Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(19): 4857-4873, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34534463

RESUMEN

The hidden world of amyloid biology has suddenly snapped into atomic-level focus, revealing over 80 amyloid protein fibrils, both pathogenic and functional. Unlike globular proteins, amyloid proteins flatten and stack into unbranched fibrils. Stranger still, a single protein sequence can adopt wildly different two-dimensional conformations, yielding distinct fibril polymorphs. Thus, an amyloid protein may define distinct diseases depending on its conformation. At the heart of this conformational variability lies structural frustrations. In functional amyloids, evolution tunes frustration levels to achieve either stability or sensitivity according to the fibril's biological function, accounting for the vast versatility of the amyloid fibril scaffold.


Asunto(s)
Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/metabolismo , Proteínas Amiloidogénicas/genética , Animales , Enfermedad/genética , Evolución Molecular , Humanos , Polimorfismo Genético , Pliegue de Proteína , Estabilidad Proteica
2.
Annu Rev Biochem ; 86: 69-95, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28125289

RESUMEN

Dozens of proteins are known to convert to the aggregated amyloid state. These include fibrils associated with systemic and neurodegenerative diseases and cancer, functional amyloid fibrils in microorganisms and animals, and many denatured proteins. Amyloid fibrils can be much more stable than other protein assemblies. In contrast to globular proteins, a single protein sequence can aggregate into several distinctly different amyloid structures, termed polymorphs, and a given polymorph can reproduce itself by seeding. Amyloid polymorphs may be the molecular basis of prion strains. Whereas the Protein Data Bank contains some 100,000 globular protein and 3,000 membrane protein structures, only a few dozen amyloid protein structures have been determined, and most of these are short segments of full amyloid-forming proteins. Regardless, these amyloid structures illuminate the architecture of the amyloid state, including its stability and its capacity for formation of polymorphs.


Asunto(s)
Proteínas Amiloidogénicas/química , Proteínas Priónicas/química , Agregación Patológica de Proteínas/metabolismo , Secuencias de Aminoácidos , Proteínas Amiloidogénicas/genética , Proteínas Amiloidogénicas/metabolismo , Animales , Microscopía por Crioelectrón , Expresión Génica , Humanos , Resonancia Magnética Nuclear Biomolecular , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Desnaturalización Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Secundaria de Proteína , Difracción de Rayos X
3.
Cell ; 161(4): 833-44, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25913193

RESUMEN

Angiotensin II type 1 receptor (AT(1)R) is a G protein-coupled receptor that serves as a primary regulator for blood pressure maintenance. Although several anti-hypertensive drugs have been developed as AT(1)R blockers (ARBs), the structural basis for AT(1)R ligand-binding and regulation has remained elusive, mostly due to the difficulties of growing high-quality crystals for structure determination using synchrotron radiation. By applying the recently developed method of serial femtosecond crystallography at an X-ray free-electron laser, we successfully determined the room-temperature crystal structure of the human AT(1)R in complex with its selective antagonist ZD7155 at 2.9-Å resolution. The AT(1)R-ZD7155 complex structure revealed key structural features of AT(1)R and critical interactions for ZD7155 binding. Docking simulations of the clinically used ARBs into the AT(1)R structure further elucidated both the common and distinct binding modes for these anti-hypertensive drugs. Our results thereby provide fundamental insights into AT(1)R structure-function relationship and structure-based drug design.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Receptor de Angiotensina Tipo 1/química , Secuencia de Aminoácidos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/química , Cristalografía por Rayos X , Humanos , Datos de Secuencia Molecular , Mutagénesis , Naftiridinas/química , Naftiridinas/farmacología , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Alineación de Secuencia
4.
Nature ; 605(7909): 304-309, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35344984

RESUMEN

Frontotemporal lobar degeneration (FTLD) is the third most common neurodegenerative condition after Alzheimer's and Parkinson's diseases1. FTLD typically presents in 45 to 64 year olds with behavioural changes or progressive decline of language skills2. The subtype FTLD-TDP is characterized by certain clinical symptoms and pathological neuronal inclusions with TAR DNA-binding protein (TDP-43) immunoreactivity3. Here we extracted amyloid fibrils from brains of four patients representing four of the five FTLD-TDP subclasses, and determined their structures by cryo-electron microscopy. Unexpectedly, all amyloid fibrils examined were composed of a 135-residue carboxy-terminal fragment of transmembrane protein 106B (TMEM106B), a lysosomal membrane protein previously implicated as a genetic risk factor for FTLD-TDP4. In addition to TMEM106B fibrils, we detected abundant non-fibrillar aggregated TDP-43 by immunogold labelling. Our observations confirm that FTLD-TDP is associated with amyloid fibrils, and that the fibrils are formed by TMEM106B rather than TDP-43.


Asunto(s)
Amiloide , Proteínas de Unión al ADN , Degeneración Lobar Frontotemporal , Proteínas de la Membrana , Proteínas del Tejido Nervioso , Amiloide/ultraestructura , Microscopía por Crioelectrón , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/ultraestructura , Degeneración Lobar Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/patología , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/ultraestructura
5.
Cell ; 149(4): 753-67, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22579281

RESUMEN

Eukaryotic cells contain assemblies of RNAs and proteins termed RNA granules. Many proteins within these bodies contain KH or RRM RNA-binding domains as well as low complexity (LC) sequences of unknown function. We discovered that exposure of cell or tissue lysates to a biotinylated isoxazole (b-isox) chemical precipitated hundreds of RNA-binding proteins with significant overlap to the constituents of RNA granules. The LC sequences within these proteins are both necessary and sufficient for b-isox-mediated aggregation, and these domains can undergo a concentration-dependent phase transition to a hydrogel-like state in the absence of the chemical. X-ray diffraction and EM studies revealed the hydrogels to be composed of uniformly polymerized amyloid-like fibers. Unlike pathogenic fibers, the LC sequence-based polymers described here are dynamic and accommodate heterotypic polymerization. These observations offer a framework for understanding the function of LC sequences as well as an organizing principle for cellular structures that are not membrane bound.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Hidrogel de Polietilenoglicol-Dimetacrilato/metabolismo , Proteínas de Unión al ARN/análisis , ARN/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Sistema Libre de Células , Gránulos Citoplasmáticos/química , Células Madre Embrionarias/metabolismo , Masculino , Ratones , Modelos Moleculares , Células 3T3 NIH , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Testículo/citología , Testículo/metabolismo , Difracción de Rayos X
6.
Proc Natl Acad Sci U S A ; 120(37): e2305494120, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37669364

RESUMEN

Cryoelectron microscopy (Cryo-EM) has enabled structural determination of proteins larger than about 50 kDa, including many intractable by any other method, but it has largely failed for smaller proteins. Here, we obtain structures of small proteins by binding them to a rigid molecular scaffold based on a designed protein cage, revealing atomic details at resolutions reaching 2.9 Å. We apply this system to the key cancer signaling protein KRAS (19 kDa in size), obtaining four structures of oncogenic mutational variants by cryo-EM. Importantly, a structure for the key G12C mutant bound to an inhibitor drug (AMG510) reveals significant conformational differences compared to prior data in the crystalline state. The findings highlight the promise of cryo-EM scaffolds for advancing the design of drug molecules against small therapeutic protein targets in cancer and other human diseases.


Asunto(s)
Diagnóstico por Imagen , Humanos , Microscopía por Crioelectrón
7.
Proc Natl Acad Sci U S A ; 120(41): e2300258120, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37801475

RESUMEN

Despite much effort, antibody therapies for Alzheimer's disease (AD) have shown limited efficacy. Challenges to the rational design of effective antibodies include the difficulty of achieving specific affinity to critical targets, poor expression, and antibody aggregation caused by buried charges and unstructured loops. To overcome these challenges, we grafted previously determined sequences of fibril-capping amyloid inhibitors onto a camel heavy chain antibody scaffold. These sequences were designed to cap fibrils of tau, known to form the neurofibrillary tangles of AD, thereby preventing fibril elongation. The nanobodies grafted with capping inhibitors blocked tau aggregation in biosensor cells seeded with postmortem brain extracts from AD and progressive supranuclear palsy (PSP) patients. The tau capping nanobody inhibitors also blocked seeding by recombinant tau oligomers. Another challenge to the design of effective antibodies is their poor blood-brain barrier (BBB) penetration. In this study, we also designed a bispecific nanobody composed of a nanobody that targets a receptor on the BBB and a tau capping nanobody inhibitor, conjoined by a flexible linker. We provide evidence that the bispecific nanobody improved BBB penetration over the tau capping inhibitor alone after intravenous administration in mice. Our results suggest that the design of synthetic antibodies that target sequences that drive protein aggregation may be a promising approach to inhibit the prion-like seeding of tau and other proteins involved in AD and related proteinopathies.


Asunto(s)
Enfermedad de Alzheimer , Anticuerpos de Dominio Único , Parálisis Supranuclear Progresiva , Humanos , Animales , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Proteínas tau/metabolismo , Anticuerpos de Dominio Único/farmacología , Anticuerpos de Dominio Único/metabolismo , Ovillos Neurofibrilares/metabolismo , Parálisis Supranuclear Progresiva/metabolismo , Anticuerpos/metabolismo , Encéfalo/metabolismo
8.
J Biol Chem ; 300(2): 105531, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38072051

RESUMEN

Heterogeneous nuclear ribonucleoprotein A2 (hnRNPA2) is a human ribonucleoprotein that transports RNA to designated locations for translation via its ability to phase separate. Its mutated form, D290V, is implicated in multisystem proteinopathy known to afflict two families, mainly with myopathy and Paget's disease of bone. Here, we investigate this mutant form of hnRNPA2 by determining cryo-EM structures of the recombinant D290V low complexity domain. We find that the mutant form of hnRNPA2 differs from the WT fibrils in four ways. In contrast to the WT fibrils, the PY-nuclear localization signals in the fibril cores of all three mutant polymorphs are less accessible to chaperones. Also, the mutant fibrils are more stable than WT fibrils as judged by phase separation, thermal stability, and energetic calculations. Similar to other pathogenic amyloids, the mutant fibrils are polymorphic. Thus, these structures offer evidence to explain how a D-to-V missense mutation diverts the assembly of reversible, functional amyloid-like fibrils into the assembly of pathogenic amyloid, and may shed light on analogous conversions occurring in other ribonucleoproteins that lead to neurological diseases such as amyotrophic lateral sclerosis and frontotemporal dementia.


Asunto(s)
Microscopía por Crioelectrón , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B , Modelos Moleculares , Humanos , Separación de Fases , Dominios Proteicos , Mutación , Concentración de Iones de Hidrógeno , Estabilidad Proteica , Estructura Terciaria de Proteína , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/química , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo
9.
Proc Natl Acad Sci U S A ; 119(15): e2119952119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35377792

RESUMEN

In neurodegenerative diseases including Alzheimer's and amyotrophic lateral sclerosis, proteins that bind RNA are found in aggregated forms in autopsied brains. Evidence suggests that RNA aids nucleation of these pathological aggregates; however, the mechanism has not been investigated at the level of atomic structure. Here, we present the 3.4-Å resolution structure of fibrils of full-length recombinant tau protein in the presence of RNA, determined by electron cryomicroscopy (cryo-EM). The structure reveals the familiar in-register cross-ß amyloid scaffold but with a small fibril core spanning residues Glu391 to Ala426, a region disordered in the fuzzy coat in all previously studied tau polymorphs. RNA is bound on the fibril surface to the positively charged residues Arg406 and His407 and runs parallel to the fibril axis. The fibrils dissolve when RNase is added, showing that RNA is necessary for fibril integrity. While this structure cannot exist simultaneously with the tau fibril structures extracted from patients' brains, it could conceivably account for the nucleating effects of RNA cofactors followed by remodeling as fibrils mature.


Asunto(s)
Amiloide , ARN , Proteínas tau , Amiloide/química , Microscopía por Crioelectrón , Humanos , ARN/química , Proteínas tau/química
10.
J Biol Chem ; 299(4): 104566, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36871760

RESUMEN

Synucleinopathies like Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple systems atrophy (MSA), have the same pathologic feature of misfolded α-synuclein protein (α-syn) accumulation in the brain. PD patients who carry α-syn hereditary mutations tend to have earlier onset and more severe clinical symptoms than sporadic PD patients. Therefore, revealing the effect of hereditary mutations to the α-syn fibril structure can help us understand these synucleinopathies' structural basis. Here, we present a 3.38 Å cryo-electron microscopy structure of α-synuclein fibrils containing the hereditary A53E mutation. The A53E fibril is symmetrically composed of two protofilaments, similar to other fibril structures of WT and mutant α-synuclein. The new structure is distinct from all other synuclein fibrils, not only at the interface between proto-filaments, but also between residues packed within the same proto-filament. A53E has the smallest interface with the least buried surface area among all α-syn fibrils, consisting of only two contacting residues. Within the same protofilament, A53E reveals distinct residue re-arrangement and structural variation at a cavity near its fibril core. Moreover, the A53E fibrils exhibit slower fibril formation and lower stability compared to WT and other mutants like A53T and H50Q, while also demonstrate strong cellular seeding in α-synuclein biosensor cells and primary neurons. In summary, our study aims to highlight structural differences - both within and between the protofilaments of A53E fibrils - and interpret fibril formation and cellular seeding of α-synuclein pathology in disease, which could further our understanding of the structure-activity relationship of α-synuclein mutants.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Humanos , alfa-Sinucleína/metabolismo , Microscopía por Crioelectrón , Amiloide/química , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Mutación
11.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33372136

RESUMEN

Proteins are commonly known to transfer electrons over distances limited to a few nanometers. However, many biological processes require electron transport over far longer distances. For example, soil and sediment bacteria transport electrons, over hundreds of micrometers to even centimeters, via putative filamentous proteins rich in aromatic residues. However, measurements of true protein conductivity have been hampered by artifacts due to large contact resistances between proteins and electrodes. Using individual amyloid protein crystals with atomic-resolution structures as a model system, we perform contact-free measurements of intrinsic electronic conductivity using a four-electrode approach. We find hole transport through micrometer-long stacked tyrosines at physiologically relevant potentials. Notably, the transport rate through tyrosines (105 s-1) is comparable to cytochromes. Our studies therefore show that amyloid proteins can efficiently transport charges, under ordinary thermal conditions, without any need for redox-active metal cofactors, large driving force, or photosensitizers to generate a high oxidation state for charge injection. By measuring conductivity as a function of molecular length, voltage, and temperature, while eliminating the dominant contribution of contact resistances, we show that a multistep hopping mechanism (composed of multiple tunneling steps), not single-step tunneling, explains the measured conductivity. Combined experimental and computational studies reveal that proton-coupled electron transfer confers conductivity; both the energetics of the proton acceptor, a neighboring glutamine, and its proximity to tyrosine influence the hole transport rate through a proton rocking mechanism. Surprisingly, conductivity increases 200-fold upon cooling due to higher availability of the proton acceptor by increased hydrogen bonding.


Asunto(s)
Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/fisiología , Proteínas/fisiología , Citocromos/química , Citocromos/fisiología , Conductividad Eléctrica , Transporte de Electrón/fisiología , Electrones , Enlace de Hidrógeno , Modelos Biológicos , Simulación de Dinámica Molecular , Oxidación-Reducción , Proteínas/química , Protones , Tirosina/química
12.
J Biol Chem ; 298(10): 102396, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988647

RESUMEN

Amyloid protein aggregation is commonly associated with progressive neurodegenerative diseases, however not all amyloid fibrils are pathogenic. The neuronal cytoplasmic polyadenylation element binding protein is a regulator of synaptic mRNA translation and has been shown to form functional amyloid aggregates that stabilize long-term memory. In adult Drosophila neurons, the cytoplasmic polyadenylation element binding homolog Orb2 is expressed as 2 isoforms, of which the Orb2B isoform is far more abundant, but the rarer Orb2A isoform is required to initiate Orb2 aggregation. The N terminus is a distinctive feature of the Orb2A isoform and is critical for its aggregation. Intriguingly, replacement of phenylalanine in the fifth position of Orb2A with tyrosine (F5Y) in Drosophila impairs stabilization of long-term memory. The structure of endogenous Orb2B fibers was recently determined by cryo-EM, but the structure adopted by fibrillar Orb2A is less certain. Here we use micro-electron diffraction to determine the structure of the first 9 N-terminal residues of Orb2A, at a resolution of 1.05 Å. We find that this segment (which we term M9I) forms an amyloid-like array of parallel in-register ß-sheets, which interact through side chain interdigitation of aromatic and hydrophobic residues. Our structure provides an explanation for the decreased aggregation observed for the F5Y mutant and offers a hypothesis for how the addition of a single atom (the tyrosyl oxygen) affects long-term memory. We also propose a structural model of Orb2A that integrates our structure of the M9I segment with the published Orb2B cryo-EM structure.


Asunto(s)
Péptidos beta-Amiloides , Amiloide , Proteínas de Drosophila , Drosophila melanogaster , Agregado de Proteínas , Factores de Transcripción , Factores de Escisión y Poliadenilación de ARNm , Animales , Amiloide/química , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Electrones , Factores de Escisión y Poliadenilación de ARNm/química , Factores de Escisión y Poliadenilación de ARNm/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Neuronas/metabolismo , Conformación Proteica en Lámina beta , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Drosophila melanogaster/metabolismo
13.
J Biol Chem ; 298(7): 102111, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35690147

RESUMEN

Mevalonate 3,5-bisphosphate decarboxylase is involved in the recently discovered Thermoplasma-type mevalonate pathway. The enzyme catalyzes the elimination of the 3-phosphate group from mevalonate 3,5-bisphosphate as well as concomitant decarboxylation of the substrate. This entire reaction of the enzyme resembles the latter half-reactions of its homologs, diphosphomevalonate decarboxylase and phosphomevalonate decarboxylase, which also catalyze ATP-dependent phosphorylation of the 3-hydroxyl group of their substrates. However, the crystal structure of mevalonate 3,5-bisphosphate decarboxylase and the structural reasons of the difference between reactions catalyzed by the enzyme and its homologs are unknown. In this study, we determined the X-ray crystal structure of mevalonate 3,5-bisphosphate decarboxylase from Picrophilus torridus, a thermoacidophilic archaeon of the order Thermoplasmatales. Structural and mutational analysis demonstrated the importance of a conserved aspartate residue for enzyme activity. In addition, although crystallization was performed in the absence of substrate or ligands, residual electron density having the shape of a fatty acid was observed at a position overlapping the ATP-binding site of the homologous enzyme, diphosphomevalonate decarboxylase. This finding is in agreement with the expected evolutionary route from phosphomevalonate decarboxylase (ATP-dependent) to mevalonate 3,5-bisphosphate decarboxylase (ATP-independent) through the loss of kinase activity. We found that the binding of geranylgeranyl diphosphate, an intermediate of the archeal isoprenoid biosynthesis pathway, evoked significant activation of mevalonate 3,5-bisphosphate decarboxylase, and several mutations at the putative geranylgeranyl diphosphate-binding site impaired this activation, suggesting the physiological importance of ligand binding as well as a possible novel regulatory system employed by the Thermoplasma-type mevalonate pathway.


Asunto(s)
Carboxiliasas/química , Thermoplasmales/enzimología , Adenosina Trifosfato/metabolismo , Carboxiliasas/metabolismo , Redes y Vías Metabólicas , Ácido Mevalónico/metabolismo
14.
J Am Chem Soc ; 145(47): 25917-25926, 2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-37972334

RESUMEN

The rippled ß-sheet was theorized by Pauling and Corey in 1953 as a structural motif in which mirror image peptide strands assemble into hydrogen-bonded periodic arrays with strictly alternating chirality. Structural characterization of the rippled ß-sheet was limited to biophysical methods until 2022 when atomic resolution structures of the motif were first obtained. The crystal structural foundation is restricted to four model tripeptides composed exclusively of aromatic residues. Here, we report five new rippled sheet crystal structures derived from amyloid ß and amylin, the aggregating toxic peptides of Alzheimer's disease and type II diabetes, respectively. Despite the variation in peptide sequence composition, all five structures form antiparallel rippled ß-sheets that extend, like a fibril, along the entire length of the crystalline needle. The long-range packing of the crystals, however, varies. In three of the crystals, the sheets pack face-to-face and exclude water, giving rise to cross-ß architectures grossly resembling the steric zipper motif of amyloid fibrils but differing in fundamental details. In the other two crystals, the solvent is encapsulated between the sheets, yielding fibril architectures capable of host-guest chemistry. Our study demonstrates that the formation of rippled ß-sheets from aggregating racemic peptide mixtures in three-dimensional (3D) assemblies is a general phenomenon and provides a structural basis for targeting intrinsically disordered proteins.


Asunto(s)
Péptidos beta-Amiloides , Diabetes Mellitus Tipo 2 , Humanos , Péptidos beta-Amiloides/química , Conformación Proteica en Lámina beta , Polipéptido Amiloide de los Islotes Pancreáticos , Modelos Moleculares , Amiloide/química
15.
Cell ; 134(6): 1007-18, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18805093

RESUMEN

Drosophila Dscam encodes a vast family of immunoglobulin (Ig)-containing proteins that exhibit isoform-specific homophilic binding. This diversity is essential for cell recognition events required for wiring the brain. Each isoform binds to itself but rarely to other isoforms. Specificity is determined by "matching" of three variable Ig domains within an approximately 220 kD ectodomain. Here, we present the structure of the homophilic binding region of Dscam, comprising the eight N-terminal Ig domains (Dscam(1-8)). Dscam(1-8) forms a symmetric homodimer of S-shaped molecules. This conformation, comprising two reverse turns, allows each pair of the three variable domains to "match" in an antiparallel fashion. Structural, genetic, and biochemical studies demonstrate that, in addition to variable domain "matching," intramolecular interactions between constant domains promote homophilic binding. These studies provide insight into how "matching" at all three pairs of variable domains in Dscam mediates isoform-specific recognition.


Asunto(s)
Proteínas de Drosophila/química , Drosophila melanogaster/metabolismo , Animales , Sitios de Unión , Moléculas de Adhesión Celular , Cristalografía por Rayos X , Proteínas de Drosophila/metabolismo , Inmunoglobulinas/química , Inmunoglobulinas/metabolismo , Modelos Moleculares , Pliegue de Proteína , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
16.
Mol Cell ; 57(6): 1011-1021, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25728769

RESUMEN

Eosinophils are white blood cells that function in innate immunity and participate in the pathogenesis of various inflammatory and neoplastic disorders. Their secretory granules contain four cytotoxic proteins, including the eosinophil major basic protein (MBP-1). How MBP-1 toxicity is controlled within the eosinophil itself and activated upon extracellular release is unknown. Here we show how intragranular MBP-1 nanocrystals restrain toxicity, enabling its safe storage, and characterize them with an X-ray-free electron laser. Following eosinophil activation, MBP-1 toxicity is triggered by granule acidification, followed by extracellular aggregation, which mediates the damage to pathogens and host cells. Larger non-toxic amyloid plaques are also present in tissues of eosinophilic patients in a feedback mechanism that likely limits tissue damage under pathological conditions of MBP-1 oversecretion. Our results suggest that MBP-1 aggregation is important for innate immunity and immunopathology mediated by eosinophils and clarify how its polymorphic self-association pathways regulate toxicity intra- and extracellularly.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Eosinófilos/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Celulitis (Flemón)/metabolismo , Celulitis (Flemón)/patología , Proteínas de Unión al ADN/toxicidad , Dermatitis Atópica/metabolismo , Dermatitis Atópica/patología , Eosinofilia/metabolismo , Eosinofilia/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Escherichia coli/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/fisiología , Ratones Endogámicos C57BL , Nanopartículas/metabolismo , Nanopartículas/toxicidad , Vesículas Secretoras/metabolismo , Piel/efectos de los fármacos , Piel/patología
17.
Proc Natl Acad Sci U S A ; 117(7): 3592-3602, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32015135

RESUMEN

Aggregation of α-synuclein is a defining molecular feature of Parkinson's disease, Lewy body dementia, and multiple systems atrophy. Hereditary mutations in α-synuclein are linked to both Parkinson's disease and Lewy body dementia; in particular, patients bearing the E46K disease mutation manifest a clinical picture of parkinsonism and Lewy body dementia, and E46K creates more pathogenic fibrils in vitro. Understanding the effect of these hereditary mutations on α-synuclein fibril structure is fundamental to α-synuclein biology. We therefore determined the cryo-electron microscopy (cryo-EM) structure of α-synuclein fibrils containing the hereditary E46K mutation. The 2.5-Å structure reveals a symmetric double protofilament in which the molecules adopt a vastly rearranged, lower energy fold compared to wild-type fibrils. We propose that the E46K misfolding pathway avoids electrostatic repulsion between K46 and K80, a residue pair which form the E46-K80 salt bridge in the wild-type fibril structure. We hypothesize that, under our conditions, the wild-type fold does not reach this deeper energy well of the E46K fold because the E46-K80 salt bridge diverts α-synuclein into a kinetic trap-a shallower, more accessible energy minimum. The E46K mutation apparently unlocks a more stable and pathogenic fibril structure.


Asunto(s)
Enfermedad por Cuerpos de Lewy/genética , Mutación Missense , Enfermedad de Parkinson/genética , alfa-Sinucleína/química , alfa-Sinucleína/genética , Secuencias de Aminoácidos , Microscopía por Crioelectrón , Humanos , Enfermedad por Cuerpos de Lewy/congénito , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad de Parkinson/congénito , Enfermedad de Parkinson/metabolismo , Pliegue de Proteína , alfa-Sinucleína/metabolismo
18.
Nature ; 539(7627): 43-47, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27680699

RESUMEN

BinAB is a naturally occurring paracrystalline larvicide distributed worldwide to combat the devastating diseases borne by mosquitoes. These crystals are composed of homologous molecules, BinA and BinB, which play distinct roles in the multi-step intoxication process, transforming from harmless, robust crystals, to soluble protoxin heterodimers, to internalized mature toxin, and finally to toxic oligomeric pores. The small size of the crystals-50 unit cells per edge, on average-has impeded structural characterization by conventional means. Here we report the structure of Lysinibacillus sphaericus BinAB solved de novo by serial-femtosecond crystallography at an X-ray free-electron laser. The structure reveals tyrosine- and carboxylate-mediated contacts acting as pH switches to release soluble protoxin in the alkaline larval midgut. An enormous heterodimeric interface appears to be responsible for anchoring BinA to receptor-bound BinB for co-internalization. Remarkably, this interface is largely composed of propeptides, suggesting that proteolytic maturation would trigger dissociation of the heterodimer and progression to pore formation.


Asunto(s)
Bacillus/química , Toxinas Bacterianas/química , Culicidae , Insecticidas/química , Larva , Rayos Láser , Animales , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Culicidae/metabolismo , Concentración de Iones de Hidrógeno , Larva/química , Larva/metabolismo , Modelos Moleculares , Multimerización de Proteína , Proteolisis , Tirosina/química
19.
J Biol Chem ; 295(31): 10662-10676, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32493775

RESUMEN

Soluble oligomers of aggregated tau accompany the accumulation of insoluble amyloid fibrils, a histological hallmark of Alzheimer disease (AD) and two dozen related neurodegenerative diseases. Both oligomers and fibrils seed the spread of Tau pathology, and by virtue of their low molecular weight and relative solubility, oligomers may be particularly pernicious seeds. Here, we report the formation of in vitro tau oligomers formed by an ionic liquid (IL15). Using IL15-induced recombinant tau oligomers and a dot blot assay, we discovered a mAb (M204) that binds oligomeric tau, but not tau monomers or fibrils. M204 and an engineered single-chain variable fragment (scFv) inhibited seeding by IL15-induced tau oligomers and pathological extracts from donors with AD and chronic traumatic encephalopathy. This finding suggests that M204-scFv targets pathological structures that are formed by tau in neurodegenerative diseases. We found that M204-scFv itself partitions into oligomeric forms that inhibit seeding differently, and crystal structures of the M204-scFv monomer, dimer, and trimer revealed conformational differences that explain differences among these forms in binding and inhibition. The efficiency of M204-scFv antibodies to inhibit the seeding by brain tissue extracts from different donors with tauopathies varied among individuals, indicating the possible existence of distinct amyloid polymorphs. We propose that by binding to oligomers, which are hypothesized to be the earliest seeding-competent species, M204-scFv may have potential as an early-stage diagnostic for AD and tauopathies, and also could guide the development of promising therapeutic antibodies.


Asunto(s)
Enfermedad de Alzheimer , Multimerización de Proteína , Anticuerpos de Cadena Única/química , Proteínas tau/química , Cristalografía por Rayos X , Humanos
20.
PLoS Pathog ; 15(4): e1007723, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31002736

RESUMEN

Staphylococcus aureus and other bacterial pathogens affix wall teichoic acids (WTAs) to their surface. These highly abundant anionic glycopolymers have critical functions in bacterial physiology and their susceptibility to ß-lactam antibiotics. The membrane-associated TagA glycosyltransferase (GT) catalyzes the first-committed step in WTA biosynthesis and is a founding member of the WecB/TagA/CpsF GT family, more than 6,000 enzymes that synthesize a range of extracellular polysaccharides through a poorly understood mechanism. Crystal structures of TagA from T. italicus in its apo- and UDP-bound states reveal a novel GT fold, and coupled with biochemical and cellular data define the mechanism of catalysis. We propose that enzyme activity is regulated by interactions with the bilayer, which trigger a structural change that facilitates proper active site formation and recognition of the enzyme's lipid-linked substrate. These findings inform upon the molecular basis of WecB/TagA/CpsF activity and could guide the development of new anti-microbial drugs.


Asunto(s)
Proteínas Bacterianas/química , Pared Celular/metabolismo , Lipoproteínas/química , Staphylococcus aureus/enzimología , Ácidos Teicoicos/metabolismo , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Lipoproteínas/metabolismo , Modelos Moleculares , Multimerización de Proteína , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA