Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nat Chem Biol ; 13(3): 317-324, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28114273

RESUMEN

Protein lysine methyltransferases (PKMTs) regulate diverse physiological processes including transcription and the maintenance of genomic integrity. Genetic studies suggest that the PKMTs SUV420H1 and SUV420H2 facilitate proficient nonhomologous end-joining (NHEJ)-directed DNA repair by catalyzing the di- and trimethylation (me2 and me3, respectively) of lysine 20 on histone 4 (H4K20). Here we report the identification of A-196, a potent and selective inhibitor of SUV420H1 and SUV420H2. Biochemical and co-crystallization analyses demonstrate that A-196 is a substrate-competitive inhibitor of both SUV4-20 enzymes. In cells, A-196 induced a global decrease in H4K20me2 and H4K20me3 and a concomitant increase in H4K20me1. A-196 inhibited 53BP1 foci formation upon ionizing radiation and reduced NHEJ-mediated DNA-break repair but did not affect homology-directed repair. These results demonstrate the role of SUV4-20 enzymatic activity in H4K20 methylation and DNA repair. A-196 represents a first-in-class chemical probe of SUV4-20 to investigate the role of histone methyltransferases in genomic integrity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Inestabilidad Genómica/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Línea Celular Tumoral , Cristalografía por Rayos X , Reparación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/química , Compuestos Heterocíclicos de 4 o más Anillos/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación/efectos de los fármacos , Modelos Moleculares , Estructura Molecular
2.
Nat Chem Biol ; 13(4): 389-395, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28135237

RESUMEN

Polycomb repressive complex 2 (PRC2) is a regulator of epigenetic states required for development and homeostasis. PRC2 trimethylates histone H3 at lysine 27 (H3K27me3), which leads to gene silencing, and is dysregulated in many cancers. The embryonic ectoderm development (EED) protein is an essential subunit of PRC2 that has both a scaffolding function and an H3K27me3-binding function. Here we report the identification of A-395, a potent antagonist of the H3K27me3 binding functions of EED. Structural studies demonstrate that A-395 binds to EED in the H3K27me3-binding pocket, thereby preventing allosteric activation of the catalytic activity of PRC2. Phenotypic effects observed in vitro and in vivo are similar to those of known PRC2 enzymatic inhibitors; however, A-395 retains potent activity against cell lines resistant to the catalytic inhibitors. A-395 represents a first-in-class antagonist of PRC2 protein-protein interactions (PPI) for use as a chemical probe to investigate the roles of EED-containing protein complexes.


Asunto(s)
Antineoplásicos/farmacología , Indanos/farmacología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Sulfonamidas/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indanos/química , Modelos Moleculares , Estructura Molecular , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/metabolismo , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad , Sulfonamidas/química , Células Tumorales Cultivadas
3.
J Infect Dis ; 209(2): 275-84, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24123773

RESUMEN

Malaria parasites are transmitted by mosquitoes, and blocking parasite transmission is critical in reducing or eliminating malaria in endemic regions. Here, we report the pharmacological characterization of a new class of malaria transmission-blocking compounds that acts via the inhibition of Plasmodia CDPK4 enzyme. We demonstrate that these compounds achieved selectivity over mammalian kinases by capitalizing on a small serine gatekeeper residue in the active site of the Plasmodium CDPK4 enzyme. To directly confirm the mechanism of action of these compounds, we generated P. falciparum parasites that express a drug-resistant methionine gatekeeper (S147 M) CDPK4 mutant. Mutant parasites showed a shift in exflagellation EC50 relative to the wild-type strains in the presence of compound 1294, providing chemical-genetic evidence that CDPK4 is the target of the compound. Pharmacokinetic analyses suggest that coformulation of this transmission-blocking agent with asexual stage antimalarials such as artemisinin combination therapy (ACT) is a promising option for drug delivery that may reduce transmission of malaria including drug-resistant strains. Ongoing studies include refining the compounds to improve efficacy and toxicological properties for efficient blocking of malaria transmission.


Asunto(s)
Antimaláricos/metabolismo , Inhibidores Enzimáticos/metabolismo , Plasmodium falciparum/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Antimaláricos/aislamiento & purificación , Antimaláricos/farmacocinética , Inhibidores Enzimáticos/aislamiento & purificación , Inhibidores Enzimáticos/farmacocinética , Flagelos/efectos de los fármacos , Flagelos/fisiología , Ratones , Plasmodium falciparum/fisiología
5.
J Med Chem ; 64(1): 417-429, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33378180

RESUMEN

Tumor necrosis factor α (TNFα) is a soluble cytokine that is directly involved in systemic inflammation through the regulation of the intracellular NF-κB and MAPK signaling pathways. The development of biologic drugs that inhibit TNFα has led to improved clinical outcomes for patients with rheumatoid arthritis and other chronic autoimmune diseases; however, TNFα has proven to be difficult to drug with small molecules. Herein, we present a two-phase, fragment-based drug discovery (FBDD) effort in which we first identified isoquinoline fragments that disrupt TNFα ligand-receptor binding through an allosteric desymmetrization mechanism as observed in high-resolution crystal structures. The second phase of discovery focused on the de novo design and optimization of fragments with improved binding efficiency and drug-like properties. The 3-indolinone-based lead presented here displays oral, in vivo efficacy in a mouse glucose-6-phosphate isomerase (GPI)-induced paw swelling model comparable to that seen with a TNFα antibody.


Asunto(s)
Productos Biológicos/síntesis química , Diseño de Fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Administración Oral , Regulación Alostérica , Animales , Artritis Reumatoide/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Ligandos , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
6.
Methods Mol Biol ; 1888: 45-71, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30519940

RESUMEN

Intracellular target affinity and residence time are fundamental aspects of pharmacological mechanism (Lu and Tonge, Curr Opin Chem Biol 14:467-474, 2010). Although various robust biochemical approaches exist to measure these binding characteristics, analysis of compound binding with isolated targets may not accurately reflect engagement in the milieu of living cells. To realize the influence of cellular context, methods are needed that are capable of quantifying affinity and residence time in the presence of the intracellular factors that may impact target engagement. Bioluminescence resonance energy transfer (BRET) offers a solution for intracellular target engagement when quantitative metrics or kinetic analyses are required.


Asunto(s)
Descubrimiento de Drogas/métodos , Transferencia Resonante de Energía de Fluorescencia , Mediciones Luminiscentes , Técnicas de Cultivo de Célula , Línea Celular , Transferencia Resonante de Energía de Fluorescencia/métodos , Ensayos Analíticos de Alto Rendimiento , Humanos , Mediciones Luminiscentes/métodos , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Permeabilidad , Reproducibilidad de los Resultados
7.
J Med Chem ; 61(19): 8504-8535, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-29718665

RESUMEN

Small-molecule (SM) leads in the early drug discovery pipeline are progressed primarily based on potency against the intended target(s) and selectivity against a very narrow slice of the proteome. So, why is there a tendency to wait until SMs are matured before probing for a deeper mechanistic understanding? For one, there is a concern about the interpretation of complex -omic data outputs and the resources needed to test these hypotheses. However, with recent advances in broad endpoint profiling assays that have companion reference databases and refined technology integration strategies, we argue that data complexity can translate into meaningful decision-making. This same strategy can also prioritize phenotypic screening hits to increase the likelihood of accessing unprecedented target space. In this Perspective. we will highlight a cohesive process that supports SM hit prosecution, providing a data-driven rationale and a suite of methods for direct identification of SM targets driving relevant biological end points.


Asunto(s)
Descubrimiento de Drogas , Proteoma/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Ensayos Analíticos de Alto Rendimiento , Humanos
8.
J Biomol Screen ; 11(7): 743-54, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16973923

RESUMEN

The authors describe the discovery of a new class of inhibitors to an essential Streptococcus pneumoniae cell wall biosyn-thesis enzyme, MurF, by a novel affinity screening method. The strategy involved screening very large mixtures of diverse small organic molecules against the protein target on the basis of equilibrium binding, followed by iterative ultrafiltration steps and ligand identification by mass spectrometry. Hits from any affinity-based screening method often can be relatively nonselective ligands, sometimes referred to as "nuisance" or "promiscuous" compounds. Ligands selective in their binding affinity for the MurF target were readily identified through electronic subtraction of an empirically determined subset of promiscuous compounds in the library without subsequent selectivity panels. The complete strategy for discovery and identification of novel specific ligands can be applied to all soluble protein targets and a wide variety of ligand libraries.


Asunto(s)
Antibacterianos/análisis , Antibacterianos/farmacología , Pared Celular/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Pruebas de Sensibilidad Microbiana , Péptido Sintasas/antagonistas & inhibidores , Streptococcus pneumoniae/enzimología , Antibacterianos/química , Espectrometría de Masas
9.
J Biomol Screen ; 11(7): 755-64, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16956998

RESUMEN

Kinase enzymes are involved in a vast array of biological processes associated with human disease; therefore, selective kinase inhibition by small molecules and therapeutic antibodies is an area of intense study. The authors show that drug candidates with immediate value for biological preclinical evaluation can be identified directly through ultra-efficient affinity screening of kinase enzymes and random compound mixtures. The screening process comprises sampling and trapping equilibrium binding between candidate ligands and protein in solution, followed by removal of unbound ligands via 3 rounds of ultrafiltration and direct identification of bound ligands by mass spectrometry. Evaluation of significant peaks is facilitated by automated integration and collation of the mass spectral data and import into custom software for analysis. One Chk1-selective ligand found by using this process is presented in detail. The compound is potent in both enzymatic and Chk1-dependent cellular assays, and specific contacts in the Chk1 active site are shown by X-ray crystallography.


Asunto(s)
Daño del ADN , Evaluación Preclínica de Medicamentos/métodos , Espectrometría de Masas/métodos , Inhibidores de Proteínas Quinasas/análisis , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Doxorrubicina/farmacología , Fase G2/efectos de los fármacos , Humanos , Ligandos , Datos de Secuencia Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Quinasas/aislamiento & purificación
10.
Curr Opin Drug Discov Devel ; 7(4): 411-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15338950

RESUMEN

Contemporary, rational small-molecule lead discovery methods, comprising target identification, assay development, high-throughput screening (HTS), hit characterization and medicinal chemistry optimization, dominate early-stage drug discovery strategies in many pharmaceutical companies. There is a growing disparity between the increasing cost of funding these methods and the decreasing number of new drugs reaching the market. New strategies must be adopted to reverse this trend. The use of genomics- and proteomics-based target discovery efforts can aid the process by dramatically increasing the number of novel, more highly validated targets entering the discovery process, but HTS must meet this increased demand with faster, cheaper technologies. Although activity-based screening strategies are typically efficient, allowing one scientist to interrogate tens of thousands of compounds per day, affinity-based screening strategies can allow much greater efficiency in the overall process. Affinity-based methods can play a role in both facilitating the screening of a greater number of targets and in efficiently characterizing the primary hits discovered.


Asunto(s)
Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Tecnología Farmacéutica/métodos , Evaluación Preclínica de Medicamentos/economía , Evaluación Preclínica de Medicamentos/tendencias , Tecnología Farmacéutica/economía , Tecnología Farmacéutica/tendencias
11.
J Med Chem ; 47(20): 4851-7, 2004 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-15369388

RESUMEN

Reversal of aberrant gene expression that is induced by the proto-oncogene c-myc is likely to be effective for treating a variety of tumors that rely on this pathway for growth. One strategy to down-regulate the c-myc pathway is to target transcription factors that regulate its own expression. A host of proteins act in coordination to regulate c-myc expression and any one of them are theoretical targets for small-molecule therapy. Experimentally, it has been shown that the far upstream element (FUSE) binding protein (FBP) is essential for c-myc expression, and reductions in FBP levels both reduce c-myc expression and correlate with slower cell growth. FBP binds to ssDNA by capturing exposed DNA bases in a hydrophobic pocket. This suggests that a small molecule could be designed to occupy this pocket and inhibit FBP function. Using a variety of screening methodologies, we have identified ligands that bind to the DNA binding pockets of the KH domains of FBP. Gel shift analyses using full length FBP and a related transcription factor confirm that a small-molecule lead compound inhibits DNA binding in a specific manner. The benzoylanthranilic acid compounds described here represent leads in the design of FBP inhibitors that can serve as useful tools in the study of c-myc regulation and in the development of therapeutics that target the c-myc pathway.


Asunto(s)
Técnicas Químicas Combinatorias/métodos , Proteínas de Unión al ADN/antagonistas & inhibidores , Genes myc , Espectroscopía de Resonancia Magnética , Regiones Promotoras Genéticas , Sitios de Unión , ADN Helicasas , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Ligandos , Modelos Moleculares , Conformación Proteica , Estructura Terciaria de Proteína , Proto-Oncogenes Mas , Proteínas de Unión al ARN , Secuencias Repetitivas de Aminoácido , Relación Estructura-Actividad
12.
ACS Med Chem Lett ; 5(1): 40-44, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24494061

RESUMEN

5-Aminopyrazole-4-carboxamide was used as an alternative scaffold to substitute for the pyrazolopyrimidine of a known "bumped kinase inhibitor" to create selective inhibitors of calcium-dependent protein kinase-1 from both Toxoplasma gondii and Cryptosporidium parvum. Compounds with low nanomolar inhibitory potencies against the target enzymes were obtained. The most selective inhibitors also exhibited submicromolar activities in T. gondii cell proliferation assays and were shown to be non-toxic to mammalian cells.

13.
J Biomol Screen ; 17(8): 1005-17, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22706350

RESUMEN

Efficient elucidation of the biological mechanism of action of novel compounds remains a major bottleneck in the drug discovery process. To address this need in the area of oncology, we report the development of a multiparametric high-content screening assay panel at the level of single cells to dramatically accelerate understanding the mechanism of action of cell growth-inhibiting compounds on a large scale. Our approach is based on measuring 10 established end points associated with mitochondrial apoptosis, cell cycle disruption, DNA damage, and cellular morphological changes in the same experiment, across three multiparametric assays. The data from all of the measurements taken together are expected to help increase our current understanding of target protein functions, constrain the list of possible targets for compounds identified using phenotypic screens, and identify off-target effects. We have also developed novel data visualization and phenotypic classification approaches for detailed interpretation of individual compound effects and navigation of large collections of multiparametric cellular responses. We expect this general approach to be valuable for drug discovery across multiple therapeutic areas.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Microscopía Fluorescente/métodos , Mitocondrias/efectos de los fármacos
14.
ACS Chem Biol ; 6(3): 234-44, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21090814

RESUMEN

Inhibition of protein kinases has validated therapeutic utility for cancer, with at least seven kinase inhibitor drugs on the market. Protein kinase inhibition also has significant potential for a variety of other diseases, including diabetes, pain, cognition, and chronic inflammatory and immunologic diseases. However, as the vast majority of current approaches to kinase inhibition target the highly conserved ATP-binding site, the use of kinase inhibitors in treating nononcology diseases may require great selectivity for the target kinase. As protein kinases are signal transducers that are involved in binding to a variety of other proteins, targeting alternative, less conserved sites on the protein may provide an avenue for greater selectivity. Here we report an affinity-based, high-throughput screening technique that allows nonbiased interrogation of small molecule libraries for binding to all exposed sites on a protein surface. This approach was used to screen both the c-Jun N-terminal protein kinase Jnk-1 (involved in insulin signaling) and p38α (involved in the formation of TNFα and other cytokines). In addition to canonical ATP-site ligands, compounds were identified that bind to novel allosteric sites. The nature, biological relevance, and mode of binding of these ligands were extensively characterized using two-dimensional (1)H/(13)C NMR spectroscopy, protein X-ray crystallography, surface plasmon resonance, and direct enzymatic activity and activation cascade assays. Jnk-1 and p38α both belong to the MAP kinase family, and the allosteric ligands for both targets bind similarly on a ledge of the protein surface exposed by the MAP insertion present in the CMGC family of protein kinases and distant from the active site. Medicinal chemistry studies resulted in an improved Jnk-1 ligand able to increase adiponectin secretion in human adipocytes and increase insulin-induced protein kinase PKB phosphorylation in human hepatocytes, in similar fashion to Jnk-1 siRNA and to rosiglitazone treatment. Together, the data suggest that these new ligand series bind to a novel, allosteric, and physiologically relevant site and therefore represent a unique approach to identify kinase inhibitors.


Asunto(s)
Descubrimiento de Drogas , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteína Quinasa 8 Activada por Mitógenos/química , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Bibliotecas de Moléculas Pequeñas , Estereoisomerismo , Relación Estructura-Actividad , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Bioorg Med Chem Lett ; 17(11): 3122-9, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17391963

RESUMEN

Survivin is one of the most tumor-specific genes in the human genome and is an attractive target for cancer therapy. However, small-molecule ligands for survivin have not yet been described. Thus, an interrogation of survivin which could potentially both validate a small-molecule therapy approach, and determine the biochemical nature of any of survivin's functions has not been possible. Here we describe the discovery and characterization of a small molecule binding site on the survivin surface distinct from the Smac peptide-binding site. The new site is located at the dimer interface and exhibits many of the features of highly druggable, biologically relevant protein binding sites. A variety of small hydrophobic compounds were found that bind with moderate affinity to this binding site, from which one lead was developed into a group of compounds with nanomolar affinity. Additionally, a subset of these compounds are adequately water-soluble and cell-permeable. Thus, the structural studies and small molecules described here provide tools that can be used to probe the biochemical role(s) of survivin, and may ultimately serve as a basis for the development of small molecule therapeutics acting via direct or allosteric disruption of binding events related to this poorly understood target.


Asunto(s)
Inhibidores de Cisteína Proteinasa/química , Proteínas Asociadas a Microtúbulos/química , Sondas Moleculares/química , Proteínas de Neoplasias/química , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Inhibidoras de la Apoptosis , Ligandos , Conformación Proteica , Survivin
16.
Chem Biol Drug Des ; 69(6): 395-404, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17581233

RESUMEN

As part of a fully integrated and comprehensive strategy to discover novel antibacterial agents, NMR- and mass spectrometry-based affinity selection screens were performed to identify compounds that bind to protein targets uniquely found in bacteria and encoded by genes essential for microbial viability. A biphenyl acid lead series emerged from an NMR-based screen with the Haemophilus influenzae protein HI0065, a member of a family of probable ATP-binding proteins found exclusively in eubacteria. The structure-activity relationships developed around the NMR-derived biphenyl acid lead were consistent with on-target antibacterial activity as the Staphylococcus aureus antibacterial activity of the series correlated extremely well with binding affinity to HI0065, while the correlation of binding affinity with B-cell cytotoxicity was relatively poor. Although further studies are needed to conclusively establish the mode of action of the biphenyl series, these compounds represent novel leads that can serve as the basis for the development of novel antibacterial agents that appear to work via an unprecedented mechanism of action. Overall, these results support the genomics-driven hypothesis that targeting bacterial essential gene products that are not present in eukaryotic cells can identify novel antibacterial agents.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Antibacterianos/química , Proteínas Bacterianas/metabolismo , Química Farmacéutica/métodos , Haemophilus influenzae/metabolismo , Secuencia de Aminoácidos , Animales , Linfocitos B/metabolismo , Diseño de Fármacos , Genoma Bacteriano , Genómica , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Datos de Secuencia Molecular , Unión Proteica , Relación Estructura-Actividad
17.
Bioorg Med Chem Lett ; 16(13): 3574-7, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16632353

RESUMEN

We have screened molecules for inhibition of MetAP2 as a novel approach toward antiangiogenesis and anticancer therapy using affinity selection/mass spectrometry (ASMS) employing MetAP2 loaded with Mn(2+) as the active site metal. After a series of anthranilic acid sulfonamides with micromolar affinities was identified, chemistry efforts were initiated. The micromolar hits were quickly improved to potent nanomolar inhibitors by chemical modifications guided by insights from X-ray crystallography.


Asunto(s)
Aminopeptidasas/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Glicoproteínas/antagonistas & inhibidores , Sulfonamidas/farmacología , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Sitios de Unión , Línea Celular , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Manganeso/química , Espectrometría de Masas/métodos , Metionil Aminopeptidasas , Modelos Moleculares , Estructura Molecular , Sensibilidad y Especificidad , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química
18.
Anal Biochem ; 328(2): 131-8, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15113688

RESUMEN

Bcl-xL is a member of the Bcl-2 family of proteins that are implicated to play a vital role in several diseases including cancer. Bcl-xL suppresses apoptosis; thus the inhibition of Bcl-xL function could restore the apoptotic process. To identify antagonists of Bcl-xL function, two ultra-high-throughput screens were implemented. An activity assay utilized fluorescence polarization, based on the binding of fluorescein-labeled peptide [the BH3 domain of BAD protein (F-Bad 6)] to Bcl-xL. A 384-well plate assay with mixtures of 10 drug compounds per well, combined with a fast plate reader, resulted in a throughput of 46,080 data points/day. Utilizing this screening format, 370,400 compounds were screened in duplicate and 425 inhibitors with an IC(50) below 100 microM were identified. The second assay format, affinity selection/mass spectrometry (ASMS), used ultrafiltration to separate Bcl-xL binders from nonbinders in mixtures of 2400 compounds. The bound species were subsequently separated from the protein and analyzed by flow injection electrospray mass spectrometry. Utilizing the ASMS format, 263,382 compounds were screened in duplicate and 29 binders with affinities below 100 microM were identified. Two novel classes of Bcl-xL inhibitors were identified by both methods and confirmed to bind (13)C-labeled Bcl-xL using heteronuclear magnetic resonance spectroscopy.


Asunto(s)
Técnicas Químicas Combinatorias/métodos , Preparaciones Farmacéuticas/análisis , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Apoptosis/genética , Apoptosis/fisiología , Unión Competitiva , Proteínas Portadoras/análisis , Proteínas Portadoras/metabolismo , Dimetilsulfóxido/química , Polarización de Fluorescencia , Concentración 50 Inhibidora , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Volumetría , Proteína bcl-X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA