Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 178(2): 290-301.e10, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31230712

RESUMEN

How the central innate immune protein, STING, is activated by its ligands remains unknown. Here, using structural biology and biochemistry, we report that the metazoan second messenger 2'3'-cGAMP induces closing of the human STING homodimer and release of the STING C-terminal tail, which exposes a polymerization interface on the STING dimer and leads to the formation of disulfide-linked polymers via cysteine residue 148. Disease-causing hyperactive STING mutations either flank C148 and depend on disulfide formation or reside in the C-terminal tail binding site and cause constitutive C-terminal tail release and polymerization. Finally, bacterial cyclic-di-GMP induces an alternative active STING conformation, activates STING in a cooperative manner, and acts as a partial antagonist of 2'3'-cGAMP signaling. Our insights explain the tight control of STING signaling given varying background activation signals and provide a therapeutic hypothesis for autoimmune syndrome treatment.


Asunto(s)
Proteínas de la Membrana/metabolismo , Sitios de Unión , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Dimerización , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Ligandos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Nucleótidos Cíclicos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Transducción de Señal
2.
Proc Natl Acad Sci U S A ; 121(6): e2300644120, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38306481

RESUMEN

It is unclear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to the strong but ineffective inflammatory response that characterizes severe Coronavirus disease 2019 (COVID-19), with amplified immune activation in diverse cell types, including cells without angiotensin-converting enzyme 2 receptors necessary for infection. Proteolytic degradation of SARS-CoV-2 virions is a milestone in host viral clearance, but the impact of remnant viral peptide fragments from high viral loads is not known. Here, we examine the inflammatory capacity of fragmented viral components from the perspective of supramolecular self-organization in the infected host environment. Interestingly, a machine learning analysis to SARS-CoV-2 proteome reveals sequence motifs that mimic host antimicrobial peptides (xenoAMPs), especially highly cationic human cathelicidin LL-37 capable of augmenting inflammation. Such xenoAMPs are strongly enriched in SARS-CoV-2 relative to low-pathogenicity coronaviruses. Moreover, xenoAMPs from SARS-CoV-2 but not low-pathogenicity homologs assemble double-stranded RNA (dsRNA) into nanocrystalline complexes with lattice constants commensurate with the steric size of Toll-like receptor (TLR)-3 and therefore capable of multivalent binding. Such complexes amplify cytokine secretion in diverse uninfected cell types in culture (epithelial cells, endothelial cells, keratinocytes, monocytes, and macrophages), similar to cathelicidin's role in rheumatoid arthritis and lupus. The induced transcriptome matches well with the global gene expression pattern in COVID-19, despite using <0.3% of the viral proteome. Delivery of these complexes to uninfected mice boosts plasma interleukin-6 and CXCL1 levels as observed in COVID-19 patients.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Animales , Ratones , Células Endoteliales , Proteoma , Péptidos
3.
Biophys J ; 122(12): 2456-2474, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37147801

RESUMEN

The cell-cell adhesion cadherin-catenin complexes recruit vinculin to the adherens junction (AJ) to modulate the mechanical couplings between neighboring cells. However, it is unclear how vinculin influences the AJ structure and function. Here, we identified two patches of salt bridges that lock vinculin in the head-tail autoinhibited conformation and reconstituted the full-length vinculin activation mimetics bound to the cadherin-catenin complex. The cadherin-catenin-vinculin complex contains multiple disordered linkers and is highly dynamic, which poses a challenge for structural studies. We determined the ensemble conformation of this complex using small-angle x-ray and selective deuteration/contrast variation small-angle neutron scattering. In the complex, both α-catenin and vinculin adopt an ensemble of flexible conformations, but vinculin has fully open conformations with the vinculin head and actin-binding tail domains well separated from each other. F-actin binding experiments show that the cadherin-catenin-vinculin complex binds and bundles F-actin. However, when the vinculin actin-binding domain is removed from the complex, only a minor fraction of the complex binds to F-actin. The results show that the dynamic cadherin-catenin-vinculin complex employs vinculin as the primary F-actin binding mode to strengthen AJ-cytoskeleton interactions.


Asunto(s)
Actinas , Cadherinas , Cadherinas/metabolismo , Actinas/metabolismo , Vinculina/metabolismo , alfa Catenina/química , Unión Proteica , Citoesqueleto de Actina/metabolismo , Adhesión Celular
4.
Proc Natl Acad Sci U S A ; 117(1): 388-394, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31848245

RESUMEN

Surface layers (S-layers) are crystalline protein coats surrounding microbial cells. S-layer proteins (SLPs) regulate their extracellular self-assembly by crystallizing when exposed to an environmental trigger. However, molecular mechanisms governing rapid protein crystallization in vivo or in vitro are largely unknown. Here, we demonstrate that the Caulobacter crescentus SLP readily crystallizes into sheets in vitro via a calcium-triggered multistep assembly pathway. This pathway involves 2 domains serving distinct functions in assembly. The C-terminal crystallization domain forms the physiological 2-dimensional (2D) crystal lattice, but full-length protein crystallizes multiple orders of magnitude faster due to the N-terminal nucleation domain. Observing crystallization using a time course of electron cryo-microscopy (Cryo-EM) imaging reveals a crystalline intermediate wherein N-terminal nucleation domains exhibit motional dynamics with respect to rigid lattice-forming crystallization domains. Dynamic flexibility between the 2 domains rationalizes efficient S-layer crystal nucleation on the curved cellular surface. Rate enhancement of protein crystallization by a discrete nucleation domain may enable engineering of kinetically controllable self-assembling 2D macromolecular nanomaterials.


Asunto(s)
Proteínas Bacterianas/metabolismo , Caulobacter crescentus/metabolismo , Membrana Celular/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/ultraestructura , Calcio/metabolismo , Caulobacter crescentus/genética , Caulobacter crescentus/ultraestructura , Membrana Celular/química , Membrana Celular/ultraestructura , Microscopía por Crioelectrón , Cristalización , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/ultraestructura , Mutagénesis
5.
Proc Natl Acad Sci U S A ; 116(43): 21545-21555, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31591245

RESUMEN

The cadherin-catenin adhesion complex is the central component of the cell-cell adhesion adherens junctions that transmit mechanical stress from cell to cell. We have determined the nanoscale structure of the adherens junction complex formed by the α-catenin•ß-catenin•epithelial cadherin cytoplasmic domain (ABE) using negative stain electron microscopy, small-angle X-ray scattering, and selective deuteration/small-angle neutron scattering. The ABE complex is highly pliable and displays a wide spectrum of flexible structures that are facilitated by protein-domain motions in α- and ß-catenin. Moreover, the 107-residue intrinsically disordered N-terminal segment of ß-catenin forms a flexible "tongue" that is inserted into α-catenin and participates in the assembly of the ABE complex. The unanticipated ensemble of flexible conformations of the ABE complex suggests a dynamic mechanism for sensitivity and reversibility when transducing mechanical signals, in addition to the catch/slip bond behavior displayed by the ABE complex under mechanical tension. Our results provide mechanistic insight into the structural dynamics for the cadherin-catenin adhesion complex in mechanotransduction.


Asunto(s)
Cadherinas/química , Cadherinas/metabolismo , Mecanotransducción Celular , alfa Catenina/química , alfa Catenina/metabolismo , beta Catenina/química , beta Catenina/metabolismo , Uniones Adherentes/química , Uniones Adherentes/genética , Uniones Adherentes/metabolismo , Secuencias de Aminoácidos , Cadherinas/genética , Humanos , Conformación Molecular , Unión Proteica , Dominios Proteicos , Dispersión del Ángulo Pequeño , alfa Catenina/genética , beta Catenina/genética
6.
Proc Natl Acad Sci U S A ; 115(37): 9098-9103, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150407

RESUMEN

Ladderane lipids are unique to anaerobic ammonium-oxidizing (anammox) bacteria and are enriched in the membrane of the anammoxosome, an organelle thought to compartmentalize the anammox process, which involves the toxic intermediate hydrazine (N2H4). Due to the slow growth rate of anammox bacteria and difficulty of isolating pure ladderane lipids, experimental evidence of the biological function of ladderanes is lacking. We have synthesized two natural and one unnatural ladderane phosphatidylcholine lipids and compared their thermotropic properties in self-assembled bilayers to distinguish between [3]- and [5]-ladderane function. We developed a hydrazine transmembrane diffusion assay using a water-soluble derivative of a hydrazine sensor and determined that ladderane membranes are as permeable to hydrazine as straight-chain lipid bilayers. However, pH equilibration across ladderane membranes occurs 5-10 times more slowly than across straight-chain lipid membranes. Langmuir monolayer analysis and the rates of fluorescence recovery after photobleaching suggest that dense ladderane packing may preclude formation of proton/hydroxide-conducting water wires. These data support the hypothesis that ladderanes prevent the breakdown of the proton motive force rather than blocking hydrazine transmembrane diffusion in anammox bacteria.


Asunto(s)
Bacterias/química , Permeabilidad de la Membrana Celular , Membrana Celular/química , Hidrazinas/química , Hidróxidos/química , Fosfolípidos/química , Protones , Anaerobiosis/fisiología , Bacterias/metabolismo , Membrana Celular/metabolismo , Hidrazinas/metabolismo , Hidróxidos/metabolismo , Fosfolípidos/metabolismo
7.
Opt Express ; 28(8): 10939-10950, 2020 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-32403615

RESUMEN

X-ray free electron lasers (XFELs) provide femtosecond high-power x-ray beams with high spatial coherence, resulting in numerous influential discoveries. Diffractive optics allow for the easy manipulation and measurement of an x-ray beam's wavefront and enable the realization of complex designed properties and specifications. For example, phase gratings can be used as x-ray beam splitters to enable beam sharing by multiple end stations or in-situ beam monitoring, including spectrum and wavefront measurements. Wavefront preservation and high efficiency and survivability under high power are requirements for such beam splitters. Diamond is the most suitable choice for phase grating fabrication, due to its high thermal conductivity that enables it to survive high average power XFEL beams. We have fabricated a large area (2×2 mm2) high aspect ratio (13:1) diamond grating on a diamond plate. Testing was performed at 9.5 keV and resulted in a high splitting efficiency (30%). Tunable efficiency was obtained via tilting the grating with respect to the x-ray beam. Wavefront fidelity of the split beams were measured to less than λ/100 using a Talbot wavefront sensor.

8.
Biophys J ; 115(4): 642-654, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30037495

RESUMEN

As a core component of the adherens junction, α-catenin stabilizes the cadherin/catenin complexes to the actin cytoskeleton for the mechanical coupling of cell-cell adhesion. α-catenin also modulates actin dynamics, cell polarity, and cell-migration functions that are independent of the adherens junction. We have determined the solution structures of the α-catenin monomer and dimer using in-line size-exclusion chromatography small-angle X-ray scattering, as well as the structure of α-catenin dimer in complex to F-actin filament using selective deuteration and contrast-matching small angle neutron scattering. We further present the first observation, to our knowledge, of the nanoscale dynamics of α-catenin by neutron spin-echo spectroscopy, which explicitly reveals the mobile regions of α-catenin that are crucial for binding to F-actin. In solution, the α-catenin monomer is more expanded than either protomer shown in the crystal structure dimer, with the vinculin-binding M fragment and the actin-binding domain being able to adopt different configurations. The α-catenin dimer in solution is also significantly more expanded than the dimer crystal structure, with fewer interdomain and intersubunit contacts than the crystal structure. When in complex to F-actin, the α-catenin dimer has an even more open and extended conformation than in solution, with the actin-binding domain further separated from the main body of the dimer. The α-catenin-assembled F-actin bundle develops into an ordered filament packing arrangement at increasing α-catenin/F-actin molar ratios. Together, the structural and dynamic studies reveal that α-catenin possesses dynamic molecular conformations that prime this protein to function as a mechanosensor protein.


Asunto(s)
Actinas/metabolismo , Nanotecnología , alfa Catenina/química , alfa Catenina/metabolismo , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Soluciones
9.
Langmuir ; 34(22): 6601-6611, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29715426

RESUMEN

To determine how different components affect the structure of pulmonary surfactant, we measured X-ray scattering by samples derived from calf surfactant. The surfactant phospholipids demonstrated the essential characteristics of the Lγ phase: a unit cell with a lattice constant appropriate for two bilayers, and crystalline chains detected by wide-angle X-ray scattering (WAXS). The electron density profile, obtained from scattering by oriented films at different relative humidities (70-97%), showed that the two bilayers, arranged as mirror images, each contain two distinct leaflets with different thicknesses and profiles. The detailed structures suggest one ordered leaflet that would contain crystalline chains and one disordered monolayer likely to contain the anionic compounds, which constitute ∼10% of the surfactant phospholipids. The spacing and temperature dependence detected by WAXS fit with an ordered leaflet composed of dipalmitoyl phosphatidylcholine. Physiological levels of cholesterol had no effect on this structure. Removing the anionic phospholipids prevented formation of the Lγ phase. The cationic surfactant proteins inhibited Lγ structures, but at levels unlikely related to charge. Because the Lγ phase, if arranged properly, could produce a self-assembled ordered interfacial monolayer, the structure could have important functional consequences. Physiological levels of the proteins, however, inhibit formation of the Lγ structures at high relative humidities, making their physiological significance uncertain.


Asunto(s)
Fosfoproteínas/química , Surfactantes Pulmonares/química , Animales , Bovinos , Fosfolípidos/química , Espectrometría por Rayos X
10.
Biophys J ; 112(9): 1841-1851, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28494955

RESUMEN

Surface layers (S-layers) are paracrystalline, proteinaceous structures found in most archaea and many bacteria. Often the outermost cell envelope component, S-layers serve diverse functions including aiding pathogenicity and protecting against predators. We report that the S-layer of Caulobacter crescentus exhibits calcium-mediated structural plasticity, switching irreversibly between an amorphous aggregate state and the crystalline state. This finding invalidates the common assumption that S-layers serve only as static wall-like structures. In vitro, the Caulobacter S-layer protein, RsaA, enters the aggregate state at physiological temperatures and low divalent calcium ion concentrations. At higher concentrations, calcium ions stabilize monomeric RsaA, which can then transition to the two-dimensional crystalline state. Caulobacter requires micromolar concentrations of calcium for normal growth and development. Without an S-layer, Caulobacter is even more sensitive to changes in environmental calcium concentration. Therefore, this structurally dynamic S-layer responds to environmental conditions as an ion sensor and protects Caulobacter from calcium deficiency stress, a unique mechanism of bacterial adaptation. These findings provide a biochemical and physiological basis for RsaA's calcium-binding behavior, which extends far beyond calcium's commonly accepted role in aiding S-layer biogenesis or oligomerization and demonstrates a connection to cellular fitness.


Asunto(s)
Calcio/metabolismo , Caulobacter crescentus/química , Caulobacter crescentus/metabolismo , Glicoproteínas de Membrana/química , Calcio/química , Caulobacter crescentus/ultraestructura , Dicroismo Circular , Cristalización , Glicoproteínas de Membrana/metabolismo , Microscopía Electrónica de Transmisión , Agregado de Proteínas , Pliegue de Proteína , Dispersión del Ángulo Pequeño , Estrés Fisiológico , Temperatura , Difracción de Rayos X
11.
Adv Exp Med Biol ; 1009: 1-10, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29218550

RESUMEN

Small angle scattering (SAS) is a powerful and versatile tool to elucidate the structure of matter at the nanometer scale. Recently, the technique has seen a tremendous growth of applications in the field of structural molecular biology. Its origins however date back to almost a century ago and even though the methods potential for studying biological macromolecules was realized already early on, it was only during the last two decades that SAS gradually became a major experimental technique for the structural biologist. This rise in popularity and application was driven by the concurrence of different key factors such as the increased accessibility to high quality SAS instruments enabled by the growing number of synchrotron facilities and neutron sources established around the world, the emerging need of the structural biology community to study large multi-domain complexes and flexible systems that are hard to crystalize, and in particular the development and availability of data analysis software together with the overall access to computational resources powerful enough to run them. Today, SAS is an established and widely used tool for structural studies on bio-macromolecules. Given the potential offered by the next generation X-ray and neutron sources as well as the development of new, innovative approaches to collect and analyze solution scattering data, the application of SAS in the field of structural molecular biology will certainly continue to thrive in the years to come.


Asunto(s)
Biología Molecular/historia , Difracción de Neutrones/historia , Dispersión del Ángulo Pequeño , Difracción de Rayos X/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Biología Molecular/instrumentación , Biología Molecular/métodos , Biología Molecular/tendencias , Difracción de Neutrones/instrumentación , Difracción de Neutrones/métodos , Sincrotrones/historia , Sincrotrones/instrumentación , Difracción de Rayos X/instrumentación , Difracción de Rayos X/métodos
12.
Proc Natl Acad Sci U S A ; 111(18): 6714-9, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753590

RESUMEN

To evade host immune mechanisms, many bacteria secrete immunomodulatory enzymes. Streptococcus pyogenes, one of the most common human pathogens, secretes a large endoglycosidase, EndoS, which removes carbohydrates in a highly specific manner from IgG antibodies. This modification renders antibodies incapable of eliciting host effector functions through either complement or Fc γ receptors, providing the bacteria with a survival advantage. On account of this antibody-specific modifying activity, EndoS is being developed as a promising injectable therapeutic for autoimmune diseases that rely on autoantibodies. Additionally, EndoS is a key enzyme used in the chemoenzymatic synthesis of homogenously glycosylated antibodies with tailored Fc γ receptor-mediated effector functions. Despite the tremendous utility of this enzyme, the molecular basis of EndoS specificity for, and processing of, IgG antibodies has remained poorly understood. Here, we report the X-ray crystal structure of EndoS and provide a model of its encounter complex with its substrate, the IgG1 Fc domain. We show that EndoS is composed of five distinct protein domains, including glycosidase, leucine-rich repeat, hybrid Ig, carbohydrate binding module, and three-helix bundle domains, arranged in a distinctive V-shaped conformation. Our data suggest that the substrate enters the concave interior of the enzyme structure, is held in place by the carbohydrate binding module, and that concerted conformational changes in both enzyme and substrate are required for subsequent antibody deglycosylation. The EndoS structure presented here provides a framework from which novel endoglycosidases could be engineered for additional clinical and biotechnological applications.


Asunto(s)
Proteínas Bacterianas/química , Glicósido Hidrolasas/química , Inmunoglobulina G/metabolismo , Streptococcus pyogenes/enzimología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Glicósido Hidrolasas/inmunología , Glicósido Hidrolasas/metabolismo , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/química , Factores Inmunológicos/química , Factores Inmunológicos/metabolismo , Modelos Moleculares , Conformación Proteica , Estructura Terciaria de Proteína , Dispersión del Ángulo Pequeño , Streptococcus pyogenes/patogenicidad , Especificidad por Sustrato , Difracción de Rayos X
13.
Nano Lett ; 16(7): 4019-24, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27322135

RESUMEN

Carbon nanotube porins (CNTPs), small segments of carbon nanotubes capable of forming defined pores in lipid membranes, are important future components for bionanoelectronic devices as they could provide a robust analog of biological membrane channels. In order to control the incorporation of these CNT channels into lipid bilayers, it is important to understand the structure of the CNTPs before and after insertion into the lipid bilayer as well as the impact of such insertion on the bilayer structure. Here we employed a noninvasive in situ probe, small-angle X-ray scattering, to study the integration of CNT porins into dioleoylphosphatidylcholine bilayers. Our results show that CNTPs in solution are stabilized by a monolayer of lipid molecules wrapped around their outer surface. We also demonstrate that insertion of CNTPs into the lipid bilayer results in decreased bilayer thickness with the magnitude of this effect increasing with the concentration of CNTPs.


Asunto(s)
Nanoporos , Nanotubos de Carbono , Membrana Dobles de Lípidos , Dispersión del Ángulo Pequeño , Difracción de Rayos X
14.
J Struct Biol ; 194(3): 272-81, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26968362

RESUMEN

Sliding clamps are opened and loaded onto primer template junctions by clamp loaders, and once loaded on DNA, confer processivity to replicative polymerases. Previously determined crystal structures of eukaryotic and T4 clamp loader-clamp complexes have captured the sliding clamps in either closed or only partially open interface conformations. In these solution structure studies, we have captured for the first time the clamp loader-sliding clamp complex from Escherichia coli using size exclusion chromatography coupled to small angle X-ray scattering (SEC-SAXS). The data suggests the sliding clamp is in an open conformation which is wide enough to permit duplex DNA binding. The data also provides information about spatial arrangement of the sliding clamp with respect to the clamp loader subunits and is compared to complex crystal structures determined from other organisms.


Asunto(s)
ADN Polimerasa III/metabolismo , Replicación del ADN , ADN Polimerasa Dirigida por ADN , Escherichia coli/enzimología , Modelos Moleculares , Adenosina Trifosfato/metabolismo , Sitios de Unión , Cromatografía en Gel , ADN Bacteriano , Escherichia coli/genética , Proteínas de Escherichia coli , Conformación Proteica , Subunidades de Proteína , Dispersión del Ángulo Pequeño , Soluciones , Difracción de Rayos X
15.
Mol Cell Proteomics ; 13(11): 2911-26, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25139911

RESUMEN

The nuclear pore complex (NPC) is the sole passageway for the transport of macromolecules across the nuclear envelope. Nup133, a major component in the essential Y-shaped Nup84 complex, is a large scaffold protein of the NPC's outer ring structure. Here, we describe an integrative modeling approach that produces atomic models for multiple states of Saccharomyces cerevisiae (Sc) Nup133, based on the crystal structures of the sequence segments and their homologs, including the related Vanderwaltozyma polyspora (Vp) Nup133 residues 55 to 502 (VpNup133(55-502)) determined in this study, small angle X-ray scattering profiles for 18 constructs of ScNup133 and one construct of VpNup133, and 23 negative-stain electron microscopy class averages of ScNup133(2-1157). Using our integrative approach, we then computed a multi-state structural model of the full-length ScNup133 and validated it with mutational studies and 45 chemical cross-links determined via mass spectrometry. Finally, the model of ScNup133 allowed us to annotate a potential ArfGAP1 lipid packing sensor (ALPS) motif in Sc and VpNup133 and discuss its potential significance in the context of the whole NPC; we suggest that ALPS motifs are scattered throughout the NPC's scaffold in all eukaryotes and play a major role in the assembly and membrane anchoring of the NPC in the nuclear envelope. Our results are consistent with a common evolutionary origin of Nup133 with membrane coating complexes (the protocoatomer hypothesis); the presence of the ALPS motifs in coatomer-like nucleoporins suggests an ancestral mechanism for membrane recognition present in early membrane coating complexes.


Asunto(s)
Kluyveromyces/enzimología , Proteínas de Complejo Poro Nuclear/química , Poro Nuclear/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Sitios de Unión/genética , Cristalografía por Rayos X , Evolución Molecular , Modelos Moleculares , Mutación , Membrana Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/ultraestructura , Unión Proteica/genética , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/ultraestructura , Homología de Secuencia de Aminoácido
16.
Proc Natl Acad Sci U S A ; 110(31): 12637-42, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23858467

RESUMEN

In neurons, soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins drive the fusion of synaptic vesicles to the plasma membrane through the formation of a four-helix SNARE complex. Members of the Sec1/Munc18 protein family regulate membrane fusion through interactions with the syntaxin family of SNARE proteins. The neuronal protein Munc18a interacts with a closed conformation of the SNARE protein syntaxin1a (Syx1a) and with an assembled SNARE complex containing Syx1a in an open conformation. The N-peptide of Syx1a (amino acids 1-24) has been implicated in the transition of Munc18a-bound Syx1a to Munc18a-bound SNARE complex, but the underlying mechanism is not understood. Here we report the X-ray crystal structures of Munc18a bound to Syx1a with and without its native N-peptide (Syx1aΔN), along with small-angle X-ray scattering (SAXS) data for Munc18a bound to Syx1a, Syx1aΔN, and Syx1a L165A/E166A (LE), a mutation thought to render Syx1a in a constitutively open conformation. We show that all three complexes adopt the same global structure, in which Munc18a binds a closed conformation of Syx1a. We also identify a possible structural connection between the Syx1a N-peptide and SNARE domain that might be important for the transition of closed-to-open Syx1a in SNARE complex assembly. Although the role of the N-peptide in Munc18a-mediated SNARE complex assembly remains unclear, our results demonstrate that the N-peptide and LE mutation have no effect on the global conformation of the Munc18a-Syx1a complex.


Asunto(s)
Sustitución de Aminoácidos , Proteínas Munc18/química , Péptidos , Eliminación de Secuencia , Sintaxina 1/química , Humanos , Proteínas Munc18/genética , Proteínas Munc18/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Proteínas SNARE/química , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Sintaxina 1/genética , Sintaxina 1/metabolismo
17.
Biophys J ; 109(4): 793-805, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287631

RESUMEN

Aberrant self-assembly, induced by structural misfolding of the prion proteins, leads to a number of neurodegenerative disorders. In particular, misfolding of the mostly α-helical cellular prion protein (PrP(C)) into a ß-sheet-rich disease-causing isoform (PrP(Sc)) is the key molecular event in the formation of PrP(Sc) aggregates. The molecular mechanisms underlying the PrP(C)-to-PrP(Sc) conversion and subsequent aggregation remain to be elucidated. However, in persistently prion-infected cell-culture models, it was shown that treatment with monoclonal antibodies against defined regions of the prion protein (PrP) led to the clearing of PrP(Sc) in cultured cells. To gain more insight into this process, we characterized PrP-antibody complexes in solution using a fast protein liquid chromatography coupled with small-angle x-ray scattering (FPLC-SAXS) procedure. High-quality SAXS data were collected for full-length recombinant mouse PrP [denoted recPrP(23-230)] and N-terminally truncated recPrP(89-230), as well as their complexes with each of two Fab fragments (HuM-P and HuM-R1), which recognize N- and C-terminal epitopes of PrP, respectively. In-line measurements by fast protein liquid chromatography coupled with SAXS minimized data artifacts caused by a non-monodispersed sample, allowing structural analysis of PrP alone and in complex with Fab antibodies. The resulting structural models suggest two mechanisms for how these Fabs may prevent the conversion of PrP(C) into PrP(Sc).


Asunto(s)
Anticuerpos Monoclonales Humanizados/metabolismo , Proteínas PrPC/química , Proteínas PrPC/inmunología , Proteínas PrPSc/química , Animales , Cromatografía Liquida , Ratones , Modelos Moleculares , Proteínas PrPC/genética , Proteínas PrPSc/genética , Proteínas Recombinantes/química , Dispersión del Ángulo Pequeño , Soluciones , Difracción de Rayos X
18.
Biochemistry ; 54(48): 7142-55, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598179

RESUMEN

Geosmin synthase from Streptomyces coelicolor (ScGS) catalyzes an unusual, metal-dependent terpenoid cyclization and fragmentation reaction sequence. Two distinct active sites are required for catalysis: the N-terminal domain catalyzes the ionization and cyclization of farnesyl diphosphate to form germacradienol and inorganic pyrophosphate (PPi), and the C-terminal domain catalyzes the protonation, cyclization, and fragmentation of germacradienol to form geosmin and acetone through a retro-Prins reaction. A unique αα domain architecture is predicted for ScGS based on amino acid sequence: each domain contains the metal-binding motifs typical of a class I terpenoid cyclase, and each domain requires Mg(2+) for catalysis. Here, we report the X-ray crystal structure of the unliganded N-terminal domain of ScGS and the structure of its complex with three Mg(2+) ions and alendronate. These structures highlight conformational changes required for active site closure and catalysis. Although neither full-length ScGS nor constructs of the C-terminal domain could be crystallized, homology models of the C-terminal domain were constructed on the basis of ∼36% sequence identity with the N-terminal domain. Small-angle X-ray scattering experiments yield low-resolution molecular envelopes into which the N-terminal domain crystal structure and the C-terminal domain homology model were fit, suggesting possible αα domain architectures as frameworks for bifunctional catalysis.


Asunto(s)
Alendronato/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Naftoles/metabolismo , Sesquiterpenos/metabolismo , Streptomyces coelicolor/enzimología , Cristalografía por Rayos X , Ciclización , Magnesio/metabolismo , Modelos Moleculares , Fosfatos de Poliisoprenilo/metabolismo , Estructura Terciaria de Proteína , Streptomyces coelicolor/química , Streptomyces coelicolor/metabolismo
19.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 1): 45-56, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25615859

RESUMEN

Small-angle X-ray scattering (SAXS) has grown in popularity in recent times with the advent of bright synchrotron X-ray sources, powerful computational resources and algorithms enabling the calculation of increasingly complex models. However, the lack of standardized data-quality metrics presents difficulties for the growing user community in accurately assessing the quality of experimental SAXS data. Here, a series of metrics to quantitatively describe SAXS data in an objective manner using statistical evaluations are defined. These metrics are applied to identify the effects of radiation damage, concentration dependence and interparticle interactions on SAXS data from a set of 27 previously described targets for which high-resolution structures have been determined via X-ray crystallography or nuclear magnetic resonance (NMR) spectroscopy. The studies show that these metrics are sufficient to characterize SAXS data quality on a small sample set with statistical rigor and sensitivity similar to or better than manual analysis. The development of data-quality analysis strategies such as these initial efforts is needed to enable the accurate and unbiased assessment of SAXS data quality.


Asunto(s)
Dispersión del Ángulo Pequeño , Cristalografía por Rayos X , Resonancia Magnética Nuclear Biomolecular , Proteínas/química
20.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2309-27, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26527147

RESUMEN

Infection by the four serotypes of Dengue virus (DENV-1 to DENV-4) causes an important arthropod-borne viral disease in humans. The multifunctional DENV nonstructural protein 5 (NS5) is essential for capping and replication of the viral RNA and harbours a methyltransferase (MTase) domain and an RNA-dependent RNA polymerase (RdRp) domain. In this study, insights into the overall structure and flexibility of the entire NS5 of all four Dengue virus serotypes in solution are presented for the first time. The solution models derived revealed an arrangement of the full-length NS5 (NS5FL) proteins with the MTase domain positioned at the top of the RdRP domain. The DENV-1 to DENV-4 NS5 forms are elongated and flexible in solution, with DENV-4 NS5 being more compact relative to NS5 from DENV-1, DENV-2 and DENV-3. Solution studies of the individual MTase and RdRp domains show the compactness of the RdRp domain as well as the contribution of the MTase domain and the ten-residue linker region to the flexibility of the entire NS5. Swapping the ten-residue linker between DENV-4 NS5FL and DENV-3 NS5FL demonstrated its importance in MTase-RdRp communication and in concerted interaction with viral and host proteins, as probed by amide hydrogen/deuterium mass spectrometry. Conformational alterations owing to RNA binding are presented.


Asunto(s)
Virus del Dengue/química , Dengue/virología , Proteínas no Estructurales Virales/química , Secuencia de Aminoácidos , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Dispersión del Ángulo Pequeño , Alineación de Secuencia , Serogrupo , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA