Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nature ; 534(7605): 63-8, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27135925

RESUMEN

The physiology of N-methyl-d-aspartate (NMDA) receptors is fundamental to brain development and function. NMDA receptors are ionotropic glutamate receptors that function as heterotetramers composed mainly of GluN1 and GluN2 subunits. Activation of NMDA receptors requires binding of neurotransmitter agonists to a ligand-binding domain (LBD) and structural rearrangement of an amino-terminal domain (ATD). Recent crystal structures of GluN1-GluN2B NMDA receptors bound to agonists and an allosteric inhibitor, ifenprodil, represent the allosterically inhibited state. However, how the ATD and LBD move to activate the NMDA receptor ion channel remains unclear. Here we applied X-ray crystallography, single-particle electron cryomicroscopy and electrophysiology to rat NMDA receptors to show that, in the absence of ifenprodil, the bi-lobed structure of GluN2 ATD adopts an open conformation accompanied by rearrangement of the GluN1-GluN2 ATD heterodimeric interface, altering subunit orientation in the ATD and LBD and forming an active receptor conformation that gates the ion channel.


Asunto(s)
Piperidinas/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/química , Animales , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Microscopía por Crioelectrón , Cristalografía por Rayos X , Electrofisiología , Activación del Canal Iónico/efectos de los fármacos , Ligandos , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Ratas , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/ultraestructura
2.
J Biol Chem ; 288(1): 274-84, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23184945

RESUMEN

Filopodia are cell surface protrusions that are essential for cell migration. This finger-like structure is supported by rigid tightly bundled actin filaments. The protein responsible for actin bundling in filopodia is fascin. However, the mechanism by which fascin functions in filopodial formation is not clear. Here we provide biochemical, cryo-electron tomographic, and x-ray crystal structural data demonstrating the unique structural characteristics of fascin. Systematic mutagenesis studies on 100 mutants of fascin indicate that there are two major actin-binding sites on fascin. Crystal structures of four fascin mutants reveal concerted conformational changes in fascin from inactive to active states in the process of actin bundling. Mutations in any one of the actin-binding sites impair the cellular function of fascin in filopodial formation. Altogether, our data reveal the molecular mechanism of fascin function in filopodial formation.


Asunto(s)
Proteínas Portadoras/química , Regulación Neoplásica de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Microfilamentos/química , Seudópodos/metabolismo , Actinas/química , Actinas/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Microscopía por Crioelectrón/métodos , Cristalografía por Rayos X/métodos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas de Microfilamentos/metabolismo , Microscopía Fluorescente/métodos , Modelos Moleculares , Conformación Molecular , Metástasis de la Neoplasia , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/química , Transducción de Señal
3.
Science ; 384(6703): eadm8693, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38935733

RESUMEN

Measles virus (MeV) presents a public health threat that is escalating as vaccine coverage in the general population declines and as populations of immunocompromised individuals, who cannot be vaccinated, increase. There are no approved therapeutics for MeV. Neutralizing antibodies targeting viral fusion are one potential therapeutic approach but have not yet been structurally characterized or advanced to clinical use. We present cryo-electron microscopy (cryo-EM) structures of prefusion F alone [2.1-angstrom (Å) resolution], F complexed with a fusion-inhibitory peptide (2.3-Å resolution), F complexed with the neutralizing and protective monoclonal antibody (mAb) 77 (2.6-Å resolution), and an additional structure of postfusion F (2.7-Å resolution). In vitro assays and examination of additional EM classes show that mAb 77 binds prefusion F, arrests F in an intermediate state, and prevents transition to the postfusion conformation. These structures shed light on antibody-mediated neutralization that involves arrest of fusion proteins in an intermediate state.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Microscopía por Crioelectrón , Virus del Sarampión , Proteínas Virales de Fusión , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/química , Virus del Sarampión/inmunología , Virus del Sarampión/química , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/química , Humanos , Conformación Proteica
4.
Cell Chem Biol ; 30(4): 403-411.e4, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-36990092

RESUMEN

The mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a globally distributed zoonotic pathogen that can be lethal in immunocompromised patients and can cause severe birth defects if acquired during pregnancy. The structure of the trimeric surface glycoprotein, essential for entry, vaccine design, and antibody neutralization, remains unknown. Here, we present the cryoelectron microscopy (cryo-EM) structure of the LCMV surface glycoprotein (GP) in its trimeric pre-fusion assembly both alone and in complex with a rationally engineered monoclonal neutralizing antibody termed 18.5C-M28 (M28). Additionally, we show that passive administration of M28, either as a prophylactic or therapeutic, protects mice from LCMV clone 13 (LCMVcl13) challenge. Our study illuminates not only the overall structural organization of LCMV GP and the mechanism for its inhibition by M28 but also presents a promising therapeutic candidate to prevent severe or fatal disease in individuals who are at risk of infection by a virus that poses a threat worldwide.


Asunto(s)
Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica , Ratones , Animales , Coriomeningitis Linfocítica/prevención & control , Microscopía por Crioelectrón , Glicoproteínas de Membrana
5.
bioRxiv ; 2023 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-37034637

RESUMEN

Donor-specific antibody (DSA) responses against human leukocyte antigen (HLA) proteins mismatched between kidney transplant donors and recipients cause allograft loss. Using single-cell, molecular, structural, and proteomic techniques, we profiled the HLA-specific (alloreactive) B cell response in kidney and blood of a transplant recipient with antibody-mediated rejection (AMR). We identified 14 distinct alloreactive B cell lineages, which spanned the rejected organ and blood and expressed high-affinity anti-donor HLA-specific B cell receptors, many of which were clonally linked to circulating DSA. The alloreactive B cell response was focused on exposed, solvent-accessible mismatched HLA residues, while also demonstrating extensive contacts with self-HLA residues. Consistent with structural evidence of self-recognition, measurable self-reactivity by donor-specific B cells was common and positively correlated with anti-donor affinity maturation. Thus, allo- and self-reactive signatures appeared to converge, suggesting that during AMR, the recognition of non-self and breaches of tolerance conspire to produce a pathogenic donor-specific adaptive response.

6.
Sci Transl Med ; 14(668): eabq0991, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36288283

RESUMEN

Developing potent therapeutics and effective vaccines are the ultimate goals in controlling infectious diseases. Lassa virus (LASV), the causative pathogen of Lassa fever (LF), infects hundreds of thousands annually, but effective antivirals or vaccines against LASV infection are still lacking. Furthermore, neutralizing antibodies against LASV are rare. Here, we describe biochemical analyses and high-resolution cryo-electron microscopy structures of a therapeutic cocktail of three broadly protective antibodies that target the LASV glycoprotein complex (GPC), previously identified from survivors of multiple LASV infections. Structural and mechanistic analyses reveal compatible neutralizing epitopes and complementary neutralization mechanisms that offer high potency, broad range, and resistance to escape. These antibodies either circumvent or exploit specific glycans comprising the extensive glycan shield of GPC. Further, they require mammalian glycosylation, native GPC cleavage, and proper GPC trimerization. These findings guided engineering of a next-generation GPC antigen suitable for future neutralizing antibody and vaccine discovery. Together, these results explain protective mechanisms of rare, broad, and potent antibodies and identify a strategy for the rational design of therapeutic modalities against LF and related infectious diseases.


Asunto(s)
Fiebre de Lassa , Vacunas Virales , Animales , Humanos , Virus Lassa , Microscopía por Crioelectrón , Anticuerpos Neutralizantes , Epítopos , Glicoproteínas , Polisacáridos , Antivirales , Mamíferos
7.
Science ; 374(6566): 472-478, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34554826

RESUMEN

Antibody-based therapeutics and vaccines are essential to combat COVID-19 morbidity and mortality after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple mutations in SARS-CoV-2 that could impair antibody defenses propagated in human-to-human transmission and spillover or spillback events between humans and animals. To develop prevention and therapeutic strategies, we formed an international consortium to map the epitope landscape on the SARS-CoV-2 spike protein, defining and structurally illustrating seven receptor binding domain (RBD)­directed antibody communities with distinct footprints and competition profiles. Pseudovirion-based neutralization assays reveal spike mutations, individually and clustered together in variants, that affect antibody function among the communities. Key classes of RBD-targeted antibodies maintain neutralization activity against these emerging SARS-CoV-2 variants. These results provide a framework for selecting antibody treatment cocktails and understanding how viral variants might affect antibody therapeutic efficacy.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Mapeo Epitopo , Epítopos Inmunodominantes/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Antígenos Virales/química , Antígenos Virales/inmunología , COVID-19/terapia , Humanos , Epítopos Inmunodominantes/química , Unión Proteica , Dominios Proteicos , Glicoproteína de la Espiga del Coronavirus/química
8.
Nature ; 428(6980): 319-23, 2004 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15029195

RESUMEN

Key questions regarding the molecular nature of prions are how different prion strains can be propagated by the same protein and whether they are only protein. Here we demonstrate the protein-only nature of prion strains in a yeast model, the [PSI] genetic element that enhances the read-through of nonsense mutations in the yeast Saccharomyces cerevisiae. Infectious fibrous aggregates containing a Sup35 prion-determining amino-terminal fragment labelled with green fluorescent protein were purified from yeast harbouring distinctive prion strains. Using the infectious aggregates as 'seeds', elongated fibres were generated in vitro from the bacterially expressed labelled prion protein. De novo generation of strain-specific [PSI] infectivity was demonstrated by introducing sheared fibres into uninfected yeast hosts. The cross-sectional morphology of the elongated fibres generated in vitro was indistinguishable from that of the short yeast seeds, as visualized by electron microscopy. Electron diffraction of the long fibres showed the 4.7 A spacing characteristic of the cross-beta structure of amyloids. The fact that the amyloid fibres nucleated in vitro propagate the strain-specific infectivity of the yeast seeds implies that the heritable information of distinct prion strains must be encoded by different, self-propagating cross-beta folding patterns of the same prion protein.


Asunto(s)
Priones/clasificación , Priones/metabolismo , Proteínas de Saccharomyces cerevisiae/clasificación , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Amiloide/química , Amiloide/genética , Amiloide/metabolismo , Amiloide/ultraestructura , Fusión Celular , Escherichia coli , Modelos Biológicos , Factores de Terminación de Péptidos , Priones/genética , Priones/ultraestructura , Conformación Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/ultraestructura , Sonicación , Transformación Genética
9.
Cell Rep ; 30(1): 53-60.e5, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31914397

RESUMEN

The large (L) proteins of non-segmented, negative-strand RNA viruses are multifunctional enzymes that produce capped, methylated, and polyadenylated mRNA and replicate the viral genome. A phosphoprotein (P), required for efficient RNA-dependent RNA polymerization from the viral ribonucleoprotein (RNP) template, regulates the function and conformation of the L protein. We report the structure of vesicular stomatitis virus L in complex with its P cofactor determined by electron cryomicroscopy at 3.0 Å resolution, enabling us to visualize bound segments of P. The contacts of three P segments with multiple L domains show how P induces a closed, compact, initiation-competent conformation. Binding of P to L positions its N-terminal domain adjacent to a putative RNA exit channel for efficient encapsidation of newly synthesized genomes with the nucleoprotein and orients its C-terminal domain to interact with an RNP template. The model shows that a conserved tryptophan in the priming loop can support the initiating 5' nucleotide.


Asunto(s)
Coenzimas/metabolismo , Fosfoproteínas/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Humanos , Modelos Moleculares , Fosfoproteínas/química , Fosfoproteínas/ultraestructura , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , ARN Polimerasa Dependiente del ARN/ultraestructura , Proteínas Virales/ultraestructura
10.
Brain Res ; 1130(1): 223-34, 2007 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-17157826

RESUMEN

The discovery of small molecule inhibitors of cytotoxicity induced by amyloid-beta (Abeta) oligomers, either applied extracellularly or accumulated intraneuronally, is an important goal of drug development for Alzheimer's disease (AD), but has been limited by the lack of efficient screening methods. Here we describe our approach using two cell-based methods. The first method takes advantage of the unique ability of extracellularly applied Abeta oligomers to rapidly induce the exocytosis of formazan formed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). We employed a short protocol to quantify this toxicity, and quickly identified two novel inhibitors, code-named CP2 and A5, from two compound libraries. A second independent screen of the same libraries using our previously published MC65 protection assay, which identifies inhibitors of toxicity related to intracellular Abeta oligomers, also selected the same two leads, suggesting that both assays select for the same anti-Abeta oligomer properties displayed by these compounds. We further demonstrated that A5 attenuated the progressive aggregation of existing Abeta oligomers, reduced the level of intracellular Abeta oligomers, and prevented the Abeta oligomer-induced death of primary cortical neurons, effects similar to those demonstrated by CP2. Our results suggest that, when combined, the two methods would generate fewer false results and give a high likelihood of identifying leads that show promises in ameliorating Abeta oligomer-induced toxicities within both intraneuronal and extracellular sites. Both assays are simple, suitable for rapid screening of a large number of medicinal libraries, and amenable for automation.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Carbazoles/farmacología , Evaluación Preclínica de Medicamentos/métodos , Formazáns/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Sales de Tetrazolio/metabolismo , Péptidos beta-Amiloides/administración & dosificación , Animales , Bioensayo/métodos , Línea Celular , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Humanos , Ratones , Proteínas del Tejido Nervioso/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología
11.
Elife ; 62017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300532

RESUMEN

ArfA rescues ribosomes stalled on truncated mRNAs by recruiting release factor RF2, which normally binds stop codons to catalyze peptide release. We report two 3.2 Šresolution cryo-EM structures - determined from a single sample - of the 70S ribosome with ArfA•RF2 in the A site. In both states, the ArfA C-terminus occupies the mRNA tunnel downstream of the A site. One state contains a compact inactive RF2 conformation. Ordering of the ArfA N-terminus in the second state rearranges RF2 into an extended conformation that docks the catalytic GGQ motif into the peptidyl-transferase center. Our work thus reveals the structural dynamics of ribosome rescue. The structures demonstrate how ArfA 'senses' the vacant mRNA tunnel and activates RF2 to mediate peptide release without a stop codon, allowing stalled ribosomes to be recycled.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestructura , Factores de Terminación de Péptidos/metabolismo , Factores de Terminación de Péptidos/ultraestructura , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/ultraestructura , Ribosomas/metabolismo , Ribosomas/ultraestructura , Microscopía por Crioelectrón , Unión Proteica
12.
Elife ; 62017 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-29027901

RESUMEN

In bacteria, mRNA transcription and translation are coupled to coordinate optimal gene expression and maintain genome stability. Coupling is thought to involve direct interactions between RNA polymerase (RNAP) and the translational machinery. We present cryo-EM structures of E. coli RNAP core bound to the small ribosomal 30S subunit. The complex is stable under cell-like ionic conditions, consistent with functional interaction between RNAP and the 30S subunit. The RNA exit tunnel of RNAP aligns with the Shine-Dalgarno-binding site of the 30S subunit. Ribosomal protein S1 forms a wall of the tunnel between RNAP and the 30S subunit, consistent with its role in directing mRNAs onto the ribosome. The nucleic-acid-binding cleft of RNAP samples distinct conformations, suggesting different functional states during transcription-translation coupling. The architecture of the 30S•RNAP complex provides a structural basis for co-localization of the transcriptional and translational machineries, and inform future mechanistic studies of coupled transcription and translation.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/enzimología , Subunidades Ribosómicas Pequeñas Bacterianas/química , Subunidades Ribosómicas Pequeñas Bacterianas/metabolismo , Microscopía por Crioelectrón , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo
13.
J Mol Biol ; 330(5): 1165-75, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12860136

RESUMEN

The seven-residue peptide GNNQQNY from the N-terminal region of the yeast prion protein Sup35, which forms amyloid fibers, colloidal aggregates and highly ordered nanocrystals, provides a model system for characterizing the elusively protean cross-beta conformation. Depending on preparative conditions, orthorhombic and monoclinic crystals with similar lath-shaped morphology have been obtained. Ultra high-resolution (<0.5A spacing) electron diffraction patterns from single nanocrystals show that the peptide chains pack in parallel cross-beta columns with approximately 4.86A axial spacing. Mosaic striations 20-50 nm wide observed by electron microscopy indicate lateral size-limiting crystal growth related to amyloid fiber formation. Frequently obtained orthorhombic forms, with apparent space group symmetry P2(1)2(1)2(1), have cell dimensions ranging from /a/=22.7-21.2A, /b/=39.9-39.3A, /c/=4.89-4.86A for wet to dried states. Electron diffraction data from single nanocrystals, recorded in tilt series of still frames, have been mapped in reciprocal space. However, reliable integrated intensities cannot be obtained from these series, and dynamical electron diffraction effects present problems in data analysis. The diversity of ordered structures formed under similar conditions has made it difficult to obtain reproducible X-ray diffraction data from powder specimens; and overlapping Bragg reflections in the powder patterns preclude separated structure factor measurements for these data. Model protofilaments, consisting of tightly paired, half-staggered beta strands related by a screw axis, can be fit in the crystal lattices, but model refinement will require accurate structure factor measurements. Nearly anhydrous packing of this hydrophilic peptide can account for the insolubility of the crystals, since the activation energy for rehydration may be extremely high. Water-excluding packing of paired cross-beta peptide segments in thin protofilaments may be characteristic of the wide variety of anomalously stable amyloid aggregates.


Asunto(s)
Amiloide/química , Péptidos/química , Proteínas de Saccharomyces cerevisiae , Cristalografía por Rayos X , Electrones , Proteínas Fúngicas/química , Microscopía Electrónica , Factores de Terminación de Péptidos , Polimorfismo Genético , Priones/química , Conformación Proteica , Agua/química , Difracción de Rayos X
14.
Prion ; 9(3): 207-27, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26038983

RESUMEN

Prion strains are different self-propagating conformers of the same infectious protein. Three strains of the [PSI] prion, infectious forms of the yeast Sup35 protein, have been previously characterized in our laboratory. Here we report the discovery of a new [PSI] strain, named W8. We demonstrate its robust cellular propagation as well as the protein-only transmission. To reveal strain-specific sequence requirement, mutations that interfered with the propagation of W8 were identified by consecutive substitution of residues 5-55 of Sup35 by proline and insertion of glycine at alternate sites in this segment. Interestingly, propagating W8 with single mutations at residues 5-7 and around residue 43 caused the strain to transmute. In contrast to the assertion that [PSI] existed as a dynamic cloud of sub-structures, no random drift in transmission characteristics was detected in mitotically propagated W8 populations. Electron diffraction and mass-per-length measurements indicate that, similar to the 3 previously characterized strains, W8 fibers are composed of about 1 prion molecule per 4.7-Å cross-ß repeat period. Thus differently folded single Sup35 molecules, not dimeric and trimeric assemblies, form the basic repeating units to build the 4 [PSI] strains.


Asunto(s)
Factores de Terminación de Péptidos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Alelos , Secuencia Conservada , Saccharomyces cerevisiae/genética
15.
Cell Rep ; 3(3): 759-68, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23434508

RESUMEN

To elucidate the structural basis of the mechanism of microtubule depolymerization by kinesin-13s, we analyzed complexes of tubulin and the Drosophila melanogaster kinesin-13 KLP10A by electron microscopy (EM) and fluorescence polarization microscopy. We report a nanometer-resolution (1.1 nm) cryo-EM three-dimensional structure of the KLP10A head domain (KLP10AHD) bound to curved tubulin. We found that binding of KLP10AHD induces a distinct tubulin configuration with displacement (shear) between tubulin subunits in addition to curvature. In this configuration, the kinesin-binding site differs from that in straight tubulin, providing an explanation for the distinct interaction modes of kinesin-13s with the microtubule lattice or its ends. The KLP10AHD-tubulin interface comprises three areas of interaction, suggesting a crossbow-type tubulin-bending mechanism. These areas include the kinesin-13 family conserved KVD residues, and as predicted from the crossbow model, mutating these residues changes the orientation and mobility of KLP10AHDs interacting with the microtubule.


Asunto(s)
Proteínas de Drosophila/química , Drosophila melanogaster/metabolismo , Cinesinas/química , Microtúbulos/metabolismo , Simulación de Dinámica Molecular , Tubulina (Proteína)/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/ultraestructura , Drosophila melanogaster/química , Cinesinas/metabolismo , Cinesinas/ultraestructura , Microtúbulos/ultraestructura , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Subunidades de Proteína/metabolismo , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/ultraestructura
16.
J Cell Biol ; 196(4): 419-34, 2012 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-22331846

RESUMEN

The nuclear pore complex (NPC) is a multiprotein assembly that serves as the sole mediator of nucleocytoplasmic exchange in eukaryotic cells. In this paper, we use an integrative approach to determine the structure of an essential component of the yeast NPC, the ~600-kD heptameric Nup84 complex, to a precision of ~1.5 nm. The configuration of the subunit structures was determined by satisfaction of spatial restraints derived from a diverse set of negative-stain electron microscopy and protein domain-mapping data. Phenotypic data were mapped onto the complex, allowing us to identify regions that stabilize the NPC's interaction with the nuclear envelope membrane and connect the complex to the rest of the NPC. Our data allow us to suggest how the Nup84 complex is assembled into the NPC and propose a scenario for the evolution of the Nup84 complex through a series of gene duplication and loss events. This work demonstrates that integrative approaches based on low-resolution data of sufficient quality can generate functionally informative structures at intermediate resolution.


Asunto(s)
Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/fisiología , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Modelos Moleculares , Poro Nuclear/ultraestructura , Proteínas de Complejo Poro Nuclear/genética , Conformación Proteica , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Eliminación de Secuencia , Relación Estructura-Actividad
17.
Virology ; 414(2): 103-9, 2011 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-21489589

RESUMEN

Cryo-electron tomography and subtomogram averaging are utilized to determine that the bacteriophage ϕ12, a member of the Cystoviridae family, contains surface complexes that are toroidal in shape, are composed of six globular domains with six-fold symmetry, and have a discrete density connecting them to the virus membrane-envelope surface. The lack of this kind of spike in a reassortant of ϕ12 demonstrates that the gene for the hexameric spike is located in ϕ12's medium length genome segment, likely to the P3 open reading frames which are the proteins involved in viral-host cell attachment. Based on this and on protein mass estimates derived from the obtained averaged structure, it is suggested that each of the globular domains is most likely composed of a total of four copies of P3a and/or P3c proteins. Our findings may have implications in the study of the evolution of the cystovirus species in regard to their host specificity.


Asunto(s)
Bacteriófagos/fisiología , Especificidad del Huésped , Pseudomonas syringae/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Acoplamiento Viral , Secuencia de Aminoácidos , Bacteriófagos/química , Bacteriófagos/genética , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Estructura Terciaria de Proteína , Proteínas del Envoltorio Viral/genética
18.
Nat Struct Mol Biol ; 18(6): 658-64, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21552261

RESUMEN

Trax-translin heteromers, also known as C3PO, have been proposed to activate the RNA-induced silencing complex (RISC) by facilitating endonucleolytic cleavage of the siRNA passenger strand. We report on the crystal structure of hexameric Drosophila C3PO formed by truncated translin and Trax, along with electron microscopic and mass spectrometric studies on octameric C3PO formed by full-length translin and Trax. Our studies establish that Trax adopts the translin fold, possesses catalytic centers essential for C3PO's endoRNase activity and interacts extensively with translin to form an octameric assembly. The catalytic pockets of Trax subunits are located within the interior chamber of the octameric scaffold. Truncated C3PO, like full-length C3PO, shows endoRNase activity that leaves 3'-hydroxyl-cleaved ends. We have measured the catalytic activity of C3PO and shown it to cleave almost stoichiometric amounts of substrate per second.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Endorribonucleasas/química , Endorribonucleasas/metabolismo , Animales , Dominio Catalítico , Cristalografía por Rayos X , Proteínas de Unión al ADN , Drosophila melanogaster/química , Cinética , Espectrometría de Masas , Microscopía Electrónica , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , ARN/metabolismo
19.
Virology ; 372(2): 273-90, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18061231

RESUMEN

We previously reported that an envelope (Env) glycoprotein immunogen (o-gp140DeltaV2SF162) containing a partial deletion in the second variable loop (V2) derived from the R5-tropic HIV-1 isolate SF162 partially protected vaccinated rhesus macaques against pathogenic SHIV(SF162P4) virus. Extending our studies to subtype C isolate TV1, we have purified o-gp140DeltaV2TV1 (subtype C DeltaV2 trimer) to homogeneity, performed glycosylation analysis, and determined its ability to bind CD4, as well as a panel of well-characterized neutralizing monoclonal antibodies (mAb). In general, critical epitopes are preserved on the subtype C DeltaV2 trimer; however, we did not observe significant binding for the b12 mAb. The molecular mass of subtype C DeltaV2 trimer was found to be 450 kDa, and the hydrodynamic radius was found to be 10.87 nm. Our data suggest that subtype C DeltaV2 trimer binds to CD4 with an affinity comparable to o-gp140DeltaV2SF162 (subtype B DeltaV2 trimer). Using isothermal titration calorimetric (ITC) analysis, we demonstrated that all three CD4 binding sites (CD4-BS) in both subtype C and B trimers are exposed and accessible. However, compared to subtype B trimer, the three CD4-BS in subtype C trimer have different affinities for CD4, suggesting a cooperativity of CD4 binding in subtype C trimer but not in subtype B trimer. Negative staining electron microscopy of the subtype C DeltaV2 trimer has demonstrated that it is in fact a trimer. These results highlight the importance of studying subtype C Env, and also of developing appropriate subtype C-specific reagents that may be used for better immunological characterization of subtype C Env for developing an AIDS vaccine.


Asunto(s)
Productos del Gen env/metabolismo , VIH-1/clasificación , VIH-1/metabolismo , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales , Antígenos CD4/metabolismo , Células CHO , Línea Celular , Cricetinae , Cricetulus , Epítopos , Regulación Viral de la Expresión Génica , Productos del Gen env/química , Productos del Gen env/genética , Variación Genética , Antígenos VIH , Humanos , Unión Proteica
20.
Proc Natl Acad Sci U S A ; 102(29): 10165-70, 2005 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-16006506

RESUMEN

Mass per length (mpl) measurements on single amyloid fibrils that specifically propagate the [VH], [VK], and [VL] strains of the yeast prion [PSI] reveal unanticipated differences in their structures. Many fibrils have approximately 1.0 prion molecule per 4.7-A cross-beta repeat period, which is consistent with a self-replicating model built by parallel beta-sheet hydrogen-bonding of like prion peptide segments, but other fibrils are definitely heavier. The predominantly straight fibrils of the dominant [VH] strain have a bimodal mpl distribution, corresponding to components with approximately 1.0 and 1.2 prions per repeat. Fibrils of the weaker [VK] strain, which are almost all wavy, have a monodisperse mpl distribution with a mean of 1.15 prions per repeat. The recessive [VL] strain sample has approximately 1.05 prions per repeat in single fibrils and includes approximately 10% double fibrils, which are rare in the duplicate [VH] and [VK] samples. All of these samples were assembled from purified recombinant Sup35 prion protein by seeded growth on nuclei extracted from yeast bearing the three [PSI] strains. Infectious and noninfectious spontaneously assembled fibrils of the recombinant prion protein also display different heterogeneous morphologies. The strain-specific morphological differences we have observed directly confirm the structural prediction of the protein-only prion theory but do not have an obvious molecular explanation.


Asunto(s)
Amiloide/ultraestructura , Priones/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Amiloide/metabolismo , Proteínas Fluorescentes Verdes , Microscopía Electrónica , Factores de Terminación de Péptidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA