Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
SLAS Discov ; 22(4): 433-439, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28328322

RESUMEN

Monoacylglycerol acyltransferase enzymes (MGAT1, MGAT2, and MGAT3) convert monoacylglycerol to diacylglycerol (DAG). MGAT1 and MGAT2 are both implicated in obesity-related metabolic diseases. Conventional MGAT enzyme assays use radioactive substrates, wherein the product of the MGAT-catalyzed reaction is usually resolved by time-consuming thin layer chromatography (TLC) analysis. Furthermore, microsomal membrane preparations typically contain endogenous diacylglycerol acyltransferase (DGAT) from the host cells, and these DGAT activities can further acylate DAG to form triglyceride (TG). Our mass spectrometry (liquid chromatography-tandem mass spectrometry, or LC/MS/MS) MGAT2 assay measures human recombinant MGAT2-catalyzed formation of didecanoyl-glycerol from 1-decanoyl-rac-glycerol and decanoyl-CoA, to produce predominantly 1,3-didecanoyl-glycerol. Unlike 1,2-DAG, 1,3-didecanoyl-glycerol is proved to be not susceptible to further acylation to TG. 1,3-Didecanoyl-glycerol product can be readily solubilized and directly subjected to high-throughput mass spectrometry (HTMS) without further extraction in a 384-well format. We also have established the LC/MS/MS MGAT activity assay in the intestinal microsomes from various species. Our assay is proved to be highly sensitive, and thus it allows measurement of endogenous MGAT activity in cell lysates and tissue preparations. The implementation of the HTMS MGAT activity assay has facilitated the robust screening and evaluation of MGAT inhibitors for the treatment of metabolic diseases.


Asunto(s)
Diglicéridos/análisis , Inhibidores Enzimáticos/análisis , Ensayos Analíticos de Alto Rendimiento , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , Acilcoenzima A/metabolismo , Animales , Chlorocebus aethiops , Cromatografía Liquida/métodos , Diglicéridos/antagonistas & inhibidores , Diglicéridos/biosíntesis , Perros , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Haplorrinos , Humanos , Intestinos/efectos de los fármacos , Intestinos/enzimología , Cinética , Ratones , Microsomas/efectos de los fármacos , Microsomas/enzimología , N-Acetilglucosaminiltransferasas/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Espectrometría de Masas en Tándem/métodos
2.
J Lipid Res ; 53(6): 1106-16, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22493088

RESUMEN

Diacylglycerol acyltransferase (DGAT) catalyzes the final step in triglyceride (TG) synthesis. There are two isoforms, DGAT1 and DGAT2, with distinct protein sequences and potentially different physiological functions. To date, the ability to determine clear functional differences between DGAT1 and DGAT2, especially with respect to hepatic TG synthesis, has been elusive. To dissect the roles of these two key enzymes, we pretreated HepG2 hepatoma cells with (13)C(3)-D(5)-glycerol or (13)C(18)-oleic acid, and profiled the major isotope-labeled TG species by liquid chromatography tandem mass spectrometry. Selective DGAT1 and DGAT2 inhibitors demonstrated that (13)C(3)-D(5)-glycerol-incorporated TG synthesis was mediated by DGAT2, not DGAT1. Conversely, (13)C(18)-oleoyl-incorporated TG synthesis was predominantly mediated by DGAT1. To trace hepatic TG synthesis and VLDL triglyceride (VLDL-TG) secretion in vivo, we administered D(5)-glycerol to mice and measured plasma levels of D(5)-glycerol-incorporated TG. Treatment with an antisense oligonucleotide (ASO) to DGAT2 led to a significant reduction in D(5)-glycerol incorporation into VLDL-TG. In contrast, the DGAT2 ASO had no effect on the incorporation of exogenously administered (13)C(18)-oleic acid into VLDL-TG. Thus, our results indicate that DGAT1 and DGAT2 mediate distinct hepatic functions: DGAT2 is primarily responsible for incorporating endogenously synthesized FAs into TG, whereas DGAT1 plays a greater role in esterifying exogenous FAs to glycerol.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/metabolismo , Pruebas de Enzimas/métodos , Glicerol/metabolismo , Hígado/enzimología , Ácido Oléico/metabolismo , Animales , Diacilglicerol O-Acetiltransferasa/antagonistas & inhibidores , Diacilglicerol O-Acetiltransferasa/genética , Inhibidores Enzimáticos/farmacología , Esterificación/efectos de los fármacos , Ácidos Grasos/biosíntesis , Ácidos Grasos/metabolismo , Células Hep G2 , Humanos , Marcaje Isotópico , Lipoproteínas VLDL/metabolismo , Masculino , Ratones , Oligonucleótidos Antisentido/genética , Triglicéridos/biosíntesis
3.
J Med Chem ; 53(22): 7979-91, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-21033679

RESUMEN

A fragment-based drug design paradigm has been successfully applied in the discovery of lead series of ketohexokinase inhibitors. The paradigm consists of three iterations of design, synthesis, and X-ray crystallographic screening to progress low molecular weight fragments to leadlike compounds. Applying electron density of fragments within the protein binding site as defined by X-ray crystallography, one can generate target specific leads without the use of affinity data. Our approach contrasts with most fragment-based drug design methodology where solution activity is a main design guide. Herein we describe the discovery of submicromolar ketohexokinase inhibitors with promising druglike properties.


Asunto(s)
Fructoquinasas/antagonistas & inhibidores , Indazoles/síntesis química , Modelos Moleculares , Piperidinas/síntesis química , Animales , Células CACO-2 , Permeabilidad de la Membrana Celular , Cristalografía por Rayos X , Electrones , Humanos , Técnicas In Vitro , Indazoles/química , Indazoles/farmacocinética , Masculino , Microsomas Hepáticos/metabolismo , Estructura Molecular , Piperidinas/química , Piperidinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
4.
Bioorg Med Chem Lett ; 18(16): 4615-9, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18653333

RESUMEN

Members of a novel class of 4-amino-6-arylamino-pyrimidine-5-carbaldehyde hydrazones were identified as potent dual ErbB-2/EGFR kinase inhibitors using concept-guided design approach. These compounds inhibited the growth of ErbB-2 over-expressing human tumor cell lines (BT474, N87, and SK-BR-3) in vitro. Compound 15 emerged as a key lead and showed significant ability to inhibit growth factor-induced receptor phosphorylation in SK-BR-3 cells (IC(50)=54 nM) and cellular proliferation in vitro (IC(50)=14, 58, and 58 nM for BT474, N87, and SK-BR-3 respectively). The X-ray co-crystal structure of EGFR with a close analog (17) was determined and validated our design rationale.


Asunto(s)
Química Farmacéutica/métodos , Receptores ErbB/antagonistas & inhibidores , Hidrazonas/química , Hidrazonas/síntesis química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/síntesis química , Receptor ErbB-2/antagonistas & inhibidores , Animales , Diseño de Fármacos , Humanos , Hidrazonas/farmacología , Concentración 50 Inhibidora , Modelos Químicos , Conformación Molecular , Oximas/química , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
5.
Bioorg Med Chem Lett ; 18(12): 3495-9, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18508264

RESUMEN

We herein disclose a novel series of 4-aminopyrimidine-5-carbaldehyde oximes that are potent and selective inhibitors of both EGFR and ErbB-2 tyrosine kinases, with IC(50) values in the nanomolar range. Structure-activity relationship (SAR) studies elucidated a critical role for the 4-amino and C-6 arylamino moieties. The X-ray co-crystal structure of EGFR with 37 was determined and validated our design rationale.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/química , Oximas/farmacología , Pirimidinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Sitios de Unión/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Enlace de Hidrógeno , Concentración 50 Inhibidora , Modelos Moleculares , Estructura Molecular , Oximas/síntesis química , Oximas/química , Pirimidinas/síntesis química , Pirimidinas/química , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad
6.
J Biomol Screen ; 12(3): 418-28, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17438070

RESUMEN

The reliable production of large amounts of stable, high-quality proteins is a major challenge facing pharmaceutical protein biochemists, necessary for fulfilling demands from structural biology, for high-throughput screening, and for assay purposes throughout early discovery. One strategy for bypassing purification challenges in problematic systems is to engineer multiple forms of a particular protein to optimize expression, purification, and stability, often resulting in a nonphysiological sub-domain. An alternative strategy is to alter process conditions to maximize wild-type construct stability, based on a specific protein stability profile (PSP). ThermoFluor, a miniaturized 384-well thermal stability assay, has been implemented as a means of monitoring solution-dependent changes in protein stability, complementing the protein engineering and purification processes. A systematic analysis of pH, buffer or salt identity and concentration, biological metals, surfactants, and common excipients in terms of an effect on protein stability rapidly identifies conditions that might be used (or avoided) during protein production. Two PSPs are presented for the kinase catalytic domains of Akt-3 and cFMS, in which information derived from a ThermoFluor PSP led to an altered purification strategy, improving the yield and quality of the protein using the primary sequences of the catalytic domains.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/química , Receptor de Factor Estimulante de Colonias de Macrófagos/biosíntesis , Receptor de Factor Estimulante de Colonias de Macrófagos/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Tampones (Química) , Fluorescencia , Concentración de Iones de Hidrógeno , Metales/farmacología , Concentración Osmolar , Estructura Cuaternaria de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/aislamiento & purificación , Receptor de Factor Estimulante de Colonias de Macrófagos/aislamiento & purificación , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/normas , Sales (Química)/farmacología , Soluciones/farmacología , Termodinámica
7.
J Biol Chem ; 282(6): 4094-101, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17132624

RESUMEN

The cFMS proto-oncogene encodes for the colony-stimulating factor-1 receptor, a receptor-tyrosine kinase responsible for the differentiation and maturation of certain macrophages. Upon binding its ligand colony-stimulating factor-1 cFMS autophosphorylates, dimerizes, and induces phosphorylation of downstream targets. We report the novel crystal structure of unphosphorylated cFMS in complex with two members of different classes of drug-like protein kinase inhibitors. cFMS exhibits a typical bi-lobal kinase fold, and its activation loop and DFG motif are found to be in the canonical inactive conformation. Both ATP competitive inhibitors are bound in the active site and demonstrate a binding mode similar to that of STI-571 bound to cABL. The DFG motif is prevented from switching into the catalytically competent conformation through interactions with the inhibitors. Activation of cFMS is also inhibited by the juxtamembrane domain, which interacts with residues of the active site and prevents formation of the activated kinase. Together the structures of cFMS provide further insight into the autoinhibition of receptor-tyrosine kinases via their respective juxtamembrane domains; additionally the binding mode of two novel classes of kinase inhibitors will guide the design of novel molecules targeting macrophage-related diseases.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/química , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/química , Amidas/química , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/antagonistas & inhibidores , Proteínas Mutantes Quiméricas/química , Estructura Terciaria de Proteína/genética , Proto-Oncogenes Mas , Quinolonas/química , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptor TIE-2/química , Receptor TIE-2/genética , Receptores de Factores de Crecimiento de Fibroblastos/química , Receptores de Factores de Crecimiento de Fibroblastos/genética
8.
J Biol Chem ; 282(6): 4085-93, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17132625

RESUMEN

A parallel approach to designing crystallization constructs for the c-FMS kinase domain was implemented, resulting in proteins suitable for structural studies. Sequence alignment and limited proteolysis were used to identify and eliminate unstructured and surface-exposed domains. A small library of chimeras was prepared in which the kinase insert domain of FMS was replaced with the kinase insert domain of previously crystallized receptor-tyrosine kinases. Characterization of the newly generated FMS constructs by enzymology and thermoshift assays demonstrated similar activities and compound binding to the FMS full-length cytoplasmic domain. Two chimeras were evaluated for crystallization in the presence and absence of a variety of ligands resulting in crystal structures, and leading to a successful structure-based drug design project for this important inflammation target.


Asunto(s)
Ingeniería de Proteínas , Proteínas Tirosina Quinasas Receptoras/síntesis química , Proteínas Tirosina Quinasas Receptoras/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/química , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Secuencia de Aminoácidos , Animales , Células Cultivadas , Cristalización , Citoplasma/química , Citoplasma/genética , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/síntesis química , Proteínas Mutantes Quiméricas/genética , Inhibidores de Proteínas Quinasas/química , Estructura Terciaria de Proteína/genética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Alineación de Secuencia , Spodoptera
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...