Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(51): 13453-13458, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29208709

RESUMEN

Phosphorylation is a major regulator of protein interactions; however, the mechanisms by which regulation occurs are not well understood. Here we identify a salt-bridge competition or "theft" mechanism that enables a phospho-triggered swap of protein partners by Raf Kinase Inhibitory Protein (RKIP). RKIP transitions from inhibiting Raf-1 to inhibiting G-protein-coupled receptor kinase 2 upon phosphorylation, thereby bridging MAP kinase and G-Protein-Coupled Receptor signaling. NMR and crystallography indicate that a phosphoserine, but not a phosphomimetic, competes for a lysine from a preexisting salt bridge, initiating a partial unfolding event and promoting new protein interactions. Structural elements underlying the theft occurred early in evolution and are found in 10% of homo-oligomers and 30% of hetero-oligomers including Bax, Troponin C, and Early Endosome Antigen 1. In contrast to a direct recognition of phosphorylated residues by binding partners, the salt-bridge theft mechanism represents a facile strategy for promoting or disrupting protein interactions using solvent-accessible residues, and it can provide additional specificity at protein interfaces through local unfolding or conformational change.


Asunto(s)
Secuencia Conservada , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Sustitución de Aminoácidos , Animales , Evolución Molecular , Humanos , Lisina/genética , Lisina/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/química , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Fosforilación , Unión Proteica , Serina/genética , Serina/metabolismo , Troponina C/química , Troponina C/genética , Troponina C/metabolismo , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteína X Asociada a bcl-2/química , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
2.
Biochemistry ; 56(13): 1865-1878, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28225601

RESUMEN

There is growing awareness of the link between drug-target residence time and in vivo drug activity, and there are increasing efforts to determine the molecular factors that control the lifetime of a drug-target complex. Rational alterations in the drug-target residence time require knowledge of both the ground and transition states on the inhibition reaction coordinate, and we have determined the structure-kinetic relationship for 22 ethyl- or hexyl-substituted diphenyl ethers that are slow-binding inhibitors of bpFabI1, the enoyl-ACP reductase FabI1 from Burkholderia pseudomallei. Analysis of enzyme inhibition using a two-dimensional kinetic map demonstrates that the ethyl and hexyl diphenyl ethers fall into two distinct clusters. Modifications to the ethyl diphenyl ether B ring result in changes to both on and off rates, where residence times of up to ∼700 min (∼11 h) are achieved by either ground state stabilization (PT444) or transition state destabilization (slower on rate) (PT404). By contrast, modifications to the hexyl diphenyl ether B ring result in residence times of 300 min (∼5 h) through changes in only ground state stabilization (PT119). Structural analysis of nine enzyme:inhibitor complexes reveals that the variation in structure-kinetic relationships can be rationalized by structural rearrangements of bpFabI1 and subtle changes to the orientation of the inhibitor in the binding pocket. Finally, we demonstrate that three compounds with residence times on bpFabI1 from 118 min (∼2 h) to 670 min (∼11 h) have in vivo efficacy in an acute B. pseudomallei murine infection model using the virulent B. pseudomallei strain Bp400.


Asunto(s)
Antibacterianos/química , Proteínas Bacterianas/antagonistas & inhibidores , Burkholderia pseudomallei/efectos de los fármacos , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Melioidosis/dietoterapia , Éteres Fenílicos/química , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Burkholderia pseudomallei/enzimología , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/crecimiento & desarrollo , Recuento de Colonia Microbiana , Cristalografía por Rayos X , Enoil-ACP Reductasa (NADH)/genética , Enoil-ACP Reductasa (NADH)/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica , Cinética , Pulmón/efectos de los fármacos , Pulmón/microbiología , Melioidosis/tratamiento farmacológico , Melioidosis/microbiología , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Éteres Fenílicos/farmacología , Unión Proteica , Estructura Secundaria de Proteína , Bazo/efectos de los fármacos , Bazo/microbiología , Relación Estructura-Actividad
3.
Biochemistry ; 55(21): 2992-3006, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27136302

RESUMEN

The enoyl-ACP reductase (ENR) catalyzes the last reaction in the elongation cycle of the bacterial type II fatty acid biosynthesis (FAS-II) pathway. While the FabI ENR is a well-validated drug target in organisms such as Mycobacterium tuberculosis and Staphylococcus aureus, alternate ENR isoforms have been discovered in other pathogens, including the FabV enzyme that is the sole ENR in Yersinia pestis (ypFabV). Previously, we showed that the prototypical ENR inhibitor triclosan was a poor inhibitor of ypFabV and that inhibitors based on the 2-pyridone scaffold were more potent [Hirschbeck, M. (2012) Structure 20 (1), 89-100]. These studies were performed with the T276S FabV variant. In the work presented here, we describe a detailed examination of the mechanism and inhibition of wild-type ypFabV and the T276S variant. The T276S mutation significantly reduces the affinity of diphenyl ether inhibitors for ypFabV (20-fold → 100-fold). In addition, while T276S ypFabV generally displays an affinity for 2-pyridone inhibitors higher than that of the wild-type enzyme, the 4-pyridone scaffold yields compounds with similar affinity for both wild-type and T276S ypFabV. T276 is located at the N-terminus of the helical substrate-binding loop, and structural studies coupled with site-directed mutagenesis reveal that alterations in this residue modulate the size of the active site portal. Subsequently, we were able to probe the mechanism of time-dependent inhibition in this enzyme family by extending the inhibition studies to include P142W ypFabV, a mutation that results in a gain of slow-onset inhibition for the 4-pyridone PT156.


Asunto(s)
Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Éteres Fenílicos/química , Piridonas/química , Yersinia pestis/enzimología , Catálisis , Dominio Catalítico , Cristalización , Cristalografía por Rayos X , Enoil-ACP Reductasa (NADH)/genética , Enoil-ACP Reductasa (NADH)/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , NAD/metabolismo , Unión Proteica , Conformación Proteica
4.
J Biol Chem ; 289(23): 15987-6005, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24739388

RESUMEN

Determining the molecular basis for target selectivity is of particular importance in drug discovery. The ideal antibiotic should be active against a broad spectrum of pathogenic organisms with a minimal effect on human targets. CG400549, a Staphylococcus-specific 2-pyridone compound that inhibits the enoyl-acyl carrier protein reductase (FabI), has recently been shown to possess human efficacy for the treatment of methicillin-resistant Staphylococcus aureus infections, which constitute a serious threat to human health. In this study, we solved the structures of three different FabI homologues in complex with several pyridone inhibitors, including CG400549. Based on these structures, we rationalize the 65-fold reduced affinity of CG400549 toward Escherichia coli versus S. aureus FabI and implement concepts to improve the spectrum of antibacterial activity. The identification of different conformational states along the reaction coordinate of the enzymatic hydride transfer provides an elegant visual depiction of the relationship between catalysis and inhibition, which facilitates rational inhibitor design. Ultimately, we developed the novel 4-pyridone-based FabI inhibitor PT166 that retained favorable pharmacokinetics and efficacy in a mouse model of S. aureus infection with extended activity against Gram-negative and mycobacterial organisms.


Asunto(s)
Antibacterianos/farmacología , Diseño de Fármacos , Enoil-ACP Reductasa (NADH)/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Piridonas/farmacología , Animales , Antibacterianos/química , Antibacterianos/farmacocinética , Secuencia de Bases , Cristalografía por Rayos X , Cartilla de ADN , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Femenino , Ratones , Ratones Endogámicos ICR , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Reacción en Cadena de la Polimerasa , Piridonas/química , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo
5.
Structure ; 20(1): 89-100, 2012 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-22244758

RESUMEN

The recently discovered FabV enoyl-ACP reductase, which catalyzes the last step of the bacterial fatty acid biosynthesis (FAS-II) pathway, is a promising but unexploited drug target against the reemerging pathogen Yersinia pestis. The structure of Y. pestis FabV in complex with its cofactor reveals that the enzyme features the common architecture of the short-chain dehydrogenase reductase superfamily, but contains additional structural elements that are mostly folded around the usually flexible substrate-binding loop, thereby stabilizing it in a very tight conformation that seals the active site. The structures of FabV in complex with NADH and two newly developed 2-pyridone inhibitors provide insights for the development of new lead compounds, and suggest a mechanism by which the substrate-binding loop opens to admit the inhibitor, a motion that could also be coupled to the interaction of FabV with the acyl-carrier protein substrate.


Asunto(s)
Enoil-ACP Reductasa (NADH)/química , Enoil-ACP Reductasa (NADH)/metabolismo , Modelos Moleculares , NAD/metabolismo , Conformación Proteica , Piridonas/antagonistas & inhibidores , Yersinia pestis/enzimología , Secuencia de Aminoácidos , Dominio Catalítico/genética , Enoil-ACP Reductasa (NADH)/genética , Datos de Secuencia Molecular , Estructura Molecular , Unión Proteica , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA