Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioorg Med Chem Lett ; 23(11): 3443-7, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23597790

RESUMEN

Selective phosphodiesterase 2 (PDE2) inhibitors are shown to have efficacy in a rat model of osteoarthritis (OA) pain. We identified potent, selective PDE2 inhibitors by optimizing residual PDE2 activity in a series of phosphodiesterase 4 (PDE4) inhibitors, while minimizing PDE4 inhibitory activity. These newly designed PDE2 inhibitors bind to the PDE2 enzyme in a cGMP-like binding mode orthogonal to the cAMP-like binding mode found in PDE4. Extensive structure activity relationship studies ultimately led to identification of pyrazolodiazepinone, 22, which was >1000-fold selective for PDE2 over recombinant, full length PDEs 1B, 3A, 3B, 4A, 4B, 4C, 7A, 7B, 8A, 8B, 9, 10 and 11. Compound 22 also retained excellent PDE2 selectivity (241-fold to 419-fold) over the remaining recombinant, full length PDEs, 1A, 4D, 5, and 6. Compound 22 exhibited good pharmacokinetic properties and excellent oral bioavailability (F=78%, rat). In an in vivo rat model of OA pain, compound 22 had significant analgesic activity 1 and 3h after a single, 10 mg/kg, subcutaneous dose.


Asunto(s)
Azepinas/química , Azirinas/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Dihidropiridinas/química , Inhibidores de Fosfodiesterasa/química , Pirazoles/química , Analgésicos/química , Analgésicos/farmacocinética , Analgésicos/uso terapéutico , Animales , Azepinas/farmacocinética , Azepinas/uso terapéutico , Azirinas/farmacocinética , Azirinas/uso terapéutico , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Dihidropiridinas/farmacocinética , Dihidropiridinas/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Semivida , Osteoartritis/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa/farmacocinética , Inhibidores de Fosfodiesterasa/uso terapéutico , Unión Proteica , Pirazoles/farmacocinética , Pirazoles/uso terapéutico , Ratas , Relación Estructura-Actividad
2.
Bioorg Med Chem Lett ; 23(11): 3438-42, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23582272

RESUMEN

We identified potent, selective PDE2 inhibitors by optimizing residual PDE2 activity in a series of PDE4 inhibitors, while simultaneously minimizing PDE4 activity. These newly designed PDE2 inhibitors bind to the PDE2 enzyme in a cGMP-like mode in contrast to the cAMP-like binding mode found in PDE4. Structure activity relationship studies coupled with an inhibitor bound crystal structure in the active site of the catalytic domain of PDE2 identified structural features required to minimize PDE4 inhibition while simultaneously maximizing PDE2 inhibition.


Asunto(s)
Azirinas/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/antagonistas & inhibidores , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Dihidropiridinas/química , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa/química , Animales , Azirinas/metabolismo , Azirinas/uso terapéutico , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Dihidropiridinas/metabolismo , Dihidropiridinas/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Osteoartritis/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/metabolismo , Inhibidores de Fosfodiesterasa/uso terapéutico , Unión Proteica , Relación Estructura-Actividad
3.
Proc Natl Acad Sci U S A ; 106(6): 1737-42, 2009 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19164768

RESUMEN

As the need for novel antibiotic classes to combat bacterial drug resistance increases, the paucity of leads resulting from target-based antibacterial screening of pharmaceutical compound libraries is of major concern. One explanation for this lack of success is that antibacterial screening efforts have not leveraged the eukaryotic bias resulting from more extensive chemistry efforts targeting eukaryotic gene families such as G protein-coupled receptors and protein kinases. Consistent with a focus on antibacterial target space resembling these eukaryotic targets, we used whole-cell screening to identify a series of antibacterial pyridopyrimidines derived from a protein kinase inhibitor pharmacophore. In bacteria, the pyridopyrimidines target the ATP-binding site of biotin carboxylase (BC), which catalyzes the first enzymatic step of fatty acid biosynthesis. These inhibitors are effective in vitro and in vivo against fastidious gram-negative pathogens including Haemophilus influenzae. Although the BC active site has architectural similarity to those of eukaryotic protein kinases, inhibitor binding to the BC ATP-binding site is distinct from the protein kinase-binding mode, such that the inhibitors are selective for bacterial BC. In summary, we have discovered a promising class of potent antibacterials with a previously undescribed mechanism of action. In consideration of the eukaryotic bias of pharmaceutical libraries, our findings also suggest that pursuit of a novel inhibitor leads for antibacterial targets with active-site structural similarity to known human targets will likely be more fruitful than the traditional focus on unique bacterial target space, particularly when structure-based and computational methodologies are applied to ensure bacterial selectivity.


Asunto(s)
Antibacterianos/química , Ligasas de Carbono-Nitrógeno/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Antibacterianos/farmacología , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/enzimología , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/enzimología , Moraxella catarrhalis/efectos de los fármacos , Moraxella catarrhalis/enzimología , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Bibliotecas de Moléculas Pequeñas
4.
J Struct Biol ; 162(1): 152-69, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18086534

RESUMEN

In this article, we describe for the first time the high-resolution crystal structure of a phenylalanine tRNA synthetase from the pathogenic bacterium Staphylococcus haemolyticus. We demonstrate the subtle yet important structural differences between this enzyme and the previously described Thermus thermophilus ortholog. We also explain the structure-activity relationship of several recently reported inhibitors. The native enzyme crystals were of poor quality--they only diffracted X-rays to 3-5A resolution. Therefore, we have executed a rational surface mutagenesis strategy that has yielded crystals of this 2300-amino acid multidomain protein, diffracting to 2A or better. This methodology is discussed and contrasted with the more traditional domain truncation approach.


Asunto(s)
Proteínas Bacterianas/química , Fenilalanina-ARNt Ligasa/química , Staphylococcus haemolyticus/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X/métodos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Fenilalanina-ARNt Ligasa/metabolismo , Ingeniería de Proteínas/métodos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Staphylococcus haemolyticus/genética
5.
Protein Sci ; 16(12): 2657-66, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18029420

RESUMEN

N-Acetylglucosamine-1-phosphate uridyltransferase (GlmU) catalyzes the first step in peptidoglycan biosynthesis in both Gram-positive and Gram-negative bacteria. The products of the GlmU reaction are essential for bacterial survival, making this enzyme an attractive target for antibiotic drug discovery. A series of Haemophilus influenzae GlmU (hiGlmU) structures were determined by X-ray crystallography in order to provide structural and functional insights into GlmU activity and inhibition. The information derived from these structures was combined with biochemical characterization of the K25A, Q76A, D105A, Y103A, V223A, and E224A hiGlmU mutants in order to map these active-site residues to catalytic activity of the enzyme and refine the mechanistic model of the GlmU uridyltransferase reaction. These studies suggest that GlmU activity follows a sequential substrate-binding order that begins with UTP binding noncovalently to the GlmU enzyme. The uridyltransferase active site then remains in an open apo-like conformation until N-acetylglucosamine-1-phosphate (GlcNAc-1-P) binds and induces a conformational change at the GlcNAc-binding subsite. Following the binding of GlcNAc-1-P to the UTP-charged uridyltransferase active site, the non-esterified oxygen of GlcNAc-1-P performs a nucleophilic attack on the alpha-phosphate group of UTP. The new data strongly suggest that the mechanism of phosphotransfer in the uridyltransferase reaction in GlmU is primarily through an associative mechanism with a pentavalent phosphate intermediate and an inversion of stereochemistry. Finally, the structural and biochemical characterization of the uridyltransferase active site and catalytic mechanism described herein provides a basis for the structure-guided design of novel antibacterial agents targeting GlmU activity.


Asunto(s)
Haemophilus influenzae/enzimología , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Acetilglucosamina/análogos & derivados , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Ligandos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína , Uridina/química , Uridina/metabolismo , Uridina Trifosfato/metabolismo
6.
J Biol Chem ; 282(38): 27781-91, 2007 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-17623656

RESUMEN

Matrix metalloproteinase-13 (MMP13) is a Zn(2+)-dependent protease that catalyzes the cleavage of type II collagen, the main structural protein in articular cartilage. Excess MMP13 activity causes cartilage degradation in osteoarthritis, making this protease an attractive therapeutic target. However, clinically tested MMP inhibitors have been associated with a painful, joint-stiffening musculoskeletal side effect that may be due to their lack of selectivity. In our efforts to develop a disease-modifying osteoarthritis drug, we have discovered MMP13 inhibitors that differ greatly from previous MMP inhibitors; they do not bind to the catalytic zinc ion, they are noncompetitive with respect to substrate binding, and they show extreme selectivity for inhibiting MMP13. By structure-based drug design, we generated an orally active MMP13 inhibitor that effectively reduces cartilage damage in vivo and does not induce joint fibroplasias in a rat model of musculoskeletal syndrome side effects. Thus, highly selective inhibition of MMP13 in patients may overcome the major safety and efficacy challenges that have limited previously tested non-selective MMP inhibitors. MMP13 inhibitors such as the ones described here will help further define the role of this protease in arthritis and other diseases and may soon lead to drugs that safely halt cartilage damage in patients.


Asunto(s)
Cartílago/metabolismo , Artropatías/metabolismo , Metaloproteinasa 13 de la Matriz/fisiología , Animales , Colágeno/química , Colágeno/metabolismo , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Humanos , Iones , Metaloproteinasa 13 de la Matriz/química , Metaloproteinasa 13 de la Matriz/metabolismo , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Conejos , Ratas , Zinc/química
7.
Nat Struct Mol Biol ; 11(12): 1192-7, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15543157

RESUMEN

MEK1 and MEK2 are closely related, dual-specificity tyrosine/threonine protein kinases found in the Ras/Raf/MEK/ERK mitogen-activated protein kinase (MAPK) signaling pathway. Approximately 30% of all human cancers have a constitutively activated MAPK pathway, and constitutive activation of MEK1 results in cellular transformation. Here we present the X-ray structures of human MEK1 and MEK2, each determined as a ternary complex with MgATP and an inhibitor to a resolution of 2.4 A and 3.2 A, respectively. The structures reveal that MEK1 and MEK2 each have a unique inhibitor-binding pocket adjacent to the MgATP-binding site. The presence of the potent inhibitor induces several conformational changes in the unphosphorylated MEK1 and MEK2 enzymes that lock them into a closed but catalytically inactive species. Thus, the structures reported here reveal a novel, noncompetitive mechanism for protein kinase inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , MAP Quinasa Quinasa 1/química , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/química , MAP Quinasa Quinasa 2/metabolismo , Sitios de Unión , Secuencia Conservada , Dimerización , Inhibidores Enzimáticos/química , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Modelos Moleculares , Estructura Molecular , Estructura Cuaternaria de Proteína , Homología Estructural de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...