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 Biol Chem ; 294(30): 11473-11485, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31175158

RESUMEN

DHX9/RNA helicase A (RHA) is a host RNA helicase that participates in many critical steps of the HIV-1 life cycle. It co-assembles with the viral RNA genome into the capsid core. Virions deficient in RHA are less infectious as a result of reduced reverse transcription efficiency, demonstrating that the virion-associated RHA promotes reverse transcription before the virion gains access to the new host's RHA. Here, we quantified reverse-transcription intermediates in HIV-1-infected T cells to clarify the mechanism by which RHA enhances HIV-1 reverse transcription efficiency. Consistently, purified recombinant human RHA promoted reverse transcription efficiency under in vitro conditions that mimic the early reverse transcription steps prior to capsid core uncoating. We did not observe RHA-mediated structural remodeling of the tRNALys3-viral RNA-annealed complex. RHA did not enhance the DNA synthesis rate until incorporation of the first few nucleotides, suggesting that RHA participates primarily in the elongation phase of reverse transcription. Pre-steady-state and steady-state kinetic studies revealed that RHA has little impact on the kinetics of single-nucleotide incorporation. Primer extension assays performed in the presence of trap dsDNA disclosed that RHA enhances the processivity of HIV-1 reverse transcriptase (RT). The biochemical assays used here effectively reflected and explained the low RT activity in HIV-1 virions produced from RHA-depleted cells. Moreover, RT activity in our assays indicated that RHA in HIV-1 virions is required for the efficient catalysis of (-)cDNA synthesis during viral infection before capsid uncoating. Our study identifies RHA as a processivity factor of HIV-1 RT.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Transcriptasa Inversa del VIH/metabolismo , VIH-1/patogenicidad , Interacciones Huésped-Patógeno , Proteínas de Neoplasias/fisiología , ARN/metabolismo , Virión/fisiología , Células HEK293 , VIH-1/genética , Humanos , Cinética , Transcripción Reversa
2.
Vet Immunol Immunopathol ; 185: 20-33, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28241999

RESUMEN

A reverse vaccinology system, Vaxign, was used to identify and select a subset of five African Swine Fever (ASF) antigens that were successfully purified from human embryonic kidney 293 (HEK) cells and produced in Modified vaccinia virus Ankara (MVA) viral vectors. Three HEK-purified antigens [B646L (p72), E183L (p54), and O61R (p12)], and three MVA-vectored antigens [B646L, EP153R, and EP402R (CD2v)] were evaluated using a prime-boost immunization regimen swine safety and immunogenicity study. Antibody responses were detected in pigs following prime-boost immunization four weeks apart with the HEK-293-purified p72, p54, and p12 antigens. Notably, sera from the vaccinees were positive by immunofluorescence on ASFV (Georgia 2007/1)-infected primary macrophages. Although MVA-vectored p72, CD2v, and EP153R failed to induce antibody responses, interferon-gamma (IFN-γ+) spot forming cell responses against all three antigens were detected one week post-boost. The highest IFN-γ+ spot forming cell responses were detected against p72 in pigs primed with MVA-p72 and boosted with the recombinant p72. Antigen-specific (p12, p72, CD2v, and EP153R) T-cell proliferative responses were also detected post-boost. Collectively, these results are the first demonstration that ASFV subunit antigens purified from mammalian cells or expressed in MVA vectors are safe and can induce ASFV-specific antibody and T-cell responses following a prime-boost immunization regimen in swine.


Asunto(s)
Virus de la Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/prevención & control , Antígenos Virales/inmunología , Vacunas Virales/inmunología , Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/genética , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Vectores Genéticos , Genoma Viral , Células HEK293 , Humanos , Inmunogenicidad Vacunal , Masculino , Porcinos , Linfocitos T/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Virus Vaccinia/genética , Vacunas Virales/genética
3.
ACS Med Chem Lett ; 7(12): 1092-1096, 2016 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-27994744

RESUMEN

Aberrant activation of the complement system is associated with diseases, including paroxysmal nocturnal hemoglobinuria and age-related macular degeneration. Complement factor D is the rate-limiting enzyme for activating the alternative pathway in the complement system. Recent development led to a class of potent amide containing pyrrolidine derived factor D inhibitors. Here, we used biochemical enzymatic and biolayer interferometry assays to demonstrate that the amide group improves the inhibitor potency by more than 80-fold. Our crystal structures revealed buried hydrogen bond interactions are important. Molecular orbital analysis from quantum chemistry calculations dissects the chemical groups participating in these interactions. Free energy calculation supports the differential contributions of the amide group to the binding affinities of these inhibitors. Cell-based hemolysis assay confirmed these compounds inhibit factor D mediated complement activation via the alternative pathway. Our study highlights the important interactions contributing to the high potency of factor D inhibitors reported recently.

4.
J Mol Biol ; 428(11): 2418-2429, 2016 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-27107641

RESUMEN

Cellular RNA-binding proteins incorporated into virions during human immunodeficiency virus type 1 (HIV-1) assembly promote the replication efficiency of progeny virions. Despite its critical role in bolstering virion infectivity, the molecular basis for the incorporation of DHX9/RNA helicase A (RHA) to virions remains unclear. Here, cell-based experiments demonstrate that the truncation of segments of the HIV-1 5'-untranslated region (5'-UTR) distinct from the core encapsidation sequence eliminated virion incorporation of RHA, indicating that RHA recruitment is mediated by specific interactions with the HIV-1 5'-UTR. In agreement with biological data, isothermal titration calorimetry determined that the dimer conformation of the 5'-UTR binds one RHA molecule per RNA strand, and the interaction is independent of nucleocapsid protein binding. NMR spectra employing a deuterium-labeling approach enabled resolution of the dimeric 5'-UTR in complex with the RHA N-terminal domain. The structure of the large molecular mass complex was dependent on RHA binding to a double-stranded region of the primer binding site (PBS)-segment of the 5'-UTR. A single A-to-C substitution was sufficient to disrupt biophysical conformation and attenuate virion infectivity in cell-based assays. Taken together, our studies demonstrate the structural basis for HIV-1 genomic RNA to recruit beneficial cellular cofactor to virions. The support of progeny virion infectivity by RHA is attributable to structure-dependent binding at the PBS-segment of the HIV-1 5'-UTR during virus assembly.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Cartilla de ADN/genética , Infecciones por VIH/genética , VIH-1/genética , Proteínas de Neoplasias/metabolismo , ARN Viral/genética , Virión/genética , Ensamble de Virus/genética , Regiones no Traducidas 5'/genética , Sitios de Unión/genética , Genómica , Infecciones por VIH/virología , Humanos , Replicación Viral/genética
5.
PLoS One ; 11(1): e0146522, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26735493

RESUMEN

Suppression of Tumorigenicity 2 (ST2), a member of the interleukin-1 receptor (IL-1R) family, activates type 2 immune responses to pathogens and tissue damage via binding to IL-33. Dysregulated responses contribute to asthma, graft-versus-host and autoinflammatory diseases and disorders. To study ST2 structure for inhibitor development, we performed the principal component (PC) analysis on the crystal structures of IL1-1R1, IL1-1R2, ST2 and the refined ST2 ectodomain (ST2ECD) models, constructed from previously reported small-angle X-ray scattering data. The analysis facilitates mapping of the ST2ECD conformations to PC subspace for characterizing structural changes. Extensive coverage of ST2ECD conformations was then obtained using the accelerated molecular dynamics simulations started with the IL-33 bound ST2ECD structure as instructed by their projected locations on the PC subspace. Cluster analysis of all conformations further determined representative conformations of ST2ECD ensemble in solution. Alignment of the representative conformations with the ST2/IL-33 structure showed that the D3 domain of ST2ECD (containing D1-D3 domains) in most conformations exhibits no clashes with IL-33 in the crystal structure. Our experimental binding data informed that the D1-D2 domain of ST2ECD contributes predominantly to the interaction between ST2ECD and IL-33 underscoring the importance of the D1-D2 domain in binding. Computational binding site assessment revealed one third of the total detected binding sites in the representative conformations may be suitable for binding to potent small molecules. Locations of these sites include the D1-D2 domain ST2ECD and modulation sites conformed to ST2ECD conformations. Our study provides structural models and analyses of ST2ECD that could be useful for inhibitor discovery.


Asunto(s)
Receptores de Superficie Celular/metabolismo , Sitios de Unión , Análisis por Conglomerados , Cristalografía por Rayos X , Humanos , Interferometría , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33/química , Interleucina-33/metabolismo , Simulación de Dinámica Molecular , Análisis de Componente Principal , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
6.
J Biol Chem ; 290(9): 5226-39, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25568319

RESUMEN

PLP synthase (PLPS) is a remarkable single-enzyme biosynthetic pathway that produces pyridoxal 5'-phosphate (PLP) from glutamine, ribose 5-phosphate, and glyceraldehyde 3-phosphate. The intact enzyme includes 12 synthase and 12 glutaminase subunits. PLP synthesis occurs in the synthase active site by a complicated mechanism involving at least two covalent intermediates at a catalytic lysine. The first intermediate forms with ribose 5-phosphate. The glutaminase subunit is a glutamine amidotransferase that hydrolyzes glutamine and channels ammonia to the synthase active site. Ammonia attack on the first covalent intermediate forms the second intermediate. Glyceraldehyde 3-phosphate reacts with the second intermediate to form PLP. To investigate the mechanism of the synthase subunit, crystal structures were obtained for three intermediate states of the Geobacillus stearothermophilus intact PLPS or its synthase subunit. The structures capture the synthase active site at three distinct steps in its complicated catalytic cycle, provide insights into the elusive mechanism, and illustrate the coordinated motions within the synthase subunit that separate the catalytic states. In the intact PLPS with a Michaelis-like intermediate in the glutaminase active site, the first covalent intermediate of the synthase is fully sequestered within the enzyme by the ordering of a generally disordered 20-residue C-terminal tail. Following addition of ammonia, the synthase active site opens and admits the Lys-149 side chain, which participates in formation of the second intermediate and PLP. Roles are identified for conserved Asp-24 in the formation of the first intermediate and for conserved Arg-147 in the conversion of the first to the second intermediate.


Asunto(s)
Proteínas Bacterianas/química , Geobacillus stearothermophilus/enzimología , Glutaminasa/química , Fosfato de Piridoxal/química , Amoníaco/química , Amoníaco/metabolismo , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biocatálisis , Vías Biosintéticas , Dominio Catalítico , Cristalografía por Rayos X , Geobacillus stearothermophilus/genética , Glutaminasa/genética , Glutaminasa/metabolismo , Glutamina/química , Glutamina/metabolismo , Gliceraldehído 3-Fosfato/química , Gliceraldehído 3-Fosfato/metabolismo , Cinética , Lisina/química , Lisina/metabolismo , Modelos Moleculares , Estructura Molecular , Mutación , Conformación Proteica , Fosfato de Piridoxal/metabolismo , Ribosamonofosfatos/química , Ribosamonofosfatos/metabolismo , Espectrometría de Masa por Ionización de Electrospray
7.
Protein Sci ; 21(2): 239-48, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22124946

RESUMEN

DmmA is a haloalkane dehalogenase (HLD) identified and characterized from the metagenomic DNA of a marine microbial consortium. Dehalogenase activity was detected with 1,3-dibromopropane as substrate, with steady-state kinetic parameters typical of HLDs (K(m) = 0.24 ± 0.05 mM, k(cat) = 2.4 ± 0.1 s(-1) ). The 2.2-Å crystal structure of DmmA revealed a fold and active site similar to other HLDs, but with a substantially larger active site binding pocket, suggestive of an ability to act on bulky substrates. This enhanced cavity was shown to accept a range of linear and cyclic substrates, suggesting that DmmA will contribute to the expanding industrial applications of HLDs.


Asunto(s)
Organismos Acuáticos/enzimología , Hidrolasas/química , Hidrolasas/metabolismo , Animales , Organismos Acuáticos/química , Sitios de Unión , Cristalografía por Rayos X , Cianobacterias/química , Cianobacterias/enzimología , Cianobacterias/metabolismo , Activación Enzimática , Cinética , Modelos Biológicos , Modelos Moleculares , Multimerización de Proteína/fisiología , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Especificidad por Sustrato
8.
Protein Expr Purif ; 77(1): 34-45, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21262364

RESUMEN

Biomedical research has undergone a major shift in emphasis over the past decade from characterizing the genomes of organisms to characterizing their proteomes. The high-throughput approaches that were successfully applied to sequencing of genomes, such as miniaturization and automation, have been adapted for high-throughput cloning and protein production. High-throughput platforms allow for a multi-construct, multi-parallel approach to expression optimization and construct evaluation. We describe here a series of baculovirus transfer and expression vectors that contain ligation-independent cloning regions originally designed for use in high-throughput Escherichia coli expression evaluation. These new vectors allow for parallel cloning of the same gene construct into a variety of baculovirus or E. coli expression vectors. A high-throughput platform for construct expression evaluation in baculovirus-infected insect cells was developed to utilize these vectors. Data from baculovirus infection expression trials for multiple constructs of two target protein systems relevant to the study of human diseases are presented. The target proteins exhibit a wide variation in behavior and illustrate the benefit of investigating multiple cell types, fusion partners and secretion signals in optimization of constructs and conditions for eukaryotic protein production.


Asunto(s)
Baculoviridae/genética , Clonación Molecular/métodos , ADN Recombinante/administración & dosificación , Spodoptera/metabolismo , Spodoptera/virología , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Línea Celular , ADN Recombinante/genética , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/metabolismo , Vectores Genéticos/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Reproducibilidad de los Resultados , Proteínas no Estructurales Virales/biosíntesis , Proteínas no Estructurales Virales/genética
9.
J Biol Chem ; 285(7): 4520-35, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19959473

RESUMEN

Major histocompatibility complex (MHC) class I molecules are ligands for T-cell receptors of CD8(+) T cells and inhibitory receptors of natural killer cells. Assembly of the heavy chain, light chain, and peptide components of MHC class I molecules occurs in the endoplasmic reticulum (ER). Specific assembly factors and generic ER chaperones, collectively called the MHC class I peptide loading complex (PLC), are required for MHC class I assembly. Calreticulin has an important role within the PLC and induces MHC class I cell surface expression, but the interactions and mechanisms involved are incompletely understood. We show that interactions with the thiol oxidoreductase ERp57 and substrate glycans are important for the recruitment of calreticulin into the PLC and for its functional activities in MHC class I assembly. The glycan and ERp57 binding sites of calreticulin contribute directly or indirectly to complexes between calreticulin and the MHC class I assembly factor tapasin and are important for maintaining steady-state levels of both tapasin and MHC class I heavy chains. A number of destabilizing conditions and mutations induce generic polypeptide binding sites on calreticulin and contribute to calreticulin-mediated suppression of misfolded protein aggregation in vitro. We show that generic polypeptide binding sites per se are insufficient for stable recruitment of calreticulin to PLC substrates in cells. However, such binding sites could contribute to substrate stabilization in a step that follows the glycan and ERp57-dependent recruitment of calreticulin to the PLC.


Asunto(s)
Calreticulina/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Transducción de Señal/fisiología , Animales , Calreticulina/genética , Línea Celular , Cromatografía en Gel , Citometría de Flujo , Vectores Genéticos , Humanos , Inmunoprecipitación , Ratones , Mutagénesis Sitio-Dirigida , Polisacáridos/metabolismo , Unión Proteica/genética , Unión Proteica/fisiología , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Transducción de Señal/genética , Relación Estructura-Actividad
10.
Protein Expr Purif ; 63(1): 40-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18824232

RESUMEN

A persistent problem in heterologous protein production is insolubility of the target protein when expressed to high level in the host cell. A widely employed strategy for overcoming this problem is the use of fusion tags. The best fusion tags promote solubility, may function as purification handles and either do not interfere with downstream applications or may be removed from the passenger protein preparation. A novel fusion tag is identified that meets these criteria. This fusion tag is a monomeric mutant of the Ocr protein (0.3 gene product) of bacteriophage T7. This fusion tag displays solubilizing activity with a variety of different passenger proteins. We show that it may be used as a purification handle similar to other fusion tags. Its small size and compact structure are compatible with its use in downstream applications of the passenger protein or it may be removed and purified away from the passenger protein. The use of monomeric Ocr (Mocr) as a complement to other fusion tags such as maltose-binding protein will provide greater flexibility in protein production and processing for a wide variety of protein applications.


Asunto(s)
Proteínas Virales/química , Sustitución de Aminoácidos , Cromatografía Liquida , Clonación Molecular , Etanolaminas , Reacción en Cadena de la Polimerasa , Análisis por Matrices de Proteínas , Estructura Cuaternaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Secuencia de Proteína , Solubilidad , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA