Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Pharmacol Exp Ther ; 383(1): 56-69, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35926871

RESUMEN

Ion channels are targets of considerable therapeutic interest to address a wide variety of neurologic indications, including pain perception. Current pharmacological strategies have focused mostly on small molecule approaches that can be limited by selectivity requirements within members of a channel family or superfamily. Therapeutic antibodies have been proposed, designed, and characterized to alleviate this selectivity limitation; however, there are no Food and Drug Administration-approved therapeutic antibody-based drugs targeting ion channels on the market to date. Here, in an effort to identify novel classes of engineered ion channel modulators for potential neurologic therapeutic applications, we report the generation and characterization of six (EC50 < 25nM) Cys-loop receptor family monoclonal antibodies with modulatory function against rat and human glycine receptor alpha 1 (GlyRα1) and/or GlyRα3. These antibodies have activating (i.e., positive modulator) or inhibiting (i.e., negative modulator) profiles. Moreover, GlyRα3 selectivity was successfully achieved for two of the three positive modulators identified. When dosed intravenously, the antibodies achieved sufficient brain exposure to cover their calculated in vitro EC50 values. When compared head-to-head at identical exposures, the GlyRα3-selective antibody showed a more desirable safety profile over the nonselective antibody, thus demonstrating, for the first time, an advantage for GlyRα3-selectivity. Our data show that ligand-gated ion channels of the glycine receptor family within the central nervous system can be functionally modulated by engineered biologics in a dose-dependent manner and that, despite high protein homology between the alpha subunits, selectivity can be achieved within this receptor family, resulting in future therapeutic candidates with more desirable drug safety profiles. SIGNIFICANCE STATEMENT: This study presents immunization and multiplatform screening approaches to generate a diverse library of functional antibodies (agonist, potentiator, or inhibitory) raised against human glycine receptors (GlyRs). This study also demonstrates the feasibility of acquiring alpha subunit selectivity, a desirable therapeutic profile. When tested in vivo, these tool molecules demonstrated an increased safety profile in favor of GlyRα3-selectivity. These are the first reported functional GlyR antibodies that may open new avenues to treating central nervous system diseases with subunit selective biologics.


Asunto(s)
Anticuerpos Monoclonales , Receptores de Glicina , Animales , Ratas , Humanos , Receptores de Glicina/metabolismo , Ligandos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Transmisión Sináptica
2.
J Lipid Res ; 61(5): 790-807, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32102800

RESUMEN

Membrane-bound proteins have been proposed to mediate the transport of long-chain FA (LCFA) transport through the plasma membrane (PM). These proposals are based largely on reports that PM transport of LCFAs can be blocked by a number of enzymes and purported inhibitors of LCFA transport. Here, using the ratiometric pH indicator (2',7'-bis-(2-carboxyethyl)-5-(and-6-)-carboxyfluorescein and acrylodated intestinal FA-binding protein-based dual fluorescence assays, we investigated the effects of nine inhibitors of the putative FA transporter protein CD36 on the binding and transmembrane movement of LCFAs. We particularly focused on sulfosuccinimidyl oleate (SSO), reported to be a competitive inhibitor of CD36-mediated LCFA transport. Using these assays in adipocytes and inhibitor-treated protein-free lipid vesicles, we demonstrate that rapid LCFA transport across model and biological membranes remains unchanged in the presence of these purported inhibitors. We have previously shown in live cells that CD36 does not accelerate the transport of unesterified LCFAs across the PM. Our present experiments indicated disruption of LCFA metabolism inside the cell within minutes upon treatment with many of the "inhibitors" previously assumed to inhibit LCFA transport across the PM. Furthermore, using confocal microscopy and a specific anti-SSO antibody, we found that numerous intracellular and PM-bound proteins are SSO-modified in addition to CD36. Our results support the hypothesis that LCFAs diffuse rapidly across biological membranes and do not require an active protein transporter for their transmembrane movement.


Asunto(s)
Antígenos CD36/metabolismo , Ácidos Grasos/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Embrión de Pollo , Interacciones Farmacológicas , Concentración de Iones de Hidrógeno , Ácido Oléico/metabolismo , Palmitatos/farmacología
3.
Chembiochem ; 17(11): 990-4, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26991964

RESUMEN

The receptor tyrosine kinase EGFR is regulated by complex conformational changes, and this conformational control is disturbed in certain types of cancer. Many ligands are known to bind EGFR in its active conformation, thereby preventing ATP from binding. Only a few ligands are known to stabilize EGFR in its inactive conformation, thus providing novel strategies for perturbing EGFR activity. We report a direct binding assay that enables the identification of novel ligands that bind to and stabilize the inactive conformation of EGFR.


Asunto(s)
Receptores ErbB/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Sitios de Unión , Receptores ErbB/química , Receptores ErbB/genética , Clorhidrato de Erlotinib/química , Clorhidrato de Erlotinib/metabolismo , Lapatinib , Ligandos , Mutagénesis Sitio-Dirigida , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Estructura Terciaria de Proteína , Quinazolinas/química , Quinazolinas/metabolismo , Espectrometría de Fluorescencia
4.
J Am Chem Soc ; 135(18): 6838-41, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23611635

RESUMEN

Normal cellular function, such as signal transduction, is largely controlled by the reversible phosphorylation of cellular proteins catalyzed by two major classes of enzymes, kinases and phosphatases. A misbalance in this complex and dynamic interplay leads to a variety of severe diseases, such as cancer, inflammation, or autoimmune diseases. This makes kinases as well as phosphatases equally attractive targets for therapeutic manipulation by small molecules. While the development of kinase inhibitors has resulted in several blockbuster drugs, such as imatinib, with remarkable success in the clinic and sales of many billions of U.S. dollars per year, not a single phosphatase inhibitor has yet been approved for clinical use. Similar to the kinase world, substrate-competitive phosphatase inhibitors have been developed but were not suitable for further development into clinical candidates due to their charge and limited selectivity. Research efforts, therefore, have shifted to the exploitation of allosteric sites that can regulate phosphatase activity and may enable the discovery of novel modulators of phosphatase activity with much improved pharmacological properties. However, assay systems, which enable the straightforward discovery of these inhibitor types, are missing. Here, we present a novel binding assay capable of detecting ligands of an allosteric pocket of the protein tyrosine phosphatase 1B. This assay is suitable for high-throughput screening and selectively detects ligands which bind to this unique site with a clear discrimination from substrate-competitive ligands.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Regulación Alostérica/efectos de los fármacos , Inhibidores Enzimáticos/química , Humanos , Ligandos , Modelos Moleculares , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Relación Estructura-Actividad
5.
J Am Chem Soc ; 135(22): 8400-8, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23672540

RESUMEN

In the attempt to discover novel chemical scaffolds that can modulate the activity of disease-associated enzymes, such as kinases, biochemical assays are usually deployed in high-throughput screenings. First-line assays, such as activity-based assays, often rely on fluorescent molecules by measuring a change in the total emission intensity, polarization state, or energy transfer to another fluorescent molecule. However, under certain conditions, intrinsic compound fluorescence can lead to difficult data analysis and to false-positive, as well as false-negative, hits. We have reported previously on a powerful direct binding assay called fluorescent labels in kinases ('FLiK'), which enables a sensitive measurement of conformational changes in kinases upon ligand binding. In this assay system, changes in the emission spectrum of the fluorophore acrylodan, induced by the binding of a ligand, are translated into a robust assay readout. However, under the excitation conditions of acrylodan, intrinsic compound fluorescence derived from highly conjugated compounds complicates data analysis. We therefore optimized this method by identifying novel fluorophores that excite in the far red, thereby avoiding compound fluorescence. With this advancement, even rigid compounds with multiple π-conjugated ring systems can now be measured reliably. This study was performed on three different kinase constructs with three different labeling sites, each undergoing distinct conformational changes upon ligand binding. It may therefore serve as a guideline for the establishment of novel fluorescence-based detection assays.


Asunto(s)
2-Naftilamina/análogos & derivados , Pruebas de Enzimas , Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Proteínas Quinasas/química , 2-Naftilamina/química , 2-Naftilamina/metabolismo , Modelos Moleculares , Estructura Molecular , Proteínas Quinasas/metabolismo
6.
J Am Chem Soc ; 134(22): 9138-41, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22612329

RESUMEN

Abelson (Abl) tyrosine kinase is an important cellular enzyme that is rendered constitutively active in the breakpoint cluster region (BCR)-Abl fusion protein, contributing to several forms of leukemia. Although inhibiting BCR-Abl activity with imatinib shows great clinical success, many patients acquire secondary mutations that result in resistance to imatinib. Second-generation inhibitors such as dasatinib and nilotinib can overcome the majority of these mutations but fail to treat patients with an especially prevalent T315I mutation at the gatekeeper position of the kinase domain. However, a combination of nilotinib with an allosteric type IV inhibitor was recently shown to overcome this clinically relevant point mutation. In this study, we present the development of a direct binding assay that enables the straightforward detection of allosteric inhibitors which bind within the myristate pocket of Abl. The assay is amenable to high-throughput screening and exclusively detects the binding of ligands to this unique allosteric site.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-abl/metabolismo , Relación Estructura-Actividad
7.
Nat Chem Biol ; 5(6): 394-6, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19396179

RESUMEN

Targeting kinases outside the highly conserved ATP pocket is thought to be a promising strategy for overcoming bottlenecks in kinase inhibitor research, such as limited selectivity and drug resistance. Here we report the development and application of a direct binding assay to detect small molecules that stabilize the inactive conformation of the tyrosine kinase cSrc. Protein X-ray crystallography validated the assay results and confirmed an exclusively allosteric binding mode.


Asunto(s)
Regulación Alostérica , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Enlace de Hidrógeno , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo
8.
Bioorg Med Chem ; 19(1): 429-39, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21130659

RESUMEN

Here we present the synthesis and biological activity of a series of 7-substituted-1-(3-bromophenylamino)isoquinoline-4-carbonitriles as inhibitors of myosin light chain kinase (MLCK) and the epidermal growth factor receptor kinase (EGFR). The inhibitory effect of these molecules was found to be dependent on the nature of the substituents at the 7-position of the isoquinoline scaffold.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Isoquinolinas/farmacología , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Trifosfato/metabolismo , Humanos , Modelos Moleculares , Relación Estructura-Actividad
9.
SLAS Discov ; 26(4): 503-517, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33430712

RESUMEN

The aberrant regulation of protein expression and function can drastically alter cellular physiology and lead to numerous pathophysiological conditions such as cancer, inflammatory diseases, and neurodegeneration. The steady-state expression levels of endogenous proteins are controlled by a balance of de novo synthesis rates and degradation rates. Moreover, the levels of activated proteins in signaling cascades can be further modulated by a variety of posttranslational modifications and protein-protein interactions. The field of targeted protein degradation is an emerging area for drug discovery in which small molecules are used to recruit E3 ubiquitin ligases to catalyze the ubiquitination and subsequent degradation of disease-causing target proteins by the proteasome in both a dose- and time-dependent manner. Traditional approaches for quantifying protein level changes in cells, such as Western blots, are typically low throughput with limited quantification, making it hard to drive the rapid development of therapeutics that induce selective, rapid, and sustained protein degradation. In the last decade, a number of techniques and technologies have emerged that have helped to accelerate targeted protein degradation drug discovery efforts, including the use of fluorescent protein fusions and reporter tags, flow cytometry, time-resolved fluorescence energy transfer (TR-FRET), and split luciferase systems. Here we discuss the advantages and disadvantages associated with these technologies and their application to the development and optimization of degraders as therapeutics.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento , Terapia Molecular Dirigida/métodos , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina-Proteína Ligasas/metabolismo , Células Eucariotas/citología , Células Eucariotas/efectos de los fármacos , Células Eucariotas/metabolismo , Citometría de Flujo/métodos , Humanos , Ligandos , Unión Proteica , Proteolisis/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Espectrometría de Fluorescencia/métodos , Coloración y Etiquetado/métodos , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/efectos de los fármacos
10.
SLAS Discov ; 26(4): 547-559, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33780296

RESUMEN

Recent advances in targeted protein degradation have enabled chemical hijacking of the ubiquitin-proteasome system to treat disease. The catalytic rate of cereblon (CRBN)-dependent bifunctional degradation activating compounds (BiDAC), which recruit CRBN to a chosen target protein, resulting in its ubiquitination and proteasomal degradation, is an important parameter to consider during the drug discovery process. In this work, an in vitro system was developed to measure the kinetics of BRD4 bromodomain 1 (BD1) ubiquitination by fitting an essential activator kinetic model to these data. The affinities between BiDACs, BD1, and CRBN in the binary complex, ternary complex, and full ubiquitination complex were characterized. Together, this work provides a new tool for understanding and optimizing the catalytic and thermodynamic properties of BiDACs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Bioensayo , Proteínas de Ciclo Celular/metabolismo , Oxindoles/farmacología , Ftalimidas/farmacología , Procesamiento Proteico-Postraduccional , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Sistema Libre de Células/química , Sistema Libre de Células/metabolismo , Células HeLa , Humanos , Cinética , Oxindoles/síntesis química , Ftalimidas/síntesis química , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Unión Proteica , Dominios Proteicos , Proteolisis/efectos de los fármacos , Termodinámica , Factores de Transcripción/química , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/efectos de los fármacos
11.
J Lipid Res ; 51(5): 914-22, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20388923

RESUMEN

Ectopic expression of caveolin-1 in HEK293 cells enhances FA sequestration in membranes as measured by a pH-sensitive fluorescent dye (1). We hypothesized that sequestration of FA is due to the enrichment of caveolin in the cytosolic leaflet and its ability to facilitate the formation of lipid rafts to buffer high FA levels. Here we show that ec-topic expression of caveolin-3 also results in enhanced FA sequestration. To further discriminate the effect that caveolins have on transmembrane FA movement and distribution, we labeled the outer membrane leaflet with fluorescein-phosphatidylethanolamine (FPE), whose emission is quenched by the presence of FA anions. Real-time measurements made with FPE and control experiments with positively charged fatty amines support our hypothesis that caveolins promote localization of FA anions through interactions with basic amino acid residues (lysines and arginines) present at the C termini of caveolins-1 and -3.


Asunto(s)
Caveolinas/metabolismo , Membrana Celular/metabolismo , Citoplasma/metabolismo , Ácidos Grasos/metabolismo , Ácidos Grasos/toxicidad , Triglicéridos/biosíntesis , Aminas/química , Aminas/metabolismo , Caveolina 1/química , Caveolina 1/metabolismo , Caveolina 3/química , Caveolina 3/metabolismo , Caveolinas/química , Línea Celular , Relación Dosis-Respuesta a Droga , Espacio Extracelular/metabolismo , Fluoresceínas/metabolismo , Regulación de la Expresión Génica , Movimiento , Fosfatidiletanolaminas/metabolismo
12.
J Biol Chem ; 284(48): 33296-304, 2009 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19801636

RESUMEN

Abnormalities in the transport of saturated very long chain fatty acids (VLCFA; >C18:0) contribute to their toxic levels in peroxisomal disorders of fatty acid metabolism, such as adrenoleukodystrophy and adrenomyeloneuropathy. We previously showed that VLCFA desorb much slower than normal dietary fatty acids from both albumin and protein-free lipid bilayers. The important step of transbilayer movement (flip-flop) was not measured directly as a consequence of this very slow desorption from donors, and the extremely low aqueous solubility of VLCFA precludes addition of unbound VLCFA to lipid membranes. We have overcome these limitations using methyl-beta-cyclodextrin to solubilize VLCFA for rapid delivery to "acceptor" phosphatidylcholine vesicles (small and large unilamellar) and to cells. VLCFA binding was monitored in real time with the fluorescent probe fluorescein-labeled phosphatidylethanolamine in the outer membrane leaflet, and entrapped pyranine was used to detect flip-flop across the membrane. The upper limit of the rate of flip-flop across the membrane was independent of temperature and media viscosity and was similar for model raft and non-raft membranes as well as living cells. We further showed that cyclodextrins can extract VLCFA rapidly (within seconds) from vesicles and cells, which have implications for the mechanism and potential alternative approaches to treat adrenoleukodystrophy. Because VLCFA diffuse through the lipid bilayer, proteins may not be required for their transport across the peroxisomal membrane.


Asunto(s)
Ácidos Grasos/metabolismo , Membrana Dobles de Lípidos/metabolismo , beta-Ciclodextrinas/química , Adrenoleucodistrofia/metabolismo , Transporte Biológico , Línea Celular , Membrana Celular/química , Membrana Celular/metabolismo , Difusión , Ácidos Eicosanoicos/química , Ácidos Eicosanoicos/metabolismo , Ácidos Grasos/química , Colorantes Fluorescentes/química , Humanos , Membrana Dobles de Lípidos/química , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Espectrometría de Fluorescencia
13.
J Am Chem Soc ; 132(12): 4152-60, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20201574

RESUMEN

Targeting protein kinases with small organic molecules is a promising strategy to regulate unwanted kinase activity in both chemical biology and medicinal chemistry research. Traditionally, kinase inhibitors are identified in activity-based screening assays using enzymatically active kinase preparations to measure the perturbation of substrate phosphorylation, often resulting in the enrichment of classical ATP competitive (Type I) inhibitors. However, addressing enzymatically incompetent kinase conformations offers new opportunities for targeted therapies and is moving to the forefront of kinase inhibitor research. Here we report the development of a new FLiK (Fluorescent Labels in Kinases) binding assay to detect small molecules that induce changes in the conformation of the glycine-rich loop. Due to cross-talk between the glycine-rich loop and the activation loop in kinases, this alternative labeling approach can also detect ligands that stabilize inactive kinase conformations, including slow-binding Type II and Type III kinase inhibitors. Protein X-ray crystallography validated the assay results and identified a novel DFG-out binding mode for a quinazoline-based inhibitor in p38alpha kinase. We also detected the high-affinity binding of a clinically relevant and specific VEGFR2 inhibitor, and we provide structural details of its binding mode in p38alpha, in which it stabilizes the DFG-out conformation. Last, we demonstrate the power of this new FLiK labeling strategy to detect the binding of Type I ligands that induce conformational changes in the glycine-rich loop as a means of gaining affinity for the target kinase. This approach may be a useful alternative to develop direct binding assays for kinases that do not adopt the DFG-out conformation while also avoiding the use of expensive kits, detection reagents, or radioactivity frequently employed with activity-based assays.


Asunto(s)
Colorantes Fluorescentes/química , Glicina/química , Inhibidores de Proteínas Quinasas , Cristalografía por Rayos X , Glicina/antagonistas & inhibidores , Glicina/metabolismo , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
14.
Chembiochem ; 11(18): 2557-66, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21080395

RESUMEN

Targeting protein kinases in cancer therapy with irreversible small-molecule inhibitors is moving to the forefront of kinase-inhibitor research and is thought to be an effective means of overcoming mutation-associated drug resistance in epidermal growth factor receptor kinase (EGFR). We generated a detection technique that allows direct measurements of covalent bond formation without relying on kinase activity, thereby allowing the straightforward investigation of the influence of steric clashes on covalent inhibitors in different resistant kinase mutants. The obtained results are discussed together with structural biology and biochemical studies of catalytic activity in both wild-type and gatekeeper mutated kinase variants to draw conclusions about the impact of steric hindrance and increased catalytic activity in drug-resistant kinase variants.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Espectrometría de Fluorescencia/métodos , Animales , Pollos , Cristalografía por Rayos X , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Mutación , Neoplasias/tratamiento farmacológico , Proteínas Quinasas/química , Proteínas Quinasas/genética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/química , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
15.
Arch Pharm (Weinheim) ; 343(4): 193-206, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20336692

RESUMEN

The 512 protein kinases encoded by the human genome are a prime example of nature's ability to create diversity by introducing variations to a highly conserved theme. The activity of each kinase domain is controlled by layers of regulatory mechanisms involving different combinations of post-translational modifications, intramolecular contacts, and intermolecular interactions. Ultimately, they all achieve their effect by favoring particular conformations that promote or prevent the kinase domain from catalyzing protein phosphorylation. The central role of kinases in various diseases has encouraged extensive investigations of their biological function and three-dimensional structures, yielding a more detailed understanding of the mechanisms that regulate protein kinase activity by conformational changes. In the present review, we discuss these regulatory mechanisms and show how conformational changes can be exploited for the design of specific inhibitors that lock protein kinases in inactive conformations. In addition, we highlight recent developments to monitor ligand-induced structural changes in protein kinases and for screening and identifying inhibitors that stabilize enzymatically incompetent kinase conformations.


Asunto(s)
Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Regulación Alostérica , Animales , Dominio Catalítico , Estabilidad de Enzimas , Humanos , Ligandos , Modelos Moleculares , Estructura Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Relación Estructura-Actividad
16.
J Am Chem Soc ; 131(37): 13286-96, 2009 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-19572644

RESUMEN

Kinase disregulation disrupts the intricate network of intracellular signaling pathways and contributes to the onset of diseases such as cancer. Although several kinase inhibitors are on the market, inhibitor selectivity and drug resistance mutations persist as fundamental challenges in the development of effective long-term treatments. Chemical entities binding to less conserved allosteric sites would be expected to offer new opportunities for scaffold development. Because no high-throughput method was previously available, we developed a fluorescence-based kinase binding assay for identifying and characterizing ligands which stabilize the inactive kinase conformation. Here, we present a description of the development and validation of this assay using the serine/threonine kinase p38alpha. By covalently attaching fluorophores to the activation loop of the kinase, we were able to detect conformational changes and measure the K(d), k(on), and k(off) associated with the binding and dissociation of ligands to the allosteric pocket. We report the SAR of a synthesized focused library of pyrazolourea derivatives, a scaffold known to bind with high affinity to the allosteric pocket of p38alpha. Additionally, we used protein X-ray crystallography together with our assay to examine the binding and dissociation kinetics to characterize potent quinazoline- and quinoline-based type II inhibitors, which also utilize this binding pocket in p38alpha. Last, we identified the b-Raf inhibitor sorafenib as a potent low nanomolar inhibitor of p38alpha and used protein X-ray crystallography to confirm a unique binding mode to the inactive kinase conformation.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Colorantes Fluorescentes/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Bencenosulfonatos/farmacología , Unión Competitiva , Dominio Catalítico , Cristalografía por Rayos X , Activación Enzimática , Estabilidad de Enzimas , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Pirazoles/química , Pirazoles/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Sorafenib , Relación Estructura-Actividad , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
J Am Chem Soc ; 131(51): 18478-88, 2009 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-19950957

RESUMEN

Small molecule kinase inhibitors are an attractive means to modulate kinase activities in medicinal chemistry and chemical biology research. In the physiological setting of a cell, kinase function is orchestrated by a plethora of regulatory processes involving the structural transition of kinases between inactive and enzymatically competent conformations and vice versa. The development of novel kinase inhibitors is mainly fostered by high-throughput screening initiatives where the small molecule perturbation of the phosphorylation reaction is measured to identify inhibitors. Such setups require enzymatically active kinase preparations and present a risk of solely identifying classical ATP-competitive Type I inhibitors. Here we report the high-throughput screening of a library of approximately 35000 small organic molecules with an assay system that utilizes enzymatically inactive human p38alpha MAP kinase to detect stabilizers of the pharmacologically more desirable DFG-out conformation. We used protein X-ray crystallography to characterize the binding mode of hit compounds and reveal structural features which explain how these ligands stabilize and/or induce the DFG-out conformation. Lastly, we show that although some of the hit compounds were confirmed by protein X-ray crystallography, they were not detected in classic phosphorylation assays, thus validating the unique sensitivity of the assay system used in this study and highlighting the potential of screening with inactive kinase preparations.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Cristalografía por Rayos X , Estabilidad de Enzimas , Humanos , Ligandos , Proteína Quinasa 14 Activada por Mitógenos/química , Unión Proteica , Conformación Proteica , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
18.
Biophys J ; 94(11): 4493-503, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18296488

RESUMEN

Fatty acids (FA) are important nutrients that the body uses to regulate the storage and use of energy resources. The predominant mechanism by which long-chain fatty acids enter cells is still debated widely as it is unclear whether long-chain fatty acids require protein transporters to catalyze their transmembrane movement. We use stopped-flow fluorescence (millisecond time resolution) with three fluorescent probes to monitor different aspects of FA binding to phospholipid vesicles. In addition to acrylodan-labeled fatty acid binding protein, a probe that detects unbound FA in equilibrium with the lipid bilayer, and cis-parinaric acid, which detects the insertion of the FA acyl chain into the membrane, we introduce fluorescein-labeled phosphatidylethanolamine as a new probe to measure the binding of FA anions to the outer membrane leaflet. We combined these three approaches with measurement of intravesicular pH to show very fast FA binding and translocation in the same experiment. We validated quantitative predictions of our flip-flop model by measuring the number of H(+) delivered across the membrane by a single dose of FA with the probe 6-methoxy-N-(3-sulfopropyl) quinolinium. These studies provide a framework and basis for evaluation of the potential roles of proteins in binding and transport of FA in biological membranes.


Asunto(s)
Ácidos Grasos/química , Colorantes Fluorescentes/química , Membrana Dobles de Lípidos/química , Fosfolípidos/química , Espectrometría de Fluorescencia/métodos , Liposomas Unilamelares/química , Adsorción
19.
Biochemistry ; 47(35): 9081-9, 2008 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-18693753

RESUMEN

Fatty acids (FA) are known to diffuse (flip-flop) rapidly across protein-free phospholipid bilayers in their un-ionized form. However, whether flip-flop through the hydrophobic core of the bilayer or desorption from the membrane into the aqueous phase is the rate-limiting step in FA transport through membranes is still debated. The issue has remained unresolved in part by disagreements over whether some methods of adding FA create artifacts that lead to erroneous conclusions and in part by the lack of fluorescence methods to monitor each individual step. Here we study the kinetics of FA transfer from donors to phospholipid vesicles (small and large unilamellar vesicles) by a dual fluorescence approach that utilizes the probes fluorescein phosphatidylethanolamine (FPE) and pyranine. FPE detects the concentration of FA anions in the outer membrane leaflet, allowing a precise measurement of kinetics of FA adsorption or desorption. Our results showed that as soon as FPE detects adsorption of FA into the outer leaflet, pyranine detects its movement to the inner leaflet. We further demonstrated that (i) flip-flop for FA with 14-22 carbons is much faster than the rates of desorption and therefore cannot be the rate-limiting step of FA translocation across membranes; (ii) fluorescence changes detected by probes located on or in acceptor vesicles are dependent upon the method used to deliver the FA (i.e., uncomplexed, or complexed to albumin or phospholipid bilayers); however, (iii) transfer kinetics observed in the presence of different donors is rate-limited by the desorption of FA from the donor into the aqueous phase rather than by flip-flop.


Asunto(s)
Ácidos Grasos/química , Ácidos Grasos/metabolismo , Membranas/química , Membranas/metabolismo , Arilsulfonatos/química , Arilsulfonatos/metabolismo , Transporte Biológico , Cinética , Modelos Biológicos , Fosfatidiletanolaminas/metabolismo , Liposomas Unilamelares/metabolismo
20.
Bioorg Med Chem ; 16(7): 3482-8, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18316192

RESUMEN

Resistance to kinase-targeted cancer drugs has recently been linked to a single point mutation in the ATP binding site of the kinase. In EGFR, the crucial Thr790 gatekeeper residue is mutated to a Met and prevents reversible ATP competitive inhibitors from binding. Irreversible 4-(phenylamino)quinazolines have been shown to overcome this drug resistance and are currently in clinical trials. In order to obtain a detailed structural understanding of how irreversible inhibitors overcome drug resistance, we used Src kinase as a model system for drug resistant EGFR-T790M. We report the first crystal structure of a drug resistant kinase in complex with an irreversible inhibitor. This 4-(phenylamino)quinazoline inhibits wild type and drug resistant EGFR in vitro at low nM concentrations. The co-crystal structure of drug resistant cSrc-T338M kinase domain provides the structural basis of this activity.


Asunto(s)
Resistencia a Medicamentos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Cristalografía por Rayos X , Receptores ErbB/química , Receptores ErbB/genética , Clorhidrato de Erlotinib , Modelos Moleculares , Estructura Molecular , Mutación/genética , Inhibidores de Proteínas Quinasas/síntesis química , Quinazolinas/química , Quinazolinas/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA