Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 67(5): 3935-3958, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38365209

RESUMEN

As SARS-CoV-2 continues to circulate, antiviral treatments are needed to complement vaccines. The virus's main protease, 3CLPro, is an attractive drug target in part because it recognizes a unique cleavage site, which features a glutamine residue at the P1 position and is not utilized by human proteases. Herein, we report the invention of MK-7845, a novel reversible covalent 3CLPro inhibitor. While most covalent inhibitors of SARS-CoV-2 3CLPro reported to date contain an amide as a Gln mimic at P1, MK-7845 bears a difluorobutyl substituent at this position. SAR analysis and X-ray crystallographic studies indicate that this group interacts with His163, the same residue that forms a hydrogen bond with the amide substituents typically found at P1. In addition to promising in vivo efficacy and an acceptable projected human dose with unboosted pharmacokinetics, MK-7845 exhibits favorable properties for both solubility and absorption that may be attributable to the unusual difluorobutyl substituent.


Asunto(s)
COVID-19 , Glutamina , Humanos , Glutamina/química , SARS-CoV-2 , Cisteína Endopeptidasas/química , Invenciones , Inhibidores de Proteasas/farmacología , Amidas , Antivirales/farmacología , Antivirales/química
2.
Nat Commun ; 12(1): 2971, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34016973

RESUMEN

The leukotriene B4 receptor 1 (BLT1) regulates the recruitment and chemotaxis of different cell types and plays a role in the pathophysiology of infectious, allergic, metabolic, and tumorigenic human diseases. Here we present a crystal structure of human BLT1 (hBLT1) in complex with a selective antagonist MK-D-046, developed for the treatment of type 2 diabetes and other inflammatory conditions. Comprehensive analysis of the structure and structure-activity relationship data, reinforced by site-directed mutagenesis and docking studies, reveals molecular determinants of ligand binding and selectivity toward different BLT receptor subtypes and across species. The structure helps to identify a putative membrane-buried ligand access channel as well as potential receptor binding modes of endogenous agonists. These structural insights of hBLT1 enrich our understanding of its ligand recognition and open up future avenues in structure-based drug design.


Asunto(s)
Hipoglucemiantes/química , Receptores de Leucotrieno B4/ultraestructura , Animales , Sitios de Unión/genética , Cristalografía por Rayos X , Diabetes Mellitus Tipo 2 , Células HEK293 , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Ligandos , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Receptores de Leucotrieno B4/agonistas , Receptores de Leucotrieno B4/antagonistas & inhibidores , Receptores de Leucotrieno B4/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Células Sf9 , Spodoptera , Relación Estructura-Actividad
3.
Protein Eng Des Sel ; 332020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-33341882

RESUMEN

Single-domain antibody fragments known as VHH have emerged in the pharmaceutical industry as useful biotherapeutics. These molecules, which are naturally produced by camelids, share the characteristics of high affinity and specificity with traditional human immunoglobulins, while consisting of only a single heavy chain. Currently, the most common method for generating VHH is via animal immunization, which can be costly and time-consuming. Here we describe the development of a synthetic VHH library for in vitro selection of single domain binders. We combine structure-based design and next-generation sequencing analysis to build a library with characteristics that closely mimic the natural repertoire. To validate the performance of our synthetic library, we isolated VHH against three model antigens (soluble mouse PD-1 ectodomain, amyloid-ß peptide, and MrgX1 GPCR) of different sizes and characteristics. We were able to isolate diverse binders targeting different epitopes with high affinity (as high as 5 nM) against all three targets. We then show that anti-mPD-1 binders have functional activity in a receptor blocking assay.


Asunto(s)
Especificidad de Anticuerpos , Antígenos/química , Epítopos/química , Biblioteca de Péptidos , Ingeniería de Proteínas , Anticuerpos de Dominio Único , Animales , Antígenos/inmunología , Camélidos del Nuevo Mundo/genética , Camélidos del Nuevo Mundo/inmunología , Camelus/genética , Camelus/inmunología , Epítopos/inmunología , Ratones , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Relación Estructura-Actividad
4.
Nature ; 533(7602): 274-7, 2016 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-27111510

RESUMEN

Glucagon is a 29-amino-acid peptide released from the α-cells of the islet of Langerhans, which has a key role in glucose homeostasis. Glucagon action is transduced by the class B G-protein-coupled glucagon receptor (GCGR), which is located on liver, kidney, intestinal smooth muscle, brain, adipose tissue, heart and pancreas cells, and this receptor has been considered an important drug target in the treatment of diabetes. Administration of recently identified small-molecule GCGR antagonists in patients with type 2 diabetes results in a substantial reduction of fasting and postprandial glucose concentrations. Although an X-ray structure of the transmembrane domain of the GCGR has previously been solved, the ligand (NNC0640) was not resolved. Here we report the 2.5 Å structure of human GCGR in complex with the antagonist MK-0893 (ref. 4), which is found to bind to an allosteric site outside the seven transmembrane (7TM) helical bundle in a position between TM6 and TM7 extending into the lipid bilayer. Mutagenesis of key residues identified in the X-ray structure confirms their role in the binding of MK-0893 to the receptor. The unexpected position of the binding site for MK-0893, which is structurally similar to other GCGR antagonists, suggests that glucagon activation of the receptor is prevented by restriction of the outward helical movement of TM6 required for G-protein coupling. Structural knowledge of class B receptors is limited, with only one other ligand-binding site defined--for the corticotropin-releasing hormone receptor 1 (CRF1R)--which was located deep within the 7TM bundle. We describe a completely novel allosteric binding site for class B receptors, providing an opportunity for structure-based drug design for this receptor class and furthering our understanding of the mechanisms of activation of these receptors.


Asunto(s)
Pirazoles/metabolismo , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/química , beta-Alanina/análogos & derivados , Sitio Alostérico/efectos de los fármacos , Cristalografía por Rayos X , Glucagón/metabolismo , Glucagón/farmacología , Humanos , Ligandos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Pirazoles/química , Pirazoles/farmacología , Receptores de Hormona Liberadora de Corticotropina/química , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Glucagón/clasificación , Receptores de Glucagón/metabolismo , beta-Alanina/química , beta-Alanina/metabolismo , beta-Alanina/farmacología
5.
Nature ; 481(7382): 469-74, 2012 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-22230955

RESUMEN

Neurotransmitter sodium symporters are integral membrane proteins that remove chemical transmitters from the synapse and terminate neurotransmission mediated by serotonin, dopamine, noradrenaline, glycine and GABA (γ-aminobutyric acid). Crystal structures of the bacterial homologue, LeuT, in substrate-bound outward-occluded and competitive inhibitor-bound outward-facing states have advanced our mechanistic understanding of neurotransmitter sodium symporters but have left fundamental questions unanswered. Here we report crystal structures of LeuT mutants in complexes with conformation-specific antibody fragments in the outward-open and inward-open states. In the absence of substrate but in the presence of sodium the transporter is outward-open, illustrating how the binding of substrate closes the extracellular gate through local conformational changes: hinge-bending movements of the extracellular halves of transmembrane domains 1, 2 and 6, together with translation of extracellular loop 4. The inward-open conformation, by contrast, involves large-scale conformational changes, including a reorientation of transmembrane domains 1, 2, 5, 6 and 7, a marked hinge bending of transmembrane domain 1a and occlusion of the extracellular vestibule by extracellular loop 4. These changes close the extracellular gate, open an intracellular vestibule, and largely disrupt the two sodium sites, thus providing a mechanism by which ions and substrate are released to the cytoplasm. The new structures establish a structural framework for the mechanism of neurotransmitter sodium symporters and their modulation by therapeutic and illicit substances.


Asunto(s)
Apoproteínas/química , Proteínas Bacterianas/química , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Apoproteínas/genética , Apoproteínas/inmunología , Apoproteínas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Fragmentos Fab de Inmunoglobulinas/inmunología , Iones/química , Modelos Moleculares , Movimiento , Conformación Proteica , Sodio/química , Sodio/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
6.
Nature ; 468(7327): 1129-32, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21179170

RESUMEN

Neurotransmitter/sodium symporters (NSSs) couple the uptake of neurotransmitter with one or more sodium ions, removing neurotransmitter from the synaptic cleft. NSSs are essential to the function of chemical synapses, are associated with multiple neurological diseases and disorders, and are the targets of therapeutic and illicit drugs. LeuT, a prokaryotic orthologue of the NSS family, is a model transporter for understanding the relationships between molecular mechanism and atomic structure in a broad range of sodium-dependent and sodium-independent secondary transporters. At present there is a controversy over whether there are one or two high-affinity substrate binding sites in LeuT. The first-reported crystal structure of LeuT, together with subsequent functional and structural studies, provided direct evidence for a single, high-affinity, centrally located substrate-binding site, defined as the S1 site. Recent binding, flux and molecular simulation studies, however, have been interpreted in terms of a model where there are two high-affinity binding sites: the central, S1, site and a second, the S2 site, located within the extracellular vestibule. Furthermore, it was proposed that the S1 and S2 sites are allosterically coupled such that occupancy of the S2 site is required for the cytoplasmic release of substrate from the S1 site. Here we address this controversy by performing direct measurement of substrate binding to wild-type LeuT and to S2 site mutants using isothermal titration calorimetry, equilibrium dialysis and scintillation proximity assays. In addition, we perform uptake experiments to determine whether the proposed allosteric coupling between the putative S2 site and the S1 site manifests itself in the kinetics of substrate flux. We conclude that LeuT harbours a single, centrally located, high-affinity substrate-binding site and that transport is well described by a simple, single-substrate kinetic mechanism.


Asunto(s)
Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/química , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Sitios de Unión , Humanos , Ionóforos/farmacología , Cinética , Leucina/genética , Modelos Moleculares , Mutación , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Transporte de Proteínas/efectos de los fármacos , Valinomicina/farmacología
7.
Nature ; 459(7245): 347-55, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19458710

RESUMEN

Transmembrane sodium-ion gradients provide energy that can be harnessed by 'secondary transporters' to drive the translocation of solute molecules into a cell. Decades of study have shown that such sodium-coupled transporters are involved in many physiological processes, making them targets for the treatment of numerous diseases. Within the past year, crystal structures of several sodium-coupled transporters from different families have been reported, showing a remarkable structural conservation between functionally unrelated transporters. These atomic-resolution structures are revealing the mechanism of the sodium-coupled transport of solutes across cellular membranes.


Asunto(s)
Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Sodio/metabolismo , Animales , Sitios de Unión , Humanos , Modelos Moleculares , Conformación Proteica
8.
Biochemistry ; 48(12): 2723-39, 2009 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19220019

RESUMEN

Backbone conformational dynamics of Thermotoga neapolitana adenylate kinase in the free form (TNAK) and inhibitor-bound form (TNAK*Ap5A) were investigated at 30 degrees C using (15)N NMR relaxation measurements and NMR monitored hydrogen-deuterium exchange. With kinetic parameters identical to those of Escherichia coli AK (ECAK) at 30 degrees C, TNAK is a unique hyperthermophilic enzyme. These catalytic properties make TNAK an interesting and novel model to study the interplay between protein rigidity, stability, and activity. Comparison of fast time scale dynamics (picosecond to nanosecond) in the open and closed states of TNAK and ECAK at 30 degrees C reveals a uniformly higher rigidity across all domains of TNAK. Within this framework of a rigid TNAK structure, several residues located in the AMP-binding domain and in the core-lid hinge regions display high picosecond to nanosecond time scale flexibility. Together with the recent comparison of ECAK dynamics with those of hyperthermophilic Aquifex aeolicus AK (AAAK), our results provide strong evidence for the role of picosecond to nanosecond time scale fluctuations in both stability and activity. In the slow time scales, TNAK's increased rigidity is not uniform but localized in the AMP-binding and lid domains. The core domain amides of ECAK and TNAK in the open and closed states show comparable protection against exchange. Significantly, the hinges framing the lid domain show similar exchange data in ECAK and TNAK open and closed forms. Our NMR relaxation and hydrogen-deuterium exchange studies therefore suggest that TNAK maintains high activity at 30 degrees C by localizing flexibility to the hinge regions that are key to facilitating conformational changes.


Asunto(s)
Adenilato Quinasa/química , Proteínas Bacterianas/química , Temperatura , Thermotoga neapolitana/enzimología , Secuencia de Aminoácidos , Medición de Intercambio de Deuterio , Modelos Biológicos , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Alineación de Secuencia
9.
Proteins ; 58(1): 88-100, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15521058

RESUMEN

The ternary complex of Escherichia coli adenylate kinase (ECAK) with its substrates adenosine monophosphate (AMP) and Mg-ATP, which catalyzes the reversible transfer of a phosphoryl group between adenosine triphosphate (ATP) and AMP, was studied using molecular dynamics. The starting structure for the simulation was assembled from the crystal structures of ECAK complexed with the bisubstrate analog diadenosine pentaphosphate (AP(5)A) and of Bacillus stearothermophilus adenylate kinase complexed with AP(5)A, Mg(2+), and 4 coordinated water molecules, and by deleting 1 phosphate group from AP(5)A. The interactions of ECAK residues with the various moieties of ATP and AMP were compared to those inferred from NMR, X-ray crystallography, site-directed mutagenesis, and enzyme kinetic studies. The simulation supports the hypothesis that hydrogen bonds between AMP's adenine and the protein are at the origin of the high nucleoside monophosphate (NMP) specificity of AK. The ATP adenine and ribose moieties are only loosely bound to the protein, while the ATP phosphates are strongly bound to surrounding residues. The coordination sphere of Mg(2+), consisting of 4 waters and oxygens of the ATP beta- and gamma-phosphates, stays approximately octahedral during the simulation. The important role of the conserved Lys13 in the P loop in stabilizing the active site by bridging the ATP and AMP phosphates is evident. The influence of Mg(2+), of its coordination waters, and of surrounding charged residues in maintaining the geometry and distances of the AMP alpha-phosphate and ATP beta- and gamma-phosphates is sufficient to support an associative reaction mechanism for phosphoryl transfer.


Asunto(s)
Adenilato Quinasa/química , Adenilato Quinasa/metabolismo , Simulación por Computador , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Modelos Moleculares , Termodinámica , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Fosforilación , Estructura Secundaria de Proteína , Especificidad por Sustrato
10.
Biochem J ; 372(Pt 2): 577-85, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12625835

RESUMEN

The adenylate kinase (AK) gene from Thermotoga neapolitana, a hyperthermophilic bacterium, was cloned and overexpressed in Escherichia coli, and the recombinant enzyme was biochemically characterized. The T. neapolitana AK (TNAK) sequence indicates that this enzyme belongs to the long bacterial AKs. TNAK contains the four cysteine residues that bind Zn(2+) in all Gram-positive AKs and in a few other Zn(2+)-containing bacterial AKs. Atomic emission spectroscopy and titration data indicate a content of 1 mol of Zn(2+)/mol of recombinant TNAK. The EDTA-treated enzyme has a melting temperature (T (m)=93.5 degrees C) 6.2 degrees C below that of the holoenzyme (99.7 degrees C), identifying Zn(2+) as a stabilizing feature in TNAK. TNAK is a monomeric enzyme with a molecular mass of approx. 25 kDa. TNAK displays V (max) and K (m) values at 30 degrees C identical with those of the E. coli AK at 30 degrees C, and displays very high activity at 80 degrees C, with a specific activity above 8000 units/mg. The unusually high activity of TNAK at 30 degrees C makes it an interesting model to test the role of enzyme flexibility in activity.


Asunto(s)
Adenilato Quinasa/metabolismo , Bacilos Gramnegativos Anaerobios Rectos, Curvos y Espirales/enzimología , Calor , Adenilato Quinasa/química , Adenilato Quinasa/genética , Secuencia de Aminoácidos , Apoenzimas , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dicroismo Circular , Clonación Molecular , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Estabilidad de Enzimas , Escherichia coli/genética , Escherichia coli/metabolismo , Bacilos Gramnegativos Anaerobios Rectos, Curvos y Espirales/genética , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , Pliegue de Proteína , Homología de Secuencia de Aminoácido , Espectrofotometría , Zinc/química , Zinc/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...