Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Chem Biol ; 13(3): 265-267, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28068312

RESUMEN

The rapid increase of antibiotic resistance has created an urgent need to develop novel antimicrobial agents. Here we describe the crystal structure of the promising bacterial target phospho-N-acetylmuramoyl-pentapeptide translocase (MraY) in complex with the nucleoside antibiotic tunicamycin. The structure not only reveals the mode of action of several related natural-product antibiotics but also gives an indication on the binding mode of the MraY UDP-MurNAc-pentapeptide and undecaprenyl-phosphate substrates.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/efectos de los fármacos , Clostridium/efectos de los fármacos , Transferasas/efectos de los fármacos , Tunicamicina/farmacología , Antibacterianos/química , Proteínas Bacterianas/química , Clostridium/enzimología , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Relación Estructura-Actividad , Transferasas/química , Transferasas (Grupos de Otros Fosfatos Sustitutos) , Tunicamicina/química
2.
Proc Natl Acad Sci U S A ; 110(10): 3806-11, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23431194

RESUMEN

Prostaglandin E2 (PGE2) is a key mediator in inflammatory response. The main source of inducible PGE2, microsomal PGE2 synthase-1 (mPGES-1), has emerged as an interesting drug target for treatment of pain. To support inhibitor design, we have determined the crystal structure of human mPGES-1 to 1.2 Å resolution. The structure reveals three well-defined active site cavities within the membrane-spanning region in each monomer interface of the trimeric structure. An important determinant of the active site cavity is a small cytosolic domain inserted between transmembrane helices I and II. This extra domain is not observed in other structures of proteins within the MAPEG (Membrane-Associated Proteins involved in Eicosanoid and Glutathione metabolism) superfamily but is likely to be present also in microsomal GST-1 based on sequence similarity. An unexpected feature of the structure is a 16-Å-deep cone-shaped cavity extending from the cytosolic side into the membrane-spanning region. We suggest a potential role for this cavity in substrate access. Based on the structure of the active site, we propose a catalytic mechanism in which serine 127 plays a key role. We have also determined the structure of mPGES-1 in complex with a glutathione-based analog, providing insight into mPGES-1 flexibility and potential for structure-based drug design.


Asunto(s)
Oxidorreductasas Intramoleculares/química , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Glutatión/análogos & derivados , Glutatión/química , Humanos , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/genética , Microsomas/enzimología , Modelos Moleculares , Datos de Secuencia Molecular , Prostaglandina-E Sintasas , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido
3.
J Med Chem ; 66(20): 14188-14207, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37797307

RESUMEN

Histone deacetylase 6 (HDAC6) is a unique member of the HDAC family mainly targeting cytosolic nonhistone substrates, such as α-tubulin, cortactin, and heat shock protein 90 to regulate cell proliferation, metastasis, invasion, and mitosis in tumors. We describe the identification and characterization of a series of 2-(difluoromethyl)-1,3,4-oxadiazoles (DFMOs) as selective nonhydroxamic acid HDAC6 inhibitors. By comparing structure-activity relationships and performing quantum mechanical calculations of the HDAC6 catalytic mechanism, we show that potent oxadiazoles are electrophilic substrates of HDAC6 and propose a mechanism for the bioactivation. We also observe that the inherent electrophilicity of the oxadiazoles makes them prone to degradation in water solution and the generation of potentially toxic products cannot be ruled out, limiting the developability for chronic diseases. However, the oxadiazoles demonstrate high oral bioavailability and low in vivo clearance and are excellent tools for studying the role of HDAC6 in vitro and in vivo in rats and mice.


Asunto(s)
Neoplasias , Oxadiazoles , Ratas , Ratones , Animales , Histona Desacetilasa 6 , Oxadiazoles/farmacología , Tubulina (Proteína)/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/química
4.
J Synchrotron Radiat ; 19(Pt 2): 288-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22338692

RESUMEN

Currently there is no rack system for the long-term storage of SPINE pucks in spite of their commercial availability and heavy usage at the ESRF. The only way to store pucks is in transport dewar canisters which presents a number of limitations and drawbacks. Here a simple affordable rack for storing SPINE pucks is described, which we believe is accessible to not only synchrotrons but also both academic and industrial research laboratories.


Asunto(s)
Diseño de Equipo , Sincrotrones/instrumentación , Automatización de Laboratorios/instrumentación , Frío , Cristalografía por Rayos X/instrumentación , Nitrógeno
5.
Bioorg Med Chem Lett ; 22(5): 1854-9, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22325942

RESUMEN

The evaluation of a series of bicyclic aminoimidazoles as potent BACE-1 inhibitors is described. The crystal structures of compounds 14 and 23 in complex with BACE-1 reveal hydrogen bond interactions with the protein important for achieving potent inhibition. The optimization of permeability and efflux properties of the compounds is discussed as well as the importance of these properties for attaining in vivo brain efficacy. Compound (R)-25 was selected for evaluation in vivo in wild type mice and 1.5h after oral co-administration of 300µmol/kg (R)-25 and efflux inhibitor GF120918 the brain Aß40 level was reduced by 17% and the plasma Aß40 level by 76%.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Encéfalo/efectos de los fármacos , Imidazoles/química , Imidazoles/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Aminas/química , Aminas/farmacocinética , Aminas/farmacología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/enzimología , Encéfalo/metabolismo , Línea Celular , Cristalografía por Rayos X , Imidazoles/farmacocinética , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Fragmentos de Péptidos/metabolismo
6.
ACS Med Chem Lett ; 13(4): 681-686, 2022 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-35450368

RESUMEN

Thiazolidinedione PPARγ agonists such as rosiglitazone and pioglitazone are effective antidiabetic drugs, but side effects have limited their use. It has been posited that their positive antidiabetic effects are mainly mediated by the inhibition of the CDK5-mediated Ser273 phosphorylation of PPARγ, whereas the side effects are linked to classical PPARγ agonism. Thus compounds that inhibit PPARγ Ser273 phosphorylation but lack classical PPARγ agonism have been sought as safer antidiabetic therapies. Herein we report the discovery by virtual screening of 10, which is a potent PPARγ binder and in vitro inhibitor of the CDK5-mediated phosphorylation of PPARγ Ser273 and displays negligible PPARγ agonism in a reporter gene assay. The pharmacokinetic properties of 10 are compatible with oral dosing, enabling preclinical in vivo testing, and a 7 day treatment demonstrated an improvement in insulin sensitivity in the ob/ob diabetic mouse model.

7.
ACS Chem Biol ; 16(1): 116-124, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33411499

RESUMEN

Branched-chain fatty acids (BCFA) are encountered in Gram-positive bacteria, but less so in other organisms. The bacterial BCFA in membranes are typically saturated, with both odd- and even-numbered carbon chain lengths, and with methyl branches at either the ω-1 (iso) or ω-2 (anteiso) positions. The acylation with BCFA also contributes to the structural diversity of microbial natural products and potentially modulates biological activity. For the tunicamycin (TUN) family of natural products, the toxicity toward eukaryotes is highly dependent upon N-acylation with trans-2,3-unsaturated BCFA. The loss of the 2,3-unsaturation gives modified TUN with reduced eukaryotic toxicity but crucially with retention of the synergistic enhancement of the ß-lactam group of antibiotics. Here, we infer from genomics, mass spectrometry, and deuterium labeling that the trans-2,3-unsaturated TUN variants and the saturated cellular lipids found in TUN-producing Streptomyces are derived from the same pool of BCFA metabolites. Moreover, non-natural primers of BCFA metabolism are selectively incorporated into the cellular lipids of TUN-producing Streptomyces and concomitantly produce structurally novel neo-branched TUN N-acyl variants.


Asunto(s)
Productos Biológicos/metabolismo , Metabolismo de los Lípidos , Streptomyces/metabolismo , Productos Biológicos/química , Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Relación Estructura-Actividad
8.
Sci Rep ; 10(1): 16167, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32999380

RESUMEN

Overproduction and purification of membrane proteins are generally challenging and time-consuming procedures due to low expression levels, misfolding, and low stability once extracted from the membrane. Reducing processing steps and shortening the timespan for purification represent attractive approaches to overcome some of these challenges. We have therefore compared a fast "teabag" purification method with conventional purification for five different membrane proteins (MraY, AQP10, ClC-1, PAR2 and KCC2). Notably, this new approach reduces the purification time significantly, and the quality of the purified membrane proteins is equal to or exceeds conventional methods as assessed by size exclusion chromatography, SDS-PAGE and downstream applications such as ITC, crystallization and cryo-EM. Furthermore, the method is scalable, applicable to a range of affinity resins and allows for parallelization. Consequently, the technique has the potential to substantially simplify purification efforts of membrane proteins in basic and applied sciences.


Asunto(s)
Proteínas de la Membrana/metabolismo , Electroforesis en Gel de Poliacrilamida
9.
ACS Chem Biol ; 15(11): 2885-2895, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33164499

RESUMEN

The alarming growth of antibiotic resistance that is currently ongoing is a serious threat to human health. One of the most promising novel antibiotic targets is MraY (phospho-MurNAc-pentapeptide-transferase), an essential enzyme in bacterial cell wall synthesis. Through recent advances in biochemical research, there is now structural information available for MraY, and for its human homologue GPT (GlcNAc-1-P-transferase), that opens up exciting possibilities for structure-based drug design. The antibiotic compound tunicamycin is a natural product inhibitor of MraY that is also toxic to eukaryotes through its binding to GPT. In this work, we have used tunicamycin and modified versions of tunicamycin as tool compounds to explore the active site of MraY and to gain further insight into what determines inhibitor potency. We have investigated tunicamycin variants where the following motifs have been modified: the length and branching of the tunicamycin fatty acyl chain, the saturation of the fatty acyl chain, the 6″-hydroxyl group of the GlcNAc ring, and the ring structure of the uracil motif. The compounds are analyzed in terms of how potently they bind to MraY, inhibit the activity of the enzyme, and affect the protein thermal stability. Finally, we rationalize these results in the context of the protein structures of MraY and GPT.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Dominio Catalítico/efectos de los fármacos , Transferasas/antagonistas & inhibidores , Transferasas/química , Tunicamicina/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Proteínas Bacterianas/metabolismo , Clostridium/enzimología , Infecciones por Clostridium/tratamiento farmacológico , Guanosina Trifosfato/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Transferasas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)
10.
J Med Chem ; 62(17): 7769-7787, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31415176

RESUMEN

While bronchodilators and inhaled corticosteroids are the mainstay of asthma treatment, up to 50% of asthmatics remain uncontrolled. Many studies show that the cysteinyl leukotriene cascade remains highly activated in some asthmatics, even those on high-dose inhaled or oral corticosteroids. Hence, inhibition of the leukotriene C4 synthase (LTC4S) enzyme could provide a new and differentiated core treatment for patients with a highly activated cysteinyl leukotriene cascade. Starting from a screening hit (3), a program to discover oral inhibitors of LTC4S led to (1S,2S)-2-({5-[(5-chloro-2,4-difluorophenyl)(2-fluoro-2-methylpropyl)amino]-3-methoxypyrazin-2-yl}carbonyl)cyclopropanecarboxylic acid (AZD9898) (36), a picomolar LTC4S inhibitor (IC50 = 0.28 nM) with high lipophilic ligand efficiency (LLE = 8.5), which displays nanomolar potency in cells (peripheral blood mononuclear cell, IC50,free = 6.2 nM) and good in vivo pharmacodynamics in a calcium ionophore-stimulated rat model after oral dosing (in vivo, IC50,free = 34 nM). Compound 36 mitigates the GABA binding, hepatic toxicity signal, and in vivo toxicology findings of an early lead compound 7 with a human dose predicted to be 30 mg once daily.


Asunto(s)
Antiasmáticos/farmacología , Asma/tratamiento farmacológico , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Glutatión Transferasa/antagonistas & inhibidores , Pirazinas/farmacología , Administración Oral , Animales , Antiasmáticos/administración & dosificación , Antiasmáticos/química , Asma/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Glutatión Transferasa/metabolismo , Humanos , Estructura Molecular , Pirazinas/síntesis química , Pirazinas/química , Ratas , Relación Estructura-Actividad
11.
Drug Discov Today ; 23(7): 1426-1435, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29778697

RESUMEN

The rapid growth of antibiotic-resistant bacterial infections is of major concern for human health. Therefore, it is of great importance to characterize novel targets for the development of antibacterial drugs. One promising protein target is MraY (UDP-N-acetylmuramyl-pentapeptide: undecaprenyl phosphate N-acetylmuramyl-pentapeptide-1-phosphate transferase or MurNAc-1-P-transferase), which is essential for bacterial cell wall synthesis. Here, we summarize recent breakthroughs in structural studies of bacterial MraYs and the closely related human GPT (UDP-N-acetylglucosamine: dolichyl phosphate N-acetylglucosamine-1-phosphate transferase or GlcNAc-1-P-transferase). We present a detailed comparison of interaction modes with the natural product inhibitors tunicamycin and muraymycin D2. Finally, we speculate on possible routes to design an antibacterial agent in the form of a potent and selective inhibitor against MraY.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Proteínas Bacterianas/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Peptidoglicano/biosíntesis , Transferasas/antagonistas & inhibidores , Animales , Antibacterianos/síntesis química , Bacterias/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/síntesis química , Humanos , Modelos Moleculares , Nucleósidos/química , Nucleósidos/farmacología , Péptidos/química , Péptidos/farmacología , Conformación Proteica , Relación Estructura-Actividad , Transferasas/química , Transferasas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/antagonistas & inhibidores , Transferasas (Grupos de Otros Fosfatos Sustitutos)/química , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Tunicamicina/química , Tunicamicina/farmacología
12.
J Mol Biol ; 321(2): 329-39, 2002 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-12144789

RESUMEN

The structure of cytochrome c oxidase from Rhodobacter sphaeroides has been solved at 2.3/2.8A (anisotropic resolution). This high-resolution structure revealed atomic details of a bacterial terminal oxidase including water molecule positions and a potential oxygen pathway, which has not been reported in other oxidase structures. A comparative study of the wild-type and the EQ(I-286) mutant enzyme revealed structural rearrangements around E(I-286) that could be crucial for proton transfer in this enzyme. In the structure of the mutant enzyme, EQ(I-286), which cannot transfer protons during oxygen reduction, the side-chain of Q(I-286) does not have the hydrogen bond to the carbonyl oxygen of M(I-107) that is seen in the wild-type structure. Furthermore, the Q(I-286) mutant has a different arrangement of water molecules and residues in the vicinity of the Q side-chain. These differences between the structures could reflect conformational changes that take place upon deprotonation of E(I-286) during turnover of the wild-type enzyme, which could be part of the proton-pumping machinery of the enzyme.


Asunto(s)
Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Mutación/genética , Rhodobacter sphaeroides/enzimología , Sitios de Unión , Catálisis , Cobre/metabolismo , Cristalografía por Rayos X , Complejo IV de Transporte de Electrones/metabolismo , Enlace de Hidrógeno , Ligandos , Modelos Moleculares , Oxidación-Reducción , Oxígeno/metabolismo , Conformación Proteica , Subunidades de Proteína , Protones , Rhodobacter sphaeroides/genética , Agua/metabolismo , Xenón/metabolismo
13.
MAbs ; 7(1): 152-66, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25484051

RESUMEN

Therapeutic monoclonal antibodies targeting G-protein-coupled receptors (GPCRs) are desirable for intervention in a wide range of disease processes. The discovery of such antibodies is challenging due to a lack of stability of many GPCRs as purified proteins. We describe here the generation of Fpro0165, a human anti-formyl peptide receptor 1 (FPR1) antibody generated by variable domain engineering of an antibody derived by immunization of transgenic mice expressing human variable region genes. Antibody isolation and subsequent engineering of affinity, potency and species cross-reactivity using phage display were achieved using FPR1 expressed on HEK cells for immunization and selection, along with calcium release cellular assays for antibody screening. Fpro0165 shows full neutralization of formyl peptide-mediated activation of primary human neutrophils. A crystal structure of the Fpro0165 Fab shows a long, protruding VH CDR3 of 24 amino acids and in silico docking with a homology model of FPR1 suggests that this long VH CDR3 is critical to the predicted binding mode of the antibody. Antibody mutation studies identify the apex of the long VH CDR3 as key to mediating the species cross-reactivity profile of the antibody. This study illustrates an approach for antibody discovery and affinity engineering to typically intractable membrane proteins such as GPCRs.


Asunto(s)
Anticuerpos Monoclonales/química , Regiones Determinantes de Complementariedad/química , Cadenas Pesadas de Inmunoglobulina/química , Simulación del Acoplamiento Molecular , Receptores de Formil Péptido/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Células CHO , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/inmunología , Cricetinae , Cricetulus , Cristalografía por Rayos X , Células HEK293 , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Ratones , Neutrófilos/inmunología , Estructura Cuaternaria de Proteína , Receptores de Formil Péptido/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA