Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Bioorg Med Chem Lett ; 22(11): 3795-9, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22546671

RESUMEN

The inhibition of hH-PGDS has been proposed as a potential target for the development of anti-allergic and anti-inflammatory drugs. Herein we describe our investigation of the binding pocket of this important enzyme and our observation that two water molecules bind to our inhibitors and the enzyme. A series of compounds were prepared to the probe the importance of the water molecules in determining the binding affinity of the inhibitors to the enzyme. The study provides insight into the binding requirements for the design of potent hH-PGDS inhibitors.


Asunto(s)
Antialérgicos/química , Antiinflamatorios/química , Inhibidores Enzimáticos/química , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Lipocalinas/antagonistas & inhibidores , Agua/química , Antialérgicos/síntesis química , Antiinflamatorios/síntesis química , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Inhibidores Enzimáticos/síntesis química , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Isoquinolinas/química , Lipocalinas/metabolismo , Naftalenos/química , Estructura Terciaria de Proteína
2.
Artículo en Inglés | MEDLINE | ID: mdl-22684055

RESUMEN

The role of ADAM-8 in cancer and inflammatory diseases such as allergy, arthritis and asthma makes it an attractive target for drug development. Therefore, the catalytic domain of human ADAM-8 was expressed, purified and crystallized in complex with a hydroxamic acid inhibitor, batimastat. The crystal structure of the enzyme-inhibitor complex was refined to 2.1 Å resolution. ADAM-8 has an overall fold similar to those of other ADAM members, including a central five-stranded ß-sheet and a catalytic Zn(2+) ion. However, unique differences within the S1' binding loop of ADAM-8 are observed which might be exploited to confer specificity and selectivity to ADAM-8 competitive inhibitors for the treatment of diseases involving this enzyme.


Asunto(s)
Proteínas ADAM/química , Dominio Catalítico , Proteínas de la Membrana/química , Fenilalanina/análogos & derivados , Inhibidores de Proteasas/química , Tiofenos/química , Proteínas ADAM/metabolismo , Humanos , Ligandos , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Fenilalanina/química , Fenilalanina/metabolismo , Inhibidores de Proteasas/metabolismo , Unión Proteica , Desplegamiento Proteico , Tiofenos/metabolismo
3.
Biochem Biophys Res Commun ; 396(2): 543-8, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20435017

RESUMEN

The Janus kinase family consists of four members: JAK-1, -2, -3 and TYK-2. While JAK-2 and JAK-3 have been well characterized biochemically, there is little data on TYK-2. Recent work suggests that TYK-2 may play a critical role in the development of a number of inflammatory processes. We have carried out a series of biochemical studies to better understand TYK-2 enzymology and its inhibition profile, in particular how the TYK-2 phosphorylated forms differ from each other and from the other JAK family members. We have expressed and purified milligram quantities of the TYK-2 kinase domain (KD) to high purity and developed a method to separate the non-, mono- (pY(1054)) and di-phosphorylated forms of the enzyme. Kinetic studies (k(cat(app))/K(m(app))) indicated that phosphorylation of the TYK-2-KD (pY(1054)) increased the catalytic efficiency 4.4-fold compared to its non-phosphorylated form, while further phosphorylation to generate the di-phosphorylated enzyme imparted no further increase in activity. These results are in contrast to those obtained with the JAK-2-KD and JAK-3-KD, where little or no increase in activity occurred upon mono-phosphorylation, while di-phosphorylation resulted in a 5.1-fold increase in activity for the JAK-2-KD. Moreover, ATP-competitive inhibitors demonstrated 10-30-fold shifts in potency (K(i(app))) as a result of the TYK-2-KD phosphorylation state, while the shifts for JAK-3-KD were only 2-3-fold and showed little or no change for JAK-2-KD. Thus, the phosphorlyation state imparted differential effects on both activity and inhibition within the JAK family of kinases.


Asunto(s)
Janus Quinasa 2/biosíntesis , Janus Quinasa 2/aislamiento & purificación , Janus Quinasa 3/biosíntesis , Janus Quinasa 3/aislamiento & purificación , TYK2 Quinasa/biosíntesis , TYK2 Quinasa/aislamiento & purificación , Animales , Catálisis , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 3/antagonistas & inhibidores , Ratones , Fosforilación , Estructura Terciaria de Proteína , TYK2 Quinasa/antagonistas & inhibidores
4.
Protein Expr Purif ; 69(1): 54-63, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19781647

RESUMEN

Janus-associated kinases (JAKs) play critical roles in cytokine signaling, and have emerged as viable therapeutic targets in inflammation and oncology related diseases. To date, targeting JAK proteins with highly selective inhibitor compounds have remained elusive. We have expressed the active kinase domains for both JAK2 and JAK3 and devised purification protocols to resolve the non-, mono- (Y1007) and diphosphorylated (Y1007 and Y1008) states of JAK2 and non- and monophosphorylated states of JAK3 (Y980). An optimal purified protein yield of 20, 29 and 69mg per 20L cell culture was obtained for the three JAK2 forms, respectively, and 12.2 and 2.3mg per 10L fermentation for the two JAK3 forms allowing detailed biochemical and biophysical studies. To monitor the purification process we developed a novel HPLC activity assay where a sequential order of phosphorylation was observed whereby the first tyrosine residue was completely phosphorylated prior to phosphorylation of the tandem tyrosine residue. A Caliper-based microfluidics assay was used to determine the kinetic parameters (K(m) and k(cat)) for each phosphorylated state, showing that monophosphorylated (Y1007) JAK2 enzyme activity increased 9-fold over that of the nonphosphorylated species, and increased an additional 6-fold for the diphosphorylated (Y1007/Y1008) species, while phosphorylation of JAK3 resulted in a negligible increase in activity. Moreover, crystal structures have been generated for each isolated state of JAK2 and JAK3 with resolutions better than 2.4A. The generation of these reagents has enabled kinetic and structural characterization to inform the design of potent and selective inhibitors of the JAK family.


Asunto(s)
Janus Quinasa 2/química , Janus Quinasa 2/aislamiento & purificación , Janus Quinasa 3/química , Janus Quinasa 3/aislamiento & purificación , Secuencia de Aminoácidos , Biocatálisis , Cromatografía Líquida de Alta Presión , Cristalización , Electroforesis en Gel de Poliacrilamida , Fermentación , Humanos , Cinética , Datos de Secuencia Molecular , Fosforilación , Estructura Terciaria de Proteína
5.
Artículo en Inglés | MEDLINE | ID: mdl-20823544

RESUMEN

Rat autotaxin has been cloned, expressed, purified to homogeneity and crystallized via hanging-drop vapour diffusion using PEG 3350 as precipitant and ammonium iodide and sodium thiocyanate as salts. The crystals diffracted to a maximum resolution of 2.05 A and belonged to space group P1, with unit-cell parameters a=53.8, b=63.3, c=70.5 A, alpha=98.8, beta=106.2, gamma=99.8 degrees. Preliminary X-ray diffraction analysis indicated the presence of one molecule per asymmetric unit, with a solvent content of 47%.


Asunto(s)
Hidrolasas Diéster Fosfóricas/química , Pirofosfatasas/química , Animales , Cristalización , Cristalografía por Rayos X , Ratas
6.
Protein Expr Purif ; 65(2): 133-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19189860

RESUMEN

Soluble guanylate cyclase (sGC) has been purified from 100 L cell culture infected by baculovirus using the newer and highly effective titerless infected-cells preservation and scale-up (TIPS) method. Successive passage of the enzyme through DEAE, Ni(2+)-NTA, and POROS Q columns obtained approximately 100mg of protein. The sGC obtained by this procedure was already about 90% pure and suitable for various studies which include high throughput screening (HTS) and hit follow-up. However, in order to obtain enzyme of greater homogeneity and purity for crystallographic and high precision spectroscopic and kinetic studies of sGC with select stimulators, the sGC solution after the POROS Q step was further purified by GTP-agarose affinity chromatography. This additional step led to the generation of 26 mg of enzyme that was about 99% pure. This highly pure and active enzyme exhibited a M(r)=144,933 by static light scattering supportive of a dimeric structure. It migrated as a two-band protein, each of equal intensity, on SDS-PAGE corresponding to the alpha (M(r) approximately 77,000) and beta (M(r) approximately 70,000) sGC subunits. It showed an A(430)/A(280)=1.01, indicating one heme per heterodimer, and a maximum of the Soret band at 430 nm indicative of a penta-coordinated ferrous heme with a histidine as the axial ligand. The Soret band shifted to 398 nm in the presence of an NO donor as expected for the formation of a penta-coordinated nitrosyl-heme complex. Non-stimulated sGC had k(cat)/K(m)=1.7 x 10(-3)s(-1)microM(-1) that increased to 5.8 x 10(-1)s(-1)microM(-1) upon stimulation with an NO donor which represents a 340-fold increase due to stimulation. The novel combination of using the TIPS method for co-expression of a heterodimeric heme-containing enzyme, along with the application of a reproducible ligand affinity purification method, has enabled us to obtain recombinant human sGC of both the quality and quantity needed to study structure-function relationships.


Asunto(s)
Baculoviridae/genética , Guanilato Ciclasa/aislamiento & purificación , Guanilato Ciclasa/metabolismo , Insectos/citología , Insectos/virología , Receptores Citoplasmáticos y Nucleares/aislamiento & purificación , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Animales , Baculoviridae/fisiología , Técnicas de Cultivo de Célula , Cromatografía de Afinidad , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Guanilato Ciclasa/química , Humanos , Cinética , Fosfoenolpiruvato Carboxiquinasa (GTP)/química , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Proteínas Recombinantes/química , Sefarosa/química , Guanilil Ciclasa Soluble
7.
Bioorg Med Chem Lett ; 19(3): 908-11, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19097791

RESUMEN

The inhibition of PKC-zeta has been proposed to be a potential drug target for immune and inflammatory diseases. A series of 2-(6-phenyl-1H indazol-3-yl)-1H-benzo[d]imidazoles with initial high crossover to CDK-2 has been optimized to afford potent and selective inhibitors of protein kinase c-zeta (PKC-zeta). The determination of the crystal structures of key inhibitor:CDK-2 complexes informed the design and analysis of the series. The most selective and potent analog was identified by variation of the aryl substituent at the 6-position of the indazole template to give a 4-NH(2) derivative. The analog displays good selectivity over other PKC isoforms (alpha, betaII, gamma, delta, epsilon, mu, theta, eta and iota/lambda) and CDK-2, however it displays marginal selectivity against a panel of other kinases (37 profiled).


Asunto(s)
Bencimidazoles/síntesis química , Química Farmacéutica/métodos , Inhibidores Enzimáticos/farmacología , Imidazoles/síntesis química , Proteína Quinasa C/química , Proteína Quinasa C/aislamiento & purificación , Bencimidazoles/farmacología , Cristalografía por Rayos X , Ciclina A/química , Quinasa 2 Dependiente de la Ciclina/metabolismo , Diseño de Fármacos , Humanos , Imidazoles/farmacología , Concentración 50 Inhibidora , Modelos Químicos , Modelos Moleculares , Conformación Molecular , Isoformas de Proteínas
8.
Protein Expr Purif ; 60(1): 58-63, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18430586

RESUMEN

Soluble guanylate cyclase (sGC), the main target of nitric oxide (NO), is a cytosolic, heme-containing, heterodimeric enzyme that catalyzes the conversion of guanosine 5'-triphosphate (GTP) to 3,5'-cyclic guanosine monophosphate (cGMP) and pyrophosphate (PPi) in the presence of Mg2+. Cyclic GMP is then involved in transmitting the NO activating signals to a variety of downstream effectors such as cyclic-nucleotide-gated channels, protein kinases, and phosphodiesterases. In this work, sGC has been purified from bovine lung. The lungs were subjected to grinding and extraction with buffer at physiological pH followed by centrifugation. The resulting solution was subjected to successive column chromatography on DEAE- and Q-Sepharose, Ceramic Hydroxyapatite, Resource Q, and GTP-agarose. The purified enzyme migrated as a two-band protein on SDS-PAGE corresponding to sGC subunits alpha (M(r)=77,532) and beta (M(r)=70,500) and had an A(280 nm)/A(430 nm) of approximately 1 indicating one heme per heterodimer. The yield of enzyme was 8-10mg from 4 to 5 kg bovine lungs. V(max) and K(m) of non-stimulated sGC were 22 nmol/mg/min and 180 microM, respectively. Upon stimulation with the NO donor 3-ethyl-3-(ethylaminoethyl)-1-hydroxy-2-oxo-1-triazene, the V(max) increased to 1330 nmol/mg/min while the K(m) dropped to 43 microM. The quality and quantity of enzyme make it suitable for studies to probe the structure and catalytic mechanism of this enzyme and for research related to drug discovery.


Asunto(s)
GMP Cíclico/metabolismo , Guanilato Ciclasa/aislamiento & purificación , Pulmón/enzimología , Receptores Citoplasmáticos y Nucleares/aislamiento & purificación , Animales , Bovinos , Cromatografía de Afinidad/métodos , Guanilato Ciclasa/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Guanilil Ciclasa Soluble
9.
J Med Chem ; 60(13): 5521-5542, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28498658

RESUMEN

Through fragment-based drug design focused on engaging the active site of IRAK4 and leveraging three-dimensional topology in a ligand-efficient manner, a micromolar hit identified from a screen of a Pfizer fragment library was optimized to afford IRAK4 inhibitors with nanomolar potency in cellular assays. The medicinal chemistry effort featured the judicious placement of lipophilicity, informed by co-crystal structures with IRAK4 and optimization of ADME properties to deliver clinical candidate PF-06650833 (compound 40). This compound displays a 5-unit increase in lipophilic efficiency from the fragment hit, excellent kinase selectivity, and pharmacokinetic properties suitable for oral administration.


Asunto(s)
Descubrimiento de Drogas , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Isoquinolinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Administración Oral , Relación Dosis-Respuesta a Droga , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Isoquinolinas/administración & dosificación , Isoquinolinas/química , Lactamas , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
10.
Nat Struct Mol Biol ; 18(2): 198-204, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21240271

RESUMEN

Autotaxin (ATX, also known as ectonucleotide pyrophosphatase/phosphodiesterase-2, ENPP2) is a secreted lysophospholipase D that generates the lipid mediator lysophosphatidic acid (LPA), a mitogen and chemoattractant for many cell types. ATX-LPA signaling is involved in various pathologies including tumor progression and inflammation. However, the molecular basis of substrate recognition and catalysis by ATX and the mechanism by which it interacts with target cells are unclear. Here, we present the crystal structure of ATX, alone and in complex with a small-molecule inhibitor. We have identified a hydrophobic lipid-binding pocket and mapped key residues for catalysis and selection between nucleotide and phospholipid substrates. We have shown that ATX interacts with cell-surface integrins through its N-terminal somatomedin B-like domains, using an atypical mechanism. Our results define determinants of substrate discrimination by the ENPP family, suggest how ATX promotes localized LPA signaling and suggest new approaches for targeting ATX with small-molecule therapeutic agents.


Asunto(s)
Integrinas/metabolismo , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/metabolismo , Pirofosfatasas/química , Pirofosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Dominio Catalítico , Línea Celular , Cristalografía por Rayos X , Humanos , Lisofosfolípidos/metabolismo , Datos de Secuencia Molecular , Mutación , Hidrolasas Diéster Fosfóricas/genética , Unión Proteica , Estructura Terciaria de Proteína , Pirofosfatasas/genética , Ratas , Especificidad por Sustrato
11.
J Mol Biol ; 400(3): 413-33, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20478313

RESUMEN

Janus kinases (JAKs) are critical regulators of cytokine pathways and attractive targets of therapeutic value in both inflammatory and myeloproliferative diseases. Although the crystal structures of active JAK1 and JAK2 kinase domains have been reported recently with the clinical compound CP-690550, the structures of both TYK2 and JAK3 with CP-690550 have remained outstanding. Here, we report the crystal structures of TYK2, a first in class structure, and JAK3 in complex with PAN-JAK inhibitors CP-690550 ((3R,4R)-3-[4-methyl-3-[N-methyl-N-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]piperidin-1-yl]-3-oxopropionitrile) and CMP-6 (tetracyclic pyridone 2-t-butyl-9-fluoro-3,6-dihydro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-one), both of which bind in the ATP-binding cavities of both JAK isozymes in orientations similar to that observed in crystal structures of JAK1 and JAK2. Additionally, a complete thermodynamic characterization of JAK/CP-690550 complex formation was completed by isothermal titration calorimetry, indicating the critical role of the nitrile group from the CP-690550 compound. Finally, computational analysis using WaterMap further highlights the critical positioning of the CP-690550 nitrile group in the displacement of an unfavorable water molecule beneath the glycine-rich loop. Taken together, the data emphasize the outstanding properties of the kinome-selective JAK inhibitor CP-690550, as well as the challenges in obtaining JAK isozyme-selective inhibitors due to the overall structural and sequence similarities between the TYK2, JAK1, JAK2 and JAK3 isozymes. Nevertheless, subtle amino acid variations of residues lining the ligand-binding cavity of the JAK enzymes, as well as the global positioning of the glycine-rich loop, might provide the initial clues to obtaining JAK-isozyme selective inhibitors.


Asunto(s)
Bencimidazoles/metabolismo , Inhibidores Enzimáticos/metabolismo , Janus Quinasa 3/química , Piridonas/metabolismo , Pirimidinas/metabolismo , Pirroles/metabolismo , TYK2 Quinasa/química , Sitios de Unión , Calorimetría , Humanos , Janus Quinasa 3/metabolismo , Cinética , Modelos Moleculares , Piperidinas , Unión Proteica , Estructura Terciaria de Proteína , TYK2 Quinasa/metabolismo
12.
ACS Med Chem Lett ; 1(2): 59-63, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24900177

RESUMEN

Hematopoietic prostaglandin D synthase (HPGDS) is primarly expressed in mast cells, antigen-presenting cells, and Th-2 cells. HPGDS converts PGH2 into PGD2, a mediator thought to play a pivotal role in airway allergy and inflammatory processes. In this letter, we report the discovery of an orally potent and selective inhibitor of HPGDS that reduces the antigen-induced response in allergic sheep.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA