the levels of kynurenine pathway metabolites in vivo, and it afforded therapeutic protection against MODS in a rat model of AP. Our findings establish KMO inhibition as a novel therapeutic strategy in the treatment of AP-MODS, and they open up a new area for drug discovery in critical illness.


Asunto(s)
Benzoxazoles/farmacología , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Insuficiencia Multiorgánica/genética , Oxazolidinonas/farmacología , Pancreatitis/genética , Propionatos/farmacología , ARN Mensajero/metabolismo , Enfermedad Aguda , Animales , Cromatografía Liquida , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Células HEK293 , Hepatocitos/metabolismo , Humanos , Técnicas In Vitro , Riñón/metabolismo , Riñón/patología , Quinurenina 3-Monooxigenasa/genética , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/patología , Páncreas/metabolismo , Páncreas/patología , Pancreatitis/complicaciones , Pancreatitis/patología , Ratas , Espectrometría de Masas en Tándem , Triptófano/metabolismo
14.
ACS Infect Dis ; 1(12): 615-26, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-27623058

RESUMEN

We have targeted the Mycobacterium tuberculosis decaprenylphosphoryl-ß-d-ribose oxidase (Mt-DprE1) for potential chemotherapeutic intervention of tuberculosis. A multicopy suppression strategy that overexpressed Mt-DprE1 in M. bovis BCG was used to profile the publically available GlaxoSmithKline antimycobacterial compound set, and one compound (GSK710) was identified that showed an 8-fold higher minimum inhibitory concentration relative to the control strain. Analogues of GSK710 show a clear relationship between whole cell potency and in vitro activity using an enzymatic assay employing recombinant Mt-DprE1, with binding affinity measured by fluorescence quenching of the flavin cofactor of the enzyme. M. bovis BCG spontaneous resistant mutants to GSK710 and a closely related analogue were isolated and sequencing of ten such mutants revealed a single point mutation at two sites, E221Q or G248S within DprE1, providing further evidence that DprE1 is the main target of these compounds. Finally, time-lapse microscopy experiments showed that exposure of M. tuberculosis to a compound of this series arrests bacterial growth rapidly followed by a slower cytolysis phase.

15.
J Biomol Screen ; 8(1): 19-33, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12854995

RESUMEN

Single-molecule detection technologies are becoming a powerful readout format to support ultra-high-throughput screening. These methods are based on the analysis of fluorescence intensity fluctuations detected from a small confocal volume element. The fluctuating signal contains information about the mass and brightness of the different species in a mixture. The authors demonstrate a number of applications of fluorescence intensity distribution analysis (FIDA), which discriminates molecules by their specific brightness. Examples for assays based on brightness changes induced by quenching/dequenching of fluorescence, fluorescence energy transfer, and multiple-binding stoichiometry are given for important drug targets such as kinases and proteases. FIDA also provides a powerful method to extract correct biological data in the presence of compound fluorescence.


Asunto(s)
Microscopía Confocal , Alcohol Deshidrogenasa/análisis , Fosfatasa Alcalina/análisis , Antígenos de Plaqueta Humana/análisis , Antígenos de Plaqueta Humana/metabolismo , Endopeptidasas/análisis , Endopeptidasas/metabolismo , Fluorescencia , Ligandos , ARN/metabolismo
16.
J Biomol Screen ; 8(6): 648-59, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14711390

RESUMEN

The rapid increase in size of compound libraries, as well as new targets emerging from the Human Genome Project, require progress in ultra-high-throughput screening (uHTS) systems. In a joint effort with scientists and engineers from the biotech and the pharmaceutical industry, a modular, fully integrated system for miniaturized uHTS was developed. The goal was to achieve high data quality in small assay volumes (1-4 microL) combined with reliable and unattended operation. Two new confocal fluorescence readers have been designed. One of the instruments is a 4-channel confocal fluorescence reader, measuring with 4 objectives in parallel. The fluorescence readout is based on single-molecule detection methods, allowing high sensitivity at low tracer concentrations and delivering an information-rich output. The other instrument is a confocal fluorescence imaging reader, where the images are analyzed in terms of generic patterns and quantified in units of intensity per pixel. Both readers are spanning the application range from assays with isolated targets in homogenous solution or membrane vesicle-based assays (4-channel reader) to cell-based assays (imaging reader). Results from a comprehensive test on these assay types demonstrate the high quality and robustness of this screening system.


Asunto(s)
Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Antibacterianos/farmacología , Muerte Celular , Línea Celular , Supervivencia Celular , Computadores , Contaminación de Medicamentos , Fluorescencia , Humanos , Concentración 50 Inhibidora , Ligandos , Microscopía Confocal , Péptidos/análisis , Proteínas/análisis , Ribosomas/efectos de los fármacos , Sensibilidad y Especificidad , Células U937
17.
J Biomol Screen ; 19(4): 508-15, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24381207

RESUMEN

Kynurenine 3-monooxygenase (KMO) is a therapeutically important target on the eukaryotic tryptophan catabolic pathway, where it converts L-kynurenine (Kyn) to 3-hydroxykynurenine (3-HK). We have cloned and expressed the human form of this membrane protein as a full-length GST-fusion in a recombinant baculovirus expression system. An enriched membrane preparation was used for a directed screen of approximately 78,000 compounds using a RapidFire mass spectrometry (RF-MS) assay. The RapidFire platform provides an automated solid-phase extraction system that gives a throughput of approximately 7 s per well to the mass spectrometer, where direct measurement of both the substrate and product allowed substrate conversion to be determined. The RF-MS methodology is insensitive to assay interference, other than where compounds have the same nominal mass as Kyn or 3-HK and produce the same mass transition on fragmentation. These instances could be identified by comparison with the product-only data. The screen ran with excellent performance (average Z' value 0.8) and provided several tractable hit series for further investigation.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Quinurenina 3-Monooxigenasa/antagonistas & inhibidores , Espectrometría de Masas , Animales , Dominio Catalítico , Línea Celular , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Humanos , Cinética , Quinurenina 3-Monooxigenasa/química , Quinurenina 3-Monooxigenasa/metabolismo , Espectrometría de Masas/métodos , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
18.
J Biomol Screen ; 17(5): 641-50, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22337655

RESUMEN

Microsomal prostaglandin E synthase-1 (mPGES-1) represents an attractive target for the treatment of rheumatoid arthritis and pain, being upregulated in response to inflammatory stimuli. Biochemical assays for prostaglandin E synthase activity are complicated by the instability of the substrate (PGH(2)) and the challenge of detection of the product (PGE(2)). A coupled fluorescent assay is described for mPGES-1 where PGH(2) is generated in situ using the action of cyclooxygenase 2 (Cox-2) on arachidonic acid. PGE(2) is detected by coupling through 15-prostaglandin dehydrogenase (15-PGDH) and diaphorase. The overall coupled reaction was miniaturized to 1536-well plates and validated for high-throughput screening. For compound progression, a novel high-throughput mass spectrometry assay was developed using the RapidFire platform. The assay employs the same in situ substrate generation step as the fluorescent assay, after which both PGE(2) and a reduced form of the unreacted substrate were detected by mass spectrometry. Pharmacology and assay quality were comparable between both assays, but the mass spectrometry assay was shown to be less susceptible to interference and false positives. Exploiting the throughput of the fluorescent assay and the label-free, direct detection of the RapidFire has proved to be a powerful lead discovery strategy for this challenging target.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/química , Ensayos Analíticos de Alto Rendimiento/métodos , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Espectrometría de Masas/métodos , Ciclooxigenasa 2/metabolismo , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Colorantes Fluorescentes/metabolismo , Humanos , Concentración 50 Inhibidora , Oxidorreductasas Intramoleculares/metabolismo , Prostaglandina-E Sintasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA