Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(7): e2217835120, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36757890

RESUMEN

The amyloid aggregation of alpha-synuclein within the brain is associated with the pathogenesis of Parkinson's disease (PD) and other related synucleinopathies, including multiple system atrophy (MSA). Alpha-synuclein aggregates are a major therapeutic target for treatment of these diseases. We identify two small molecules capable of disassembling preformed alpha-synuclein fibrils. The compounds, termed CNS-11 and CNS-11g, disaggregate recombinant alpha-synuclein fibrils in vitro, prevent the intracellular seeded aggregation of alpha-synuclein fibrils, and mitigate alpha-synuclein fibril cytotoxicity in neuronal cells. Furthermore, we demonstrate that both compounds disassemble fibrils extracted from MSA patient brains and prevent their intracellular seeding. They also reduce in vivo alpha-synuclein aggregates in C. elegans. Both compounds also penetrate brain tissue in mice. A molecular dynamics-based computational model suggests the compounds may exert their disaggregating effects on the N terminus of the fibril core. These compounds appear to be promising therapeutic leads for targeting alpha-synuclein for the treatment of synucleinopathies.


Asunto(s)
Atrofia de Múltiples Sistemas , Enfermedad de Parkinson , Sinucleinopatías , Ratones , Animales , alfa-Sinucleína/metabolismo , Sinucleinopatías/patología , Caenorhabditis elegans/metabolismo , Enfermedad de Parkinson/patología , Atrofia de Múltiples Sistemas/patología , Encéfalo/metabolismo , Amiloide/metabolismo
2.
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
3.
Proc Natl Acad Sci U S A ; 119(34): e2206240119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969734

RESUMEN

Neurodegenerative diseases are characterized by the pathologic accumulation of aggregated proteins. Known as amyloid, these fibrillar aggregates include proteins such as tau and amyloid-ß (Aß) in Alzheimer's disease (AD) and alpha-synuclein (αSyn) in Parkinson's disease (PD). The development and spread of amyloid fibrils within the brain correlates with disease onset and progression, and inhibiting amyloid formation is a possible route toward therapeutic development. Recent advances have enabled the determination of amyloid fibril structures to atomic-level resolution, improving the possibility of structure-based inhibitor design. In this work, we use these amyloid structures to design inhibitors that bind to the ends of fibrils, "capping" them so as to prevent further growth. Using de novo protein design, we develop a library of miniprotein inhibitors of 35 to 48 residues that target the amyloid structures of tau, Aß, and αSyn. Biophysical characterization of top in silico designed inhibitors shows they form stable folds, have no sequence similarity to naturally occurring proteins, and specifically prevent the aggregation of their targeted amyloid-prone proteins in vitro. The inhibitors also prevent the seeded aggregation and toxicity of fibrils in cells. In vivo evaluation reveals their ability to reduce aggregation and rescue motor deficits in Caenorhabditis elegans models of PD and AD.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Agregación Patológica de Proteínas/tratamiento farmacológico , alfa-Sinucleína/antagonistas & inhibidores , Proteínas tau/antagonistas & inhibidores , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Amiloide/química , Péptidos beta-Amiloides/metabolismo , Amiloidosis , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/química
4.
J Biol Chem ; 298(5): 101920, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35405097

RESUMEN

Low-complexity domains (LCDs) of proteins have been shown to self-associate, and pathogenic mutations within these domains often drive the proteins into amyloid aggregation associated with disease. These domains may be especially susceptible to amyloidogenic mutations because they are commonly intrinsically disordered and function in self-association. The question therefore arises whether a search for pathogenic mutations in LCDs of the human proteome can lead to identification of other proteins associated with amyloid disease. Here, we take a computational approach to identify documented pathogenic mutations within LCDs that may favor amyloid formation. Using this approach, we identify numerous known amyloidogenic mutations, including several such mutations within proteins previously unidentified as amyloidogenic. Among the latter group, we focus on two mutations within the TRK-fused gene protein (TFG), known to play roles in protein secretion and innate immunity, which are associated with two different peripheral neuropathies. We show that both mutations increase the propensity of TFG to form amyloid fibrils. We therefore conclude that TFG is a novel amyloid protein and propose that the diseases associated with its mutant forms may be amyloidoses.


Asunto(s)
Proteínas Amiloidogénicas , Amiloidosis , Biología Computacional , Amiloide/genética , Amiloide/metabolismo , Proteínas Amiloidogénicas/genética , Amiloidosis/metabolismo , Amiloidosis/patología , Humanos , Mutación , Proteoma/genética
5.
J Biol Chem ; 294(44): 16451-16464, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31537646

RESUMEN

In Alzheimer's disease (AD) and tauopathies, tau aggregation accompanies progressive neurodegeneration. Aggregated tau appears to spread between adjacent neurons and adjacent brain regions by prion-like seeding. Hence, inhibitors of this seeding offer a possible route to managing tauopathies. Here, we report the 1.0 Å resolution micro-electron diffraction structure of an aggregation-prone segment of tau with the sequence SVQIVY, present in the cores of patient-derived fibrils from AD and tauopathies. This structure illuminates how distinct interfaces of the parent segment, containing the sequence VQIVYK, foster the formation of distinct structures. Peptide-based fibril-capping inhibitors designed to target the two VQIVYK interfaces blocked proteopathic seeding by patient-derived fibrils. These VQIVYK inhibitors add to a panel of tau-capping inhibitors that targets specific polymorphs of recombinant and patient-derived tau fibrils. Inhibition of seeding initiated by brain tissue extracts differed among donors with different tauopathies, suggesting that particular fibril polymorphs of tau are associated with certain tauopathies. Donors with progressive supranuclear palsy exhibited more variation in inhibitor sensitivity, suggesting that fibrils from these donors were more polymorphic and potentially vary within individual donor brains. Our results suggest that a subset of inhibitors from our panel could be specific for particular disease-associated polymorphs, whereas inhibitors that blocked seeding by extracts from all of the tauopathies tested could be used to broadly inhibit seeding by multiple disease-specific tau polymorphs. Moreover, we show that tau-capping inhibitors can be transiently expressed in HEK293 tau biosensor cells, indicating that nucleic acid-based vectors can be used for inhibitor delivery.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Tauopatías/metabolismo , Proteínas tau/ultraestructura , Encéfalo/metabolismo , Células HEK293 , Humanos , Neuronas/metabolismo , Priones/metabolismo , Agregación Patológica de Proteínas/metabolismo , Proteínas tau/metabolismo
6.
Chemistry ; 21(10): 3987-96, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25649221

RESUMEN

The chemistry of [Fe]-hydrogenase has attracted significant interest due to its ability to activate molecular hydrogen. The intriguing properties of this enzyme have prompted the synthesis of numerous small molecule mimics aimed at activating H2. Despite considerable effort, a majority of these compounds remain nonfunctional for hydrogenation reactions. By using a recently synthesized model as an entry point, seven biomimetic complexes have been examined through DFT computations to probe the influence of ligand environment on the ability of a mimic to bind and split H2. One mimic, featuring a bidentate diphosphine group incorporating an internal nitrogen base, was found to have particularly attractive energetics, prompting a study of the role played by the proton/hydride acceptor necessary to complete the catalytic cycle. Computations revealed an experimentally accessible energetic pathway involving a benzaldehyde proton/hydride acceptor and the most promising catalyst.


Asunto(s)
Benzaldehídos/química , Hidrogenasas/química , Proteínas Hierro-Azufre/química , Biomimética , Catálisis , Computadores Moleculares , Hidrogenación , Ligandos , Modelos Moleculares , Estructura Molecular , Protones , Teoría Cuántica
7.
Nat Commun ; 14(1): 2379, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37185252

RESUMEN

The self-assembly of the Nucleocapsid protein (NCAP) of SARS-CoV-2 is crucial for its function. Computational analysis of the amino acid sequence of NCAP reveals low-complexity domains (LCDs) akin to LCDs in other proteins known to self-assemble as phase separation droplets and amyloid fibrils. Previous reports have described NCAP's propensity to phase-separate. Here we show that the central LCD of NCAP is capable of both, phase separation and amyloid formation. Within this central LCD we identified three adhesive segments and determined the atomic structure of the fibrils formed by each. Those structures guided the design of G12, a peptide that interferes with the self-assembly of NCAP and demonstrates antiviral activity in SARS-CoV-2 infected cells. Our work, therefore, demonstrates the amyloid form of the central LCD of NCAP and suggests that amyloidogenic segments of NCAP could be targeted for drug development.


Asunto(s)
Amiloide , COVID-19 , Proteínas de la Nucleocápside de Coronavirus , Humanos , Amiloide/metabolismo , Proteínas Amiloidogénicas , Proteínas de la Nucleocápside , Péptidos/química , Dominios Proteicos , SARS-CoV-2/metabolismo
8.
ACS Nano ; 16(2): 2154-2163, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35132852

RESUMEN

The assembly of proteins into fibrillar amyloid structures was once considered to be pathologic and essentially irreversible. Recent studies reveal amyloid-like structures that form reversibly, derived from protein low-complexity domains which function in cellular metabolism. Here, by comparing atomic-level structures of reversible and irreversible amyloid fibrils, we find that the ß-sheets of reversible fibrils are enriched in flattened (as opposed to pleated) ß-sheets formed by stacking of extended ß-strands. Quantum mechanical calculations show that glycine residues favor extended ß-strands which may be stabilized by intraresidue interactions between the amide proton and the carbonyl oxygen, known as C5 hydrogen-bonds. Larger residue side chains favor shorter strands and pleated sheets. These findings highlight a structural element that may regulate reversible amyloid assembly.


Asunto(s)
Amiloide , Proteínas Amiloidogénicas , Amiloide/química , Péptidos beta-Amiloides/química , Enlace de Hidrógeno , Conformación Proteica en Lámina beta , Dominios Proteicos
9.
Nat Struct Mol Biol ; 29(6): 529-536, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35637421

RESUMEN

Proteins including FUS, hnRNPA2, and TDP-43 reversibly aggregate into amyloid-like fibrils through interactions of their low-complexity domains (LCDs). Mutations in LCDs can promote irreversible amyloid aggregation and disease. We introduce a computational approach to identify mutations in LCDs of disease-associated proteins predicted to increase propensity for amyloid aggregation. We identify several disease-related mutations in the intermediate filament protein keratin-8 (KRT8). Atomic structures of wild-type and mutant KRT8 segments confirm the transition to a pleated strand capable of amyloid formation. Biochemical analysis reveals KRT8 forms amyloid aggregates, and the identified mutations promote aggregation. Aggregated KRT8 is found in Mallory-Denk bodies, observed in hepatocytes of livers with alcoholic steatohepatitis (ASH). We demonstrate that ethanol promotes KRT8 aggregation, and KRT8 amyloids co-crystallize with alcohol. Lastly, KRT8 aggregation can be seeded by liver extract from people with ASH, consistent with the amyloid nature of KRT8 aggregates and the classification of ASH as an amyloid-related condition.


Asunto(s)
Amiloide , Hígado , Amiloide/genética , Amiloide/metabolismo , Proteínas Amiloidogénicas/genética , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Mutación , Dominios Proteicos
10.
Nat Commun ; 13(1): 5451, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36114178

RESUMEN

Alzheimer's disease (AD) is the consequence of neuronal death and brain atrophy associated with the aggregation of protein tau into fibrils. Thus disaggregation of tau fibrils could be a therapeutic approach to AD. The small molecule EGCG, abundant in green tea, has long been known to disaggregate tau and other amyloid fibrils, but EGCG has poor drug-like properties, failing to fully penetrate the brain. Here we have cryogenically trapped an intermediate of brain-extracted tau fibrils on the kinetic pathway to EGCG-induced disaggregation and have determined its cryoEM structure. The structure reveals that EGCG molecules stack in polar clefts between the paired helical protofilaments that pathologically define AD. Treating the EGCG binding position as a pharmacophore, we computationally screened thousands of drug-like compounds for compatibility for the pharmacophore, discovering several that experimentally disaggregate brain-derived tau fibrils in vitro. This work suggests the potential of structure-based, small-molecule drug discovery for amyloid diseases.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Proteínas tau , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Amiloide/química , Amiloide/efectos de los fármacos , Catequina/análogos & derivados , Catequina/química , Catequina/farmacología , Microscopía por Crioelectrón , Evaluación Preclínica de Medicamentos/métodos , Humanos , Té/química , Proteínas tau/química , Proteínas tau/efectos de los fármacos , Proteínas tau/metabolismo
11.
Nat Struct Mol Biol ; 28(9): 724-730, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34518699

RESUMEN

Amyloidosis of human islet amyloid polypeptide (hIAPP) is a pathological hallmark of type II diabetes (T2D), an epidemic afflicting nearly 10% of the world's population. To visualize disease-relevant hIAPP fibrils, we extracted amyloid fibrils from islet cells of a T2D donor and amplified their quantity by seeding synthetic hIAPP. Cryo-EM studies revealed four fibril polymorphic atomic structures. Their resemblance to four unseeded hIAPP fibrils varies from nearly identical (TW3) to non-existent (TW2). The diverse repertoire of hIAPP polymorphs appears to arise from three distinct protofilament cores entwined in different combinations. The structural distinctiveness of TW1, TW2 and TW4 suggests they may be faithful replications of the pathogenic seeds. If so, the structures determined here provide the most direct view yet of hIAPP amyloid fibrils formed during T2D.


Asunto(s)
Amiloide/química , Microscopía por Crioelectrón , Polipéptido Amiloide de los Islotes Pancreáticos/química , Secuencia de Aminoácidos , Amiloide/aislamiento & purificación , Rojo Congo , Diabetes Mellitus Tipo 2/metabolismo , Genotipo , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Islotes Pancreáticos/química , Modelos Moleculares , Reacción en Cadena de la Polimerasa , Agregado de Proteínas , Conformación Proteica , Proteínas Recombinantes/química , Análisis de Secuencia de ADN , Coloración y Etiquetado
12.
bioRxiv ; 2021 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-33688654

RESUMEN

The SARS-CoV-2 Nucleoprotein (NCAP) functions in RNA packaging during viral replication and assembly. Computational analysis of its amino acid sequence reveals a central low-complexity domain (LCD) having sequence features akin to LCDs in other proteins known to function in liquid-liquid phase separation. Here we show that in the presence of viral RNA, NCAP, and also its LCD segment alone, form amyloid-like fibrils when undergoing liquid-liquid phase separation. Within the LCD we identified three 6-residue segments that drive amyloid fibril formation. We determined atomic structures for fibrils formed by each of the three identified segments. These structures informed our design of peptide inhibitors of NCAP fibril formation and liquid-liquid phase separation, suggesting a therapeutic route for Covid-19. ONE SENTENCE SUMMARY: Atomic structures of amyloid-driving peptide segments from SARS-CoV-2 Nucleoprotein inform the development of Covid-19 therapeutics.

13.
Elife ; 92020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31895037

RESUMEN

Seeding, in the context of amyloid disease, is the sequential transfer of pathogenic protein aggregates from cell-to-cell within affected tissues. The structure of pathogenic seeds provides the molecular basis and enables rapid conversion of soluble protein into fibrils. To date, there are no inhibitors that specifically target seeding of Parkinson's disease (PD)-associated α-synuclein (α-syn) fibrils, in part, due to lack of information of the structural properties of pathological seeds. Here we design small peptidic inhibitors based on the atomic structure of the core of α-syn fibrils. The inhibitors prevent α-syn aggregation in vitro and in cell culture models with binding affinities of 0.5 µM to α-syn fibril seeds. The inhibitors also show efficacy in preventing seeding by human patient-derived α-syn fibrils. Our results suggest that pathogenic seeds of α-syn contain steric zippers and suggest a therapeutic approach targeted at the spread and progression that may be applicable for PD and related synucleinopathies.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Agregado de Proteínas/fisiología , Agregación Patológica de Proteínas/fisiopatología , alfa-Sinucleína/metabolismo , Células HEK293 , Humanos
14.
Alzheimers Res Ther ; 11(1): 86, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31627745

RESUMEN

BACKGROUND: Repeated failure of drug candidates targeting Alzheimer's disease (AD) in clinical trials likely stems from a lack of understanding of the molecular mechanisms underlying AD pathogenesis. Recent research has highlighted synergistic interactions between aggregated amyloid-ß (Aß) and tau proteins in AD, but the molecular details of how these interactions drive AD pathology remain elusive and speculative. METHODS: Here, we test the hypothesis that Aß potentiates intracellular tau aggregation, and show that oligomeric Aß specifically exacerbates proteopathic seeding by tau. Using tau-biosensor cells, we show that treatment with sub-toxic concentrations of Aß oligomers, but not monomers or fibrils, "primes" cells, making them more susceptible to tau seeding. The treatment with Aß oligomers enhances intracellular tau aggregation in a dose-dependent manner when the cells are seeded with either recombinant or brain-derived tau fibrils, whereas little or no aggregation is observed in the absence of Aß-oligomer priming. RESULTS: Priming by Aß oligomers appears to be specific to tau, as α-synuclein seeding is unaffected by this treatment. Aß oligomer-enhanced tau seeding also occurs in primary mouse neurons and human neuroblastoma cells. Using fluorescently labeled tau seeds, we find that treatment with Aß oligomers significantly enhances the cellular uptake of tau seeds, whereas a known tau-uptake inhibitor blocks the effect of Aß on tau uptake. CONCLUSION: The ability of Aß to promote tau seeding suggests a specific and plausible mechanism by which extracellular Aß initiates a deleterious cascade that is unique to AD. These data suggest that the Aß-mediated potentiation of tau uptake into cells should also be taken into account when designing Aß-targeted therapeutics.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Técnicas Biosensibles , Encéfalo/patología , Línea Celular , Humanos , Neuronas/patología , Fragmentos de Péptidos/metabolismo
15.
Front Mol Neurosci ; 12: 268, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31787880

RESUMEN

Alzheimer's disease (AD) pathology is characterized by the aggregation of beta-amyloid (Aß) and tau in the form of amyloid plaques and neurofibrillary tangles in the brain. It has been found that a synergistic relationship between these two proteins may contribute to their roles in disease progression. However, how Aß and tau interact has not been fully characterized. Here, we analyze how tau seeding or aggregation is influenced by different Aß self-assemblies (fibrils and oligomers). Our cellular assays utilizing tau biosensor cells show that transduction of Aß oligomers into the cells greatly enhances seeded tau aggregation in a concentration-dependent manner. In contrast, transduced Aß fibrils slightly reduce tau seeding while untransduced Aß fibrils promote it. We also observe that the transduction of α-synuclein fibrils, another amyloid protein, has no effect on tau seeding. The enhancement of tau seeding by Aß oligomers was confirmed using tau fibril seeds derived from both recombinant tau and PS19 mouse brain extracts containing human tau. Our findings highlight the importance of considering the specific form and cellular location of Aß self-assembly when studying the relationship between Aß and tau in future AD therapeutic development.

16.
Elife ; 82019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31612856

RESUMEN

Alzheimer's disease (AD) pathology is characterized by plaques of amyloid beta (Aß) and neurofibrillary tangles of tau. Aß aggregation is thought to occur at early stages of the disease, and ultimately gives way to the formation of tau tangles which track with cognitive decline in humans. Here, we report the crystal structure of an Aß core segment determined by MicroED and in it, note characteristics of both fibrillar and oligomeric structure. Using this structure, we designed peptide-based inhibitors that reduce Aß aggregation and toxicity of already-aggregated species. Unexpectedly, we also found that these inhibitors reduce the efficiency of Aß-mediated tau aggregation, and moreover reduce aggregation and self-seeding of tau fibrils. The ability of these inhibitors to interfere with both Aß and tau seeds suggests these fibrils share a common epitope, and supports the hypothesis that cross-seeding is one mechanism by which amyloid is linked to tau aggregation and could promote cognitive decline.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Proteínas tau/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Cristalografía por Rayos X , Humanos , Estructura Molecular , Unión Proteica , Conformación Proteica
17.
Protein Sci ; 27(7): 1231-1242, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29453800

RESUMEN

The aggregation cascade of disease-related amyloidogenic proteins, terminating in insoluble amyloid fibrils, involves intermediate oligomeric states. The structural and biochemical details of these oligomers have been largely unknown. Here we report crystal structures of variants of the cytotoxic oligomer-forming segment residues 28-38 of the ALS-linked protein, SOD1. The crystal structures reveal three different architectures: corkscrew oligomeric structure, nontwisting curved sheet structure and a steric zipper proto-filament structure. Our work highlights the polymorphism of the segment 28-38 of SOD1 and identifies the molecular features of amyloidogenic entities.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Mutación , Superóxido Dismutasa-1/química , Superóxido Dismutasa-1/genética , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Pliegue de Proteína , Multimerización de Proteína , Estructura Secundaria de Proteína
18.
Nat Chem ; 10(12): 1213-1221, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30297750

RESUMEN

Inhibiting the interaction between amyloid-ß (Aß) and a neuronal cell surface receptor, LilrB2, has been suggested as a potential route for treating Alzheimer's disease. Supporting this approach, Alzheimer's-like symptoms are reduced in mouse models following genetic depletion of the LilrB2 homologue. In its pathogenic, oligomeric state, Aß binds to LilrB2, triggering a pathway to synaptic loss. Here we identify the LilrB2 binding moieties of Aß (16KLVFFA21) and identify its binding site on LilrB2 from a crystal structure of LilrB2 immunoglobulin domains D1D2 complexed to small molecules that mimic phenylalanine residues. In this structure, we observed two pockets that can accommodate the phenylalanine side chains of KLVFFA. These pockets were confirmed to be 16KLVFFA21 binding sites by mutagenesis. Rosetta docking revealed a plausible geometry for the Aß-LilrB2 complex and assisted with the structure-guided selection of small molecule inhibitors. These molecules inhibit Aß-LilrB2 interactions in vitro and on the cell surface and reduce Aß cytotoxicity, which suggests these inhibitors are potential therapeutic leads against Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/toxicidad , Diseño de Fármacos , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Glicoproteínas de Membrana/química , Ratones , Estructura Molecular , Neuronas/efectos de los fármacos , Receptores Inmunológicos/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
19.
Nat Chem ; 10(12): 1267, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30420778

RESUMEN

In the version of this Article originally published online, the upper right panel of Fig. 5a was mistakenly a repeat of the lower right panel. This has now been corrected in all versions of the Article.

20.
Nat Commun ; 9(1): 3609, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30190461

RESUMEN

α-Synuclein (aSyn) fibrillar polymorphs have distinct in vitro and in vivo seeding activities, contributing differently to synucleinopathies. Despite numerous prior attempts, how polymorphic aSyn fibrils differ in atomic structure remains elusive. Here, we present fibril polymorphs from the full-length recombinant human aSyn and their seeding capacity and cytotoxicity in vitro. By cryo-electron microscopy helical reconstruction, we determine the structures of the two predominant species, a rod and a twister, both at 3.7 Å resolution. Our atomic models reveal that both polymorphs share a kernel structure of a bent ß-arch, but differ in their inter-protofilament interfaces. Thus, different packing of the same kernel structure gives rise to distinct fibril polymorphs. Analyses of disease-related familial mutations suggest their potential contribution to the pathogenesis of synucleinopathies by altering population distribution of the fibril polymorphs. Drug design targeting amyloid fibrils in neurodegenerative diseases should consider the formation and distribution of concurrent fibril polymorphs.


Asunto(s)
alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Animales , Técnicas Biosensibles , Microscopía por Crioelectrón , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Células PC12/efectos de los fármacos , Conformación Proteica , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidad , Difracción de Rayos X , alfa-Sinucleína/genética , alfa-Sinucleína/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA