Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 184(5): 1232-1244.e16, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33626330

RESUMEN

Human cytomegalovirus (HCMV) infects the majority of the human population and represents the leading viral cause of congenital birth defects. HCMV utilizes the glycoproteins gHgLgO (Trimer) to bind to platelet-derived growth factor receptor alpha (PDGFRα) and transforming growth factor beta receptor 3 (TGFßR3) to gain entry into multiple cell types. This complex is targeted by potent neutralizing antibodies and represents an important candidate for therapeutics against HCMV. Here, we determine three cryogenic electron microscopy (cryo-EM) structures of the trimer and the details of its interactions with four binding partners: the receptor proteins PDGFRα and TGFßR3 as well as two broadly neutralizing antibodies. Trimer binding to PDGFRα and TGFßR3 is mutually exclusive, suggesting that they function as independent entry receptors. In addition, Trimer-PDGFRα interaction has an inhibitory effect on PDGFRα signaling. Our results provide a framework for understanding HCMV receptor engagement, neutralization, and the development of anti-viral strategies against HCMV.


Asunto(s)
Citomegalovirus/química , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral/química , Internalización del Virus , Microscopía por Crioelectrón , Citomegalovirus/fisiología , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Proteoglicanos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/química , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas del Envoltorio Viral/metabolismo
2.
MAbs ; 11(6): 1122-1138, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31122132

RESUMEN

IgA antibodies have broad potential as a novel therapeutic platform based on their superior receptor-mediated cytotoxic activity, potent neutralization of pathogens, and ability to transcytose across mucosal barriers via polymeric immunoglobulin receptor (pIgR)-mediated transport, compared to traditional IgG-based drugs. However, the transition of IgA into clinical development has been challenged by complex expression and characterization, as well as rapid serum clearance that is thought to be mediated by glycan receptor scavenging of recombinantly produced IgA monomer bearing incompletely sialylated N-linked glycans. Here, we present a comprehensive biochemical, biophysical, and structural characterization of recombinantly produced monomeric, dimeric and polymeric human IgA. We further explore two strategies to overcome the rapid serum clearance of polymeric IgA: removal of all N-linked glycosylation sites creating an aglycosylated polymeric IgA and engineering in FcRn binding with the generation of a polymeric IgG-IgA Fc fusion. While previous reports and the results presented in this study indicate that glycan-mediated clearance plays a major role for monomeric IgA, systemic clearance of polymeric IgA in mice is predominantly controlled by mechanisms other than glycan receptor clearance, such as pIgR-mediated transcytosis. The developed IgA platform now provides the potential to specifically target pIgR expressing tissues, while maintaining low systemic exposure.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/genética , Perros , Femenino , Glicosilación , Semivida , Humanos , Inmunoglobulina A/genética , Inmunoglobulina G/genética , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética
3.
Mol Pharm ; 15(10): 4529-4537, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30118239

RESUMEN

A critical part of the clinical development path for a therapeutic antibody involves evaluating the physical and chemical stability of candidate molecules throughout the manufacturing process. In particular, the risks of chemical liabilities that can impact antigen binding, such as deamidation, oxidation, and isomerization in the antibody CDR sequences, need to be controlled through formulation development or eliminated by replacing the amino acid motif displaying the chemical instability. Commonly, the antibody CDR sequence contains multiple sequence motifs (potential hotspots) for chemical instability. However, only a subset of these motifs results in actual chemical modification, and thus, experimental assessment of the extent of instability is necessary to identify positions for potential sequence engineering. Ideally, this information should be available prior to antibody humanization at the stage of parental rodent antibody identification. Early knowledge of liabilities allows for ranking of clones or the mitigation of liabilities by concurrent engineering with the antibody humanization process instead of time-consuming sequential activities. However, concurrent engineering of chemical liabilities and humanization requires translatability of the chemical modifications from the rodent parental antibody to the humanized. We experimentally compared the stability of all sequence motifs by mass spectrometric peptide mapping between the rodent parental antibody and the final humanized antibody and observed a linear correlation. These results have enabled a streamlined developability assessment process for therapeutic antibodies from lead discovery to clinical development.


Asunto(s)
Anticuerpos/inmunología , Secuencia de Aminoácidos , Animales , Cromatografía Liquida , Desaminación , Concentración de Iones de Hidrógeno , Isomerismo , Metionina/química , Ratones , Oxidación-Reducción , Espectrometría de Masas en Tándem , Triptófano/química
4.
Sci Rep ; 8(1): 7136, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29740124

RESUMEN

Outer membrane proteins (OMPs) in Gram-negative bacteria are essential for a number of cellular functions including nutrient transport and drug efflux. Escherichia coli BamA is an essential component of the OMP ß-barrel assembly machinery and a potential novel antibacterial target that has been proposed to undergo large (~15 Å) conformational changes. Here, we explored methods to isolate anti-BamA monoclonal antibodies (mAbs) that might alter the function of this OMP and ultimately lead to bacterial growth inhibition. We first optimized traditional immunization approaches but failed to identify mAbs that altered cell growth after screening >3000 hybridomas. We then developed a "targeted boost-and-sort" strategy that combines bacterial cell immunizations, purified BamA protein boosts, and single hybridoma cell sorting using amphipol-reconstituted BamA antigen. This unique workflow improves the discovery efficiency of FACS + mAbs by >600-fold and enabled the identification of rare anti-BamA mAbs with bacterial growth inhibitory activity in the presence of a truncated lipopolysaccharide layer. These mAbs represent novel tools for dissecting the BamA-mediated mechanism of ß-barrel folding and our workflow establishes a new template for the efficient discovery of novel mAbs against other highly dynamic membrane proteins.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/aislamiento & purificación , Proteínas de la Membrana Bacteriana Externa/inmunología , Escherichia coli/inmunología , Proteínas de Escherichia coli/inmunología , Inmunización , Conformación Proteica , Pliegue de Proteína , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Vacunación
5.
MAbs ; 10(4): 624-635, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29494273

RESUMEN

To rapidly find "best-in-class" antibody therapeutics, it has become essential to develop high throughput (HTP) processes that allow rapid assessment of antibodies for functional and molecular properties. Consequently, it is critical to have access to sufficient amounts of high quality antibody, to carry out accurate and quantitative characterization. We have developed automated workflows using liquid handling systems to conduct affinity-based purification either in batch or tip column mode. Here, we demonstrate the capability to purify >2000 antibodies per day from microscale (1 mL) cultures. Our optimized, automated process for human IgG1 purification using MabSelect SuRe resin achieves ∼70% recovery over a wide range of antibody loads, up to 500 µg. This HTP process works well for hybridoma-derived antibodies that can be purified by MabSelect SuRe resin. For rat IgG2a, which is often encountered in hybridoma cultures and is challenging to purify via an HTP process, we established automated purification with GammaBind Plus resin. Using these HTP purification processes, we can efficiently recover sufficient amounts of antibodies from mammalian transient or hybridoma cultures with quality comparable to conventional column purification.


Asunto(s)
Anticuerpos Monoclonales/análisis , Cromatografía de Afinidad/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Inmunoglobulina G/análisis , Animales , Humanos , Ratas
6.
Sci Rep ; 7(1): 3026, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28596531

RESUMEN

Amber codon suppression for the insertion of non-natural amino acids (nnAAs) is limited by competition with release factor 1 (RF1). Here we describe the genome engineering of a RF1 mutant strain that enhances suppression efficiency during cell-free protein synthesis, without significantly impacting cell growth during biomass production. Specifically, an out membrane protease (OmpT) cleavage site was engineered into the switch loop of RF1, which enables its conditional inactivation during cell lysis. This facilitates extract production without additional processing steps, resulting in a scaleable extract production process. The RF1 mutant extract allows nnAA incorporation at previously intractable sites of an IgG1 and at multiple sites in the same polypeptide chain. Conjugation of cytotoxic agents to these nnAAs, yields homogeneous antibody drug conjugates (ADCs) that can be optimized for conjugation site, drug to antibody ratio (DAR) and linker-warheads designed for efficient tumor killing. This platform provides the means to generate therapeutic ADCs inaccessible by other methods that are efficient in their cytotoxin delivery to tumor with reduced dose-limiting toxicities and thus have the potential for better clinical impact.


Asunto(s)
Aminoácidos/química , Inmunoconjugados , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/genética , Ingeniería de Proteínas , Sitios de Unión , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromatografía Liquida , Codón de Terminación , Estabilidad de Medicamentos , Humanos , Inmunoconjugados/química , Inmunoconjugados/aislamiento & purificación , Inmunoconjugados/metabolismo , Inmunoconjugados/farmacología , Inmunoglobulina G/química , Inmunoglobulina G/farmacología , Espectrometría de Masas , Modelos Moleculares , Mutación , Factores de Terminación de Péptidos/metabolismo , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad , Trastuzumab/química , Trastuzumab/farmacología
7.
Int J Biol Macromol ; 92: 779-787, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27402461

RESUMEN

A method for development of murine Fab fragments towards extracellular domains of a surface receptor is presented. The GluA4 ionotropic glutamate receptor is used as a model system. Recombinant GluA4 ectodomain comprising both the N-terminal domain (NTD) and the ligand-binding domain (LBD) in one molecule was used for immunization. A Fab-phage library was constructed and a parallel panning approach enabled selection of murine Fab fragments towards either intact ectodomain or the isolated LBD of the GluA4 receptor. One LBD-Fab (FabL9) showed exclusive selectivity for the GluA4 LBD, over a panel of LBDs from GluA2, GluK1, GluK2 and GluD2. Soluble FabL9 was produced in amounts suitable for characterization. Competitive ELISA and rat-brain immunoprecipitation experiments confirmed that the FabL9 epitope is conserved in the LBD and in the intact native receptor. By an alignment of GluA2 and GluA4, the likely binding epitope for FabL9 was predicted. This study demonstrates a simple approach for development of antibody fragments towards specific sub-domains of a large ligand-gated ion channel, and this method could be utilized for all multi-domain surface receptors where antibody domain-selectivity may be desirable. Furthermore, we present for the first time a GluA4 subtype-specific murine Fab fragment targeting the LBD of the receptor.


Asunto(s)
Epítopos/química , Fragmentos Fab de Inmunoglobulinas/aislamiento & purificación , Biblioteca de Péptidos , Receptores AMPA/química , Secuencia de Aminoácidos , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Química Encefálica , Células Clonales , Ensayo de Inmunoadsorción Enzimática/métodos , Epítopos/inmunología , Femenino , Inmunización , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Inmunoprecipitación , Ratones , Ratones Endogámicos BALB C , Dominios Proteicos , Multimerización de Proteína , Ratas , Receptores AMPA/administración & dosificación , Receptores AMPA/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Alineación de Secuencia
8.
J Biol Chem ; 291(27): 13974-13986, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27129258

RESUMEN

The prominent role of voltage-gated sodium channel 1.7 (Nav1.7) in nociception was revealed by remarkable human clinical and genetic evidence. Development of potent and subtype-selective inhibitors of this ion channel is crucial for obtaining therapeutically useful analgesic compounds. Microproteins isolated from animal venoms have been identified as promising therapeutic leads for ion channels, because they naturally evolved to be potent ion channel blockers. Here, we report the engineering of highly potent and selective inhibitors of the Nav1.7 channel based on tarantula ceratotoxin-1 (CcoTx1). We utilized a combination of directed evolution, saturation mutagenesis, chemical modification, and rational drug design to obtain higher potency and selectivity to the Nav1.7 channel. The resulting microproteins are highly potent (IC50 to Nav1.7 of 2.5 nm) and selective. We achieved 80- and 20-fold selectivity over the closely related Nav1.2 and Nav1.6 channels, respectively, and the IC50 on skeletal (Nav1.4) and cardiac (Nav1.5) sodium channels is above 3000 nm The lead molecules have the potential for future clinical development as novel therapeutics in the treatment of pain.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/química , Manejo del Dolor/métodos , Ingeniería de Proteínas , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Células HEK293 , Humanos , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Técnicas de Placa-Clamp , Filogenia , Venenos de Araña/química
9.
Protein Eng Des Sel ; 27(4): 97-109, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24586053

RESUMEN

Selection technologies such as ribosome display enable the rapid discovery of novel antibody fragments entirely in vitro. It has been assumed that the open nature of the cell-free reactions used in these technologies limits selections to single-chain protein fragments. We present a simple approach for the selection of multi-chain proteins, such as antibody Fab fragments, using ribosome display. Specifically, we show that a two-chain trastuzumab (Herceptin) Fab domain can be displayed in a format which tethers either the heavy or light chain to the ribosome while retaining functional antigen binding. Then, we constructed synthetic Fab HC and LC libraries and performed test selections against carcinoembryonic antigen (CEA) and vascular endothelial growth factor (VEGF). The Fab selection output was reformatted into full-length immunoglobulin Gs (IgGs) and directly expressed at high levels in an optimized cell-free system for immediate screening, purification and characterization. Several novel IgGs were identified using this cell-free platform that bind to purified CEA, CEA positive cells and VEGF.


Asunto(s)
Técnicas de Visualización de Superficie Celular/métodos , Sistema Libre de Células , Fragmentos Fab de Inmunoglobulinas , Biblioteca de Péptidos , Anticuerpos/genética , Anticuerpos Monoclonales Humanizados/genética , Antígeno Carcinoembrionario/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Trastuzumab , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Bioconjug Chem ; 25(2): 351-61, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24437342

RESUMEN

Antibody-drug conjugates (ADCs) are a targeted chemotherapeutic currently at the cutting edge of oncology medicine. These hybrid molecules consist of a tumor antigen-specific antibody coupled to a chemotherapeutic small molecule. Through targeted delivery of potent cytotoxins, ADCs exhibit improved therapeutic index and enhanced efficacy relative to traditional chemotherapies and monoclonal antibody therapies. The currently FDA-approved ADCs, Kadcyla (Immunogen/Roche) and Adcetris (Seattle Genetics), are produced by conjugation to surface-exposed lysines, or partial disulfide reduction and conjugation to free cysteines, respectively. These stochastic modes of conjugation lead to heterogeneous drug products with varied numbers of drugs conjugated across several possible sites. As a consequence, the field has limited understanding of the relationships between the site and extent of drug loading and ADC attributes such as efficacy, safety, pharmacokinetics, and immunogenicity. A robust platform for rapid production of ADCs with defined and uniform sites of drug conjugation would enable such studies. We have established a cell-free protein expression system for production of antibody drug conjugates through site-specific incorporation of the optimized non-natural amino acid, para-azidomethyl-l-phenylalanine (pAMF). By using our cell-free protein synthesis platform to directly screen a library of aaRS variants, we have discovered a novel variant of the Methanococcus jannaschii tyrosyl tRNA synthetase (TyrRS), with a high activity and specificity toward pAMF. We demonstrate that site-specific incorporation of pAMF facilitates near complete conjugation of a DBCO-PEG-monomethyl auristatin (DBCO-PEG-MMAF) drug to the tumor-specific, Her2-binding IgG Trastuzumab using strain-promoted azide-alkyne cycloaddition (SPAAC) copper-free click chemistry. The resultant ADCs proved highly potent in in vitro cell cytotoxicity assays.


Asunto(s)
Aminoácidos/química , Inmunoconjugados/química , Línea Celular , Sistema Libre de Células , Cromatografía Liquida , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunoconjugados/farmacología , Espectrometría de Masas en Tándem
11.
Nature ; 505(7482): 239-43, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24291791

RESUMEN

The increasing demands placed on natural resources for fuel and food production require that we explore the use of efficient, sustainable feedstocks such as brown macroalgae. The full potential of brown macroalgae as feedstocks for commercial-scale fuel ethanol production, however, requires extensive re-engineering of the alginate and mannitol catabolic pathways in the standard industrial microbe Saccharomyces cerevisiae. Here we present the discovery of an alginate monomer (4-deoxy-L-erythro-5-hexoseulose uronate, or DEHU) transporter from the alginolytic eukaryote Asteromyces cruciatus. The genomic integration and overexpression of the gene encoding this transporter, together with the necessary bacterial alginate and deregulated native mannitol catabolism genes, conferred the ability of an S. cerevisiae strain to efficiently metabolize DEHU and mannitol. When this platform was further adapted to grow on mannitol and DEHU under anaerobic conditions, it was capable of ethanol fermentation from mannitol and DEHU, achieving titres of 4.6% (v/v) (36.2 g l(-1)) and yields up to 83% of the maximum theoretical yield from consumed sugars. These results show that all major sugars in brown macroalgae can be used as feedstocks for biofuels and value-added renewable chemicals in a manner that is comparable to traditional arable-land-based feedstocks.


Asunto(s)
Biocombustibles/provisión & distribución , Metabolismo de los Hidratos de Carbono , Etanol/metabolismo , Ingeniería Genética , Phaeophyceae/metabolismo , Saccharomyces cerevisiae/metabolismo , Alginatos/metabolismo , Anaerobiosis , Ascomicetos/genética , Ascomicetos/metabolismo , Biotecnología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Evolución Molecular , Fermentación , Prueba de Complementación Genética , Ácido Glucurónico/metabolismo , Ácidos Hexurónicos/metabolismo , Manitol/metabolismo , Phaeophyceae/genética , Ácido Quínico/metabolismo , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/genética , Algas Marinas/genética , Algas Marinas/metabolismo , Ácidos Urónicos/metabolismo
12.
Front Mol Neurosci ; 4: 56, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22232575

RESUMEN

Ligand-gated ion channels couple the free energy of agonist binding to the gating of selective transmembrane ion pores, permitting cells to regulate ion flux in response to external chemical stimuli. However, the stereochemical mechanisms responsible for this coupling remain obscure. In the case of the ionotropic glutamate receptors (iGluRs), the modular nature of receptor subunits has facilitated structural analysis of the N-terminal domain (NTD), and of multiple conformations of the ligand-binding domain (LBD). Recently, the crystallographic structure of an antagonist-bound form of the receptor was determined. However, disulfide trapping of this conformation blocks channel opening, suggesting that channel activation involves additional quaternary packing arrangements. To explore the conformational space available to iGluR channels, we report here a second, clearly distinct domain architecture of homotetrameric, calcium-permeable AMPA receptors, determined by single-particle electron microscopy of untagged and fluorescently tagged constructs in a ligand-free state. It reveals a novel packing of NTD dimers, and a separation of LBD dimers across a central vestibule. In this arrangement, which reconciles diverse functional observations, agonist-induced cleft closure across LBD dimers can be converted into a twisting motion that provides a basis for receptor activation.

13.
PLoS One ; 6(3): e16788, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21408218

RESUMEN

Human lysozyme is a key component of the innate immune system, and recombinant forms of the enzyme represent promising leads in the search for therapeutic agents able to treat drug-resistant infections. The wild type protein, however, fails to participate effectively in clearance of certain infections due to inherent functional limitations. For example, wild type lysozymes are subject to electrostatic sequestration and inactivation by anionic biopolymers in the infected airway. A charge engineered variant of human lysozyme has recently been shown to possess improved antibacterial activity in the presence of disease associated inhibitory molecules. Here, the 2.04 Å crystal structure of this variant is presented along with an analysis that provides molecular level insights into the origins of the protein's enhanced performance. The charge engineered variant's two mutated amino acids exhibit stabilizing interactions with adjacent native residues, and from a global perspective, the mutations cause no gross structural perturbations or loss of stability. Importantly, the two substitutions dramatically expand the negative electrostatic potential that, in the wild type enzyme, is restricted to a small region near the catalytic residues. The net result is a reduction in the overall strength of the engineered enzyme's electrostatic potential field, and it appears that the specific nature of this remodeled field underlies the variant's reduced susceptibility to inhibition by anionic biopolymers.


Asunto(s)
Antibacterianos/química , Antibacterianos/metabolismo , Muramidasa/química , Muramidasa/metabolismo , Ingeniería de Proteínas , Actinas/metabolismo , Antibacterianos/farmacología , Biocatálisis/efectos de los fármacos , Cristalografía por Rayos X , Estabilidad de Enzimas/efectos de los fármacos , Humanos , Modelos Moleculares , Muramidasa/antagonistas & inhibidores , Muramidasa/farmacología , Proteínas Mutantes/química , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Análisis de Secuencia de Proteína , Electricidad Estática
14.
ACS Chem Biol ; 5(9): 809-18, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20604527

RESUMEN

Lysozymes contain a disproportionately large fraction of cationic residues, and are thereby attracted toward the negatively charged surface of bacterial targets. Importantly, this conserved biophysical property may inhibit lysozyme antibacterial function during acute and chronic infections. A mouse model of acute pulmonary Pseudomonas aeruginosa infection demonstrated that anionic biopolymers accumulate to high concentrations in the infected lung, and the presence of these species correlates with decreased endogenous lysozyme activity. To develop antibacterial enzymes designed specifically to be used as antimicrobial agents in the infected airway, the electrostatic potential of human lysozyme (hLYS) was remodeled by protein engineering. A novel, high-throughput screen was implemented to functionally interrogate combinatorial libraries of charge-engineered hLYS proteins, and variants with improved bactericidal activity were isolated and characterized in detail. These studies illustrate a general mechanism by which polyanions inhibit lysozyme function, and they are the first direct demonstration that decreasing hLYS's net cationic character improves its antibacterial activity in the presence of disease-associated biopolymers. In addition to avoiding electrostatic sequestration, at least one charge-engineered variant also kills bacteria more rapidly in the absence of inhibitory biopolymers; this observation supports a novel hypothesis that tuning the cellular affinity of peptidoglycan hydrolases may be a general strategy for improving kinetics of bacterial killing.


Asunto(s)
Antibacterianos/química , Antibacterianos/uso terapéutico , Bacterias/efectos de los fármacos , Muramidasa/química , Muramidasa/uso terapéutico , Ingeniería de Proteínas , Infecciones por Pseudomonas/tratamiento farmacológico , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Ratones , Micrococcus luteus/efectos de los fármacos , Modelos Moleculares , Muramidasa/genética , Muramidasa/farmacología , Mutación , Pseudomonas aeruginosa/efectos de los fármacos , Electricidad Estática
15.
J Neurosci ; 30(4): 1463-70, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20107073

RESUMEN

AMPA receptors (AMPARs) are tetrameric ligand-gated ion channels that couple the energy of glutamate binding to the opening of a transmembrane channel. Crystallographic and electrophysiological analysis of AMPARs has suggested a coupling between (1) cleft closure in the bilobate ligand-binding domain (LBD), (2) the resulting separation of transmembrane helix attachment points across subunit dimers, and (3) agonist efficacy. In general, more efficacious agonists induce greater degrees of cleft closure and transmembrane separation than partial agonists. Several apparent violations of the cleft-closure/efficacy paradigm have emerged, although in all cases, intradimer separation remains as the driving force for channel opening. Here, we examine the structural basis of partial agonism in GluA4 AMPARs. We find that the L651V substitution enhances the relative efficacy of kainate without increasing either LBD cleft closure or transmembrane separation. Instead, the conformational change relative to the wild-type:kainate complex involves a twisting motion with the efficacy contribution opposite from that expected based on previous analyses. As a result, channel opening may involve transmembrane rearrangements with a significant rotational component. Furthermore, a two-dimensional analysis of agonist-induced GluA2 LBD motions suggests that efficacy is not a linearly varying function of lobe 2 displacement vectors, but is rather determined by specific conformational requirements of the transmembrane domains.


Asunto(s)
Agonistas de Aminoácidos Excitadores/química , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Receptores AMPA/agonistas , Receptores AMPA/química , Transmisión Sináptica/fisiología , Sitios de Unión , Línea Celular , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cristalografía por Rayos X , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Canales Iónicos/agonistas , Canales Iónicos/química , Ácido Kaínico/farmacología , Ligandos , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Proteómica , Receptores AMPA/metabolismo
16.
Biochemistry ; 47(52): 13831-41, 2008 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-19102704

RESUMEN

AMPA receptors are glutamate-gated ion channels that are essential mediators of synaptic signals in the central nervous system. They form tetramers that are assembled as combinations of subunits GluR1-4, each of which contains a ligand-binding domain (LBD). Crystal structures of the GluR2 LBD have revealed an agonist-binding cleft, which is located between two lobes and which acts like a Venus flytrap. In general, agonist efficacy is correlated with the extent of cleft closure. However, recent observations show that cleft closure is not the sole determinant of the relative efficacy for glutamate receptors. In addition, these studies have focused on the GluR2 subunit, which is the specific target of a physiologically important RNA-editing modification in vivo. We therefore sought to test the generality of the cleft closure-efficacy correlation for other AMPA-R subunits. Here, we present crystal structures of the GluR4(flip) LBD in complex with both full and partial agonists. As for GluR2, both agonists stabilize a closed-cleft conformation, and the partial agonist induces a smaller cleft closure than the full agonist. However, a detailed analysis of LBD-kainate interactions reveals the importance of subtle backbone conformational changes in the ligand-binding pocket in determining the magnitude of agonist-associated conformational changes. Furthermore, the GluR4 subunit exhibits a different correlation between receptor activation and LBD cleft closure than does GluR2.


Asunto(s)
Receptores AMPA/agonistas , Receptores AMPA/química , Animales , Sitios de Unión , Cristalografía por Rayos X , Ligandos , Unión Proteica , Conformación Proteica , Ratas , Receptores AMPA/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-18765917

RESUMEN

Glutamate is the major excitatory neurotransmitter in the brain. Among the cognate ionotropic glutamate receptors, the subfamily selective for AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) is responsible for most fast excitatory synaptic signaling and plays key roles in synaptic plasticity. AMPA receptors (AMPA-Rs) have also been implicated in a number of neurological disorders. To investigate subunit-specific differences in the ligand binding and activation of AMPA-Rs, the GluR4 AMPA-R ligand-binding domain (LBD) was crystallized in complex with full and partial agonists. This is the first non-GluR2 AMPA-R LBD available for structural analysis. Standard cryoprotection protocols yielded high-resolution diffraction from flash-cooled crystals of the complex with the full agonist glutamate. However, for cocrystals with the partial agonist kainate, systematic screening and optimization of cryoprotection conditions yielded at best mosaic, weak diffraction at 100 K. In contrast, room-temperature data collection from capillary-mounted kainate cocrystals exhibited reproducible diffraction to better than 3 A resolution. Together, these crystals lay the foundation for a structural comparison of LBD-agonist interactions in distinct AMPA-R subunits.


Asunto(s)
Frío , Receptores AMPA/química , Receptores AMPA/aislamiento & purificación , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo , Animales , Clonación Molecular , Cristalización , Calor , Ligandos , Unión Proteica/fisiología , Conformación Proteica , Estructura Terciaria de Proteína , Ratas , Receptores AMPA/agonistas , Receptores AMPA/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...