Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Mol Cell ; 81(20): 4165-4175.e6, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34433090

RESUMEN

GPCR functional selectivity opens new opportunities for the design of safer drugs. Ligands orchestrate GPCR signaling cascades by modulating the receptor conformational landscape. Our study provides insights into the dynamic mechanism enabling opioid ligands to preferentially activate the G protein over the ß-arrestin pathways through the µ-opioid receptor (µOR). We combine functional assays in living cells, solution NMR spectroscopy, and enhanced-sampling molecular dynamic simulations to identify the specific µOR conformations induced by G protein-biased agonists. In particular, we describe the dynamic and allosteric communications between the ligand-binding pocket and the receptor intracellular domains, through conserved motifs in class A GPCRs. Most strikingly, the biased agonists trigger µOR conformational changes in the intracellular loop 1 and helix 8 domains, which may impair ß-arrestin binding or signaling. The findings may apply to other GPCR families and provide key molecular information that could facilitate the design of biased ligands.


Asunto(s)
Analgésicos Opioides/farmacología , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Transducción de Señal/efectos de los fármacos , Analgésicos Opioides/química , Animales , Sitios de Unión , Diseño Asistido por Computadora , Diseño de Fármacos , Agonismo Parcial de Drogas , Células HEK293 , Humanos , Ligandos , Ratones , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Células Sf9 , Relación Estructura-Actividad , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34663701

RESUMEN

Atypical chemokine receptor 1 (ACKR1) is a G protein-coupled receptor (GPCR) targeted by Staphylococcus aureus bicomponent pore-forming leukotoxins to promote bacterial growth and immune evasion. Here, we have developed an integrative molecular pharmacology and structural biology approach in order to characterize the effect of leukotoxins HlgA and HlgB on ACKR1 structure and function. Interestingly, using cell-based assays and native mass spectrometry, we found that both components HlgA and HlgB compete with endogenous chemokines through a direct binding with the extracellular domain of ACKR1. Unexpectedly, hydrogen/deuterium exchange mass spectrometry analysis revealed that toxin binding allosterically modulates the intracellular G protein-binding domain of the receptor, resulting in dissociation and/or changes in the architecture of ACKR1-Gαi1 protein complexes observed in living cells. Altogether, our study brings important molecular insights into the initial steps of leukotoxins targeting a host GPCR.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Staphylococcus aureus/fisiología , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dimerización , Sistema del Grupo Sanguíneo Duffy/aislamiento & purificación , Sistema del Grupo Sanguíneo Duffy/metabolismo , Exotoxinas/metabolismo , Humanos , Espectrometría de Masas/métodos , Unión Proteica , Receptores de Superficie Celular/aislamiento & purificación , Receptores de Superficie Celular/metabolismo , Células Sf9
3.
Microb Cell Fact ; 16(1): 6, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-28095880

RESUMEN

BACKGROUND: Animal venoms are complex molecular cocktails containing a wide range of biologically active disulphide-reticulated peptides that target, with high selectivity and efficacy, a variety of membrane receptors. Disulphide-reticulated peptides have evolved to display improved specificity, low immunogenicity and to show much higher resistance to degradation than linear peptides. These properties make venom peptides attractive candidates for drug development. However, recombinant expression of reticulated peptides containing disulphide bonds is challenging, especially when associated with the production of large libraries of bioactive molecules for drug screening. To date, as an alternative to artificial synthetic chemical libraries, no comprehensive recombinant libraries of natural venom peptides are accessible for high-throughput screening to identify novel therapeutics. RESULTS: In the accompanying paper an efficient system for the expression and purification of oxidized disulphide-reticulated venom peptides in Escherichia coli is described. Here we report the development of a high-throughput automated platform, that could be adapted to the production of other families, to generate the largest ever library of recombinant venom peptides. The peptides were produced in the periplasm of E. coli using redox-active DsbC as a fusion tag, thus allowing the efficient formation of correctly folded disulphide bridges. TEV protease was used to remove fusion tags and recover the animal venom peptides in the native state. Globally, within nine months, out of a total of 4992 synthetic genes encoding a representative diversity of venom peptides, a library containing 2736 recombinant disulphide-reticulated peptides was generated. The data revealed that the animal venom peptides produced in the bacterial host were natively folded and, thus, are putatively biologically active. CONCLUSIONS: Overall this study reveals that high-throughput expression of animal venom peptides in E. coli can generate large libraries of recombinant disulphide-reticulated peptides of remarkable interest for drug discovery programs.


Asunto(s)
Escherichia coli/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Biblioteca de Péptidos , Péptidos/genética , Proteínas Recombinantes/aislamiento & purificación , Ponzoñas/genética , Animales , Disulfuros/química , Descubrimiento de Drogas/métodos , Endopeptidasas/metabolismo , Proteínas de Escherichia coli/genética , Oxidación-Reducción , Péptidos/aislamiento & purificación , Péptidos/uso terapéutico , Periplasma/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Ponzoñas/química
4.
Microb Cell Fact ; 16(1): 4, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-28093085

RESUMEN

BACKGROUND: Animal venoms are large, complex libraries of bioactive, disulphide-rich peptides. These peptides, and their novel biological activities, are of increasing pharmacological and therapeutic importance. However, recombinant expression of venom peptides in Escherichia coli remains difficult due to the significant number of cysteine residues requiring effective post-translational processing. There is also an urgent need to develop high-throughput recombinant protocols applicable to the production of reticulated peptides to enable efficient screening of their drug potential. Here, a comprehensive study was developed to investigate how synthetic gene design, choice of fusion tag, compartment of expression, tag removal conditions and protease recognition site affect levels of solubility of oxidized venom peptides produced in E. coli. RESULTS: The data revealed that expression of venom peptides imposes significant pressure on cysteine codon selection. DsbC was the best fusion tag for venom peptide expression, in particular when the fusion was directed to the bacterial periplasm. While the redox activity of DsbC was not essential to maximize expression of recombinant fusion proteins, redox activity did lead to higher levels of correctly folded target peptides. With the exception of proline, the canonical TEV protease recognition site tolerated all other residues at its C-terminus, confirming that no non-native residues, which might affect activity, need to be incorporated at the N-terminus of recombinant peptides for tag removal. CONCLUSIONS: This study reveals that E. coli is a convenient heterologous host for the expression of soluble and functional venom peptides. Using the optimal construct design, a large and diverse range of animal venom peptides were produced in the µM scale. These results open up new possibilities for the high-throughput production of recombinant disulphide-rich peptides in E. coli.


Asunto(s)
Endopeptidasas/metabolismo , Escherichia coli/genética , Biosíntesis de Péptidos , Péptidos/genética , Ponzoñas/biosíntesis , Ponzoñas/genética , Animales , Biotecnología/métodos , Clonación Molecular , Disulfuros/química , Endopeptidasas/química , Vectores Genéticos , Ensayos Analíticos de Alto Rendimiento , Oxidación-Reducción , Péptidos/química , Péptidos/aislamiento & purificación , Periplasma/química , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Solubilidad , Ponzoñas/química , Ponzoñas/metabolismo
5.
Elife ; 112022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35311641

RESUMEN

Staphylococcus aureus (SA) leukocidin ED (LukED) belongs to a family of bicomponent pore forming toxins that play important roles in SA immune evasion and nutrient acquisition. LukED targets specific G protein-coupled chemokine receptors to lyse human erythrocytes (red blood cells) and leukocytes (white blood cells). The first recognition step of receptors is critical for specific cell targeting and lysis. The structural and molecular bases for this mechanism are not well understood but could constitute essential information to guide antibiotic development. Here, we characterized the interaction of LukE with chemokine receptors ACKR1, CCR2, and CCR5 using a combination of structural, pharmacological, and computational approaches. First, crystal structures of LukE in complex with a small molecule mimicking sulfotyrosine side chain (p-cresyl sulfate) and with peptides containing sulfotyrosines issued from receptor sequences revealed the location of receptor sulfotyrosine binding sites in the toxins. Then, by combining previous and novel experimental data with protein docking, classical and accelerated weight histogram (AWH) molecular dynamics we propose models of the ACKR1-LukE and CCR5-LukE complexes. This work provides novel insights into chemokine receptor recognition by leukotoxins and suggests that the conserved sulfotyrosine binding pocket could be a target of choice for future drug development.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Evasión Inmune , Leucocidinas/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Staphylococcus aureus/genética
6.
Methods Mol Biol ; 2025: 165-190, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31267452

RESUMEN

High-throughput production (HTP) of synthetic genes is becoming an important tool to explore the biological function of the extensive genomic and meta-genomic information currently available from various sources. One such source is animal venom, which contains thousands of novel bioactive peptides with potential uses as novel therapeutics to treat a plethora of diseases as well as in environmentally benign bioinsecticide formulations. Here, we describe a HTP platform for recombinant bacterial production of oxidized disulfide-rich proteins and peptides from animal venoms. High-throughput, host-optimized, gene synthesis and subcloning, combined with robust HTP expression and purification protocols, generate a semiautomated pipeline for the accelerated production of proteins and peptides identified from genomic or transcriptomic libraries. The platform has been applied to the production of thousands of animal venom peptide toxins for the purposes of drug discovery, but has the power to be universally applicable for high-level production of various and diverse target proteins in soluble form. This chapter details the HTP protocol for gene synthesis and production, which supported high levels of peptide expression in the E. coli periplasm using a cleavable DsbC fusion. Finally, target proteins and peptides are purified using automated HTP methods, before undergoing quality control and screening.


Asunto(s)
Escherichia coli/metabolismo , Animales , Disulfuros/metabolismo , Escherichia coli/genética , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ponzoñas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA