Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Nature ; 610(7933): 791-795, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36108674

RESUMEN

Parkinson's disease (PD) is the most common movement disorder, with resting tremor, rigidity, bradykinesia and postural instability being major symptoms1. Neuropathologically, it is characterized by the presence of abundant filamentous inclusions of α-synuclein in the form of Lewy bodies and Lewy neurites in some brain cells, including dopaminergic nerve cells of the substantia nigra2. PD is increasingly recognised as a multisystem disorder, with cognitive decline being one of its most common non-motor symptoms. Many patients with PD develop dementia more than 10 years after diagnosis3. PD dementia (PDD) is clinically and neuropathologically similar to dementia with Lewy bodies (DLB), which is diagnosed when cognitive impairment precedes parkinsonian motor signs or begins within one year from their onset4. In PDD, cognitive impairment develops in the setting of well-established PD. Besides PD and DLB, multiple system atrophy (MSA) is the third major synucleinopathy5. It is characterized by the presence of abundant filamentous α-synuclein inclusions in brain cells, especially oligodendrocytes (Papp-Lantos bodies). We previously reported the electron cryo-microscopy structures of two types of α-synuclein filament extracted from the brains of individuals with MSA6. Each filament type is made of two different protofilaments. Here we report that the cryo-electron microscopy structures of α-synuclein filaments from the brains of individuals with PD, PDD and DLB are made of a single protofilament (Lewy fold) that is markedly different from the protofilaments of MSA. These findings establish the existence of distinct molecular conformers of assembled α-synuclein in neurodegenerative disease.


Asunto(s)
Química Encefálica , Encéfalo , Microscopía por Crioelectrón , Enfermedad por Cuerpos de Lewy , alfa-Sinucleína , Humanos , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , alfa-Sinucleína/ultraestructura , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/ultraestructura , Enfermedad por Cuerpos de Lewy/patología , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/patología , Demencia/complicaciones , Demencia/patología
2.
Sci Rep ; 12(1): 351, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013421

RESUMEN

Parkinson's disease is a neurodegenerative disease characterized by the formation of neuronal inclusions of α-synuclein in patient brains. As the disease progresses, toxic α-synuclein aggregates transmit throughout the nervous system. No effective disease-modifying therapy has been established, and preventing α-synuclein aggregation is thought to be one of the most promising approaches to ameliorate the disease. In this study, we performed a two-step screening using the thioflavin T assay and a cell-based assay to identify α-synuclein aggregation inhibitors. The first screening, thioflavin T assay, allowed the identification of 30 molecules, among a total of 1262 FDA-approved small compounds, which showed inhibitory effects on α-synuclein fibrilization. In the second screening, a cell-based aggregation assay, seven out of these 30 candidates were found to prevent α-synuclein aggregation without causing substantial toxicity. Of the seven final candidates, tannic acid was the most promising compound. The robustness of our screening method was validated by a primary neuronal cell model and a Caenorhabditis elegans model, which demonstrated the effect of tannic acid against α-synuclein aggregation. In conclusion, our two-step screening system is a powerful method for the identification of α-synuclein aggregation inhibitors, and tannic acid is a promising candidate as a disease-modifying drug for Parkinson's disease.


Asunto(s)
Antiparkinsonianos/farmacología , Ensayos Analíticos de Alto Rendimiento , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Agregación Patológica de Proteínas , Taninos/farmacología , alfa-Sinucleína/metabolismo , Animales , Animales Modificados Genéticamente , Antiparkinsonianos/toxicidad , Benzotiazoles/química , Bioensayo , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Células HeLa , Humanos , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Agregado de Proteínas , Espectrometría de Fluorescencia , Taninos/toxicidad , alfa-Sinucleína/genética , alfa-Sinucleína/ultraestructura
3.
FEBS J ; 289(1): 215-230, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34268903

RESUMEN

Under certain cellular conditions, functional proteins undergo misfolding, leading to a transition into oligomers which precede the formation of amyloid fibrils. Misfolding proteins are associated with neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. While the importance of lipid membranes in misfolding and disease aetiology is broadly accepted, the influence of lipid membranes during therapeutic design has been largely overlooked. This study utilized a biophysical approach to provide mechanistic insights into the effects of two lipid membrane systems (anionic and zwitterionic) on the inhibition of amyloid-ß 40 and α-synuclein amyloid formation at the monomer, oligomer and fibril level. Large unilamellar vesicles (LUVs) were shown to increase fibrillization and largely decrease the effectiveness of two well-known polyphenol fibril inhibitors, (-)-epigallocatechin gallate (EGCG) and resveratrol; however, use of immunoblotting and ion mobility mass spectrometry revealed this occurs through varying mechanisms. Oligomeric populations in particular were differentially affected by LUVs in the presence of resveratrol, an elongation phase inhibitor, compared to EGCG, a nucleation targeted inhibitor. Ion mobility mass spectrometry showed EGCG interacts with or induces more compact forms of monomeric protein typical of off-pathway structures; however, binding is reduced in the presence of LUVs, likely due to partitioning in the membrane environment. Competing effects of the lipids and inhibitor, along with reduced inhibitor binding in the presence of LUVs, provide a mechanistic understanding of decreased inhibitor efficacy in a lipid environment. Together, this study highlights that amyloid inhibitor design may be misguided if effects of lipid membrane composition and architecture are not considered during development.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Amiloide/genética , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Amiloide/efectos de los fármacos , Amiloide/ultraestructura , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/ultraestructura , Proteínas Amiloidogénicas/antagonistas & inhibidores , Proteínas Amiloidogénicas/genética , Catequina/análogos & derivados , Catequina/farmacología , Humanos , Membrana Dobles de Lípidos/metabolismo , Lípidos de la Membrana/genética , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología , Fosfolípidos/biosíntesis , Fosfolípidos/genética , Polifenoles/farmacología , alfa-Sinucleína/ultraestructura
4.
Biochemistry ; 60(47): 3644-3658, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34730940

RESUMEN

The aggregation of α-synuclein (αSN) and increased oxidative stress leading to lipid peroxidation are pathological characteristics of Parkinson's disease (PD). Here, we report that aggregation of αSN in the presence of lipid peroxidation products 4-hydroxy-2-nonenal (HNE) and 4-oxo-2-nonenal (ONE) increases the stability and the yield of αSN oligomers (αSO). Further, we show that ONE is more efficient than HNE at inducing αSO. In addition, we demonstrate that the two αSO differ in both size and shape. ONE-αSO are smaller in size than HNE-αSO, except when they are formed at a high molar excess of aldehyde. In both monomeric and oligomeric αSN, His50 is the main target of HNE modification, and HNE-induced oligomerization is severely retarded in the mutant His50Ala αSN. In contrast, ONE-induced aggregation of His50Ala αSN occurs readily, demonstrating the different pathways for inducing αSN aggregation by HNE and ONE. Our results show different morphologies of the HNE-treated and ONE-treated αSO and different roles of His50 in their modification of αSN, but we also observe structural similarities between these αSO and the non-treated αSO, e.g., flexible C-terminus, a folded core composed of the N-terminal and NAC region. Furthermore, HNE-αSO show a similar deuterium uptake as a previously characterized oligomer formed by non-treated αSO, suggesting that the backbone conformational dynamics of their folded cores resemble one another.


Asunto(s)
Aldehídos/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Aldehídos/química , Línea Celular Tumoral , Humanos , Peroxidación de Lípido , Resonancia Magnética Nuclear Biomolecular , Agregado de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Dispersión del Ángulo Pequeño , Difracción de Rayos X , alfa-Sinucleína/química , alfa-Sinucleína/aislamiento & purificación , alfa-Sinucleína/ultraestructura
5.
Biomolecules ; 11(10)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34680054

RESUMEN

Abnormal accumulation of aggregated α-synuclein (α-Syn) is seen in a variety of neurodegenerative diseases, including Parkinson's disease (PD), multiple system atrophy (MSA), dementia with Lewy body (DLB), Parkinson's disease dementia (PDD), and even subsets of Alzheimer's disease (AD) showing Lewy-body-like pathology. These synucleinopathies exhibit differences in their clinical and pathological representations, reminiscent of prion disorders. Emerging evidence suggests that α-Syn self-assembles and polymerizes into conformationally diverse polymorphs in vitro and in vivo, similar to prions. These α-Syn polymorphs arising from the same precursor protein may exhibit strain-specific biochemical properties and the ability to induce distinct pathological phenotypes upon their inoculation in animal models. In this review, we discuss clinical and pathological variability in synucleinopathies and several aspects of α-Syn fibril polymorphism, including the existence of high-resolution molecular structures and brain-derived strains. The current review sheds light on the recent advances in delineating the structure-pathogenic relationship of α-Syn and how diverse α-Syn molecular polymorphs contribute to the existing clinical heterogeneity in synucleinopathies.


Asunto(s)
Amiloide/genética , Encéfalo/metabolismo , Agregado de Proteínas/genética , alfa-Sinucleína/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Amiloide/ultraestructura , Encéfalo/patología , Humanos , Cuerpos de Lewy/genética , Cuerpos de Lewy/patología , Atrofia de Múltiples Sistemas/genética , Atrofia de Múltiples Sistemas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Enfermedades por Prión/genética , Enfermedades por Prión/patología , alfa-Sinucleína/ultraestructura
6.
Biochemistry ; 60(48): 3676-3696, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34431665

RESUMEN

Liquid-liquid phase separation (LLPS) is a crucial phenomenon for the formation of functional membraneless organelles. However, LLPS is also responsible for protein aggregation in various neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease (PD). Recently, several reports, including ours, have shown that α-synuclein (α-Syn) undergoes LLPS and a subsequent liquid-to-solid phase transition, which leads to amyloid fibril formation. However, how the environmental (and experimental) parameters modulate the α-Syn LLPS remains elusive. Here, we show that in vitro α-Syn LLPS is strongly dependent on the presence of salts, which allows charge neutralization at both terminal segments of protein and therefore promotes hydrophobic interactions supportive for LLPS. Using various purification methods and experimental conditions, we showed, depending upon conditions, α-Syn undergoes either spontaneous (instantaneous) or delayed LLPS. Furthermore, we delineate that the kinetics of liquid droplet formation (i.e., the critical concentration and critical time) is relative and can be modulated by the salt/counterion concentration, pH, presence of surface, PD-associated multivalent cations, and N-terminal acetylation, which are all known to regulate α-Syn aggregation in vitro. Together, our observations suggest that α-Syn LLPS and subsequent liquid-to-solid phase transition could be pathological, which can be triggered only under disease-associated conditions (high critical concentration and/or conditions promoting α-Syn self-assembly). This study will significantly improve our understanding of the molecular mechanisms of α-Syn LLPS and the liquid-to-solid transition.


Asunto(s)
Amiloide/química , Agregación Patológica de Proteínas/genética , alfa-Sinucleína/química , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Amiloide/genética , Amiloide/ultraestructura , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Transición de Fase , Agregación Patológica de Proteínas/patología , alfa-Sinucleína/genética , alfa-Sinucleína/ultraestructura
7.
Int J Mol Sci ; 22(11)2021 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-34199513

RESUMEN

Intrinsic disorder is a natural feature of polypeptide chains, resulting in the lack of a defined three-dimensional structure. Conformational changes in intrinsically disordered regions of a protein lead to unstable ß-sheet enriched intermediates, which are stabilized by intermolecular interactions with other ß-sheet enriched molecules, producing stable proteinaceous aggregates. Upon misfolding, several pathways may be undertaken depending on the composition of the amino acidic string and the surrounding environment, leading to different structures. Accumulating evidence is suggesting that the conformational state of a protein may initiate signalling pathways involved both in pathology and physiology. In this review, we will summarize the heterogeneity of structures that are produced from intrinsically disordered protein domains and highlight the routes that lead to the formation of physiological liquid droplets as well as pathogenic aggregates. The most common proteins found in aggregates in neurodegenerative diseases and their structural variability will be addressed. We will further evaluate the clinical relevance and future applications of the study of the structural heterogeneity of protein aggregates, which may aid the understanding of the phenotypic diversity observed in neurodegenerative disorders.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Agregado de Proteínas/genética , Agregación Patológica de Proteínas/genética , Conformación Proteica en Lámina beta , Amiloide/genética , Amiloide/ultraestructura , Humanos , Proteínas Intrínsecamente Desordenadas , Enfermedades Neurodegenerativas/patología , alfa-Sinucleína/genética , alfa-Sinucleína/ultraestructura , Proteínas tau/genética , Proteínas tau/ultraestructura
8.
Int J Biol Macromol ; 182: 1278-1291, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33991558

RESUMEN

The aggregation of α-synuclein is linked to neurological disorders, and of these, Parkinson's disease (PD) is among the most widely studied. In this background, we have investigated here the effects of three α, ß-unsaturated carbonyl based plant metabolites, daidzein, fisetin and scopoletin on α-Syn aggregation. The ThT and light scattering kinetics studies establish that these compounds have ability to inhibit α-Syn fibrillation to different extents; this is confirmed by TEM studies. It is pertinent to note here that daidzein and scopoletin have been predicted to be able to cross the blood brain barrier. ANS binding assays demonstrate that the compounds interfere in the hydrophobic interactions. The tyrosine quenching, molecular docking and MD simulation studies showed that the compounds bind with α-Syn and provide structural rigidity which delays onset of structural transitions, which is confirmed by CD spectroscopy. The results obtained here throw light on the mechanisms underlying inhibition of α-Syn fibrillation by these compounds. Thus, the current work has significant therapeutic implications for identifying plant based potent therapeutic molecules for PD and other synucleinopathies, an area which needs extensive exploration.


Asunto(s)
Flavonoles/farmacología , Isoflavonas/farmacología , Metaboloma , Agregado de Proteínas/efectos de los fármacos , Escopoletina/farmacología , alfa-Sinucleína/metabolismo , Productos Biológicos/química , Productos Biológicos/farmacología , Línea Celular , Flavonoles/química , Fluorescencia , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Isoflavonas/química , Cinética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Conformación Proteica , Escopoletina/química , Tirosina/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/ultraestructura
9.
J Biol Chem ; 296: 100788, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019875

RESUMEN

The intrinsically disordered human protein α-synuclein (αSN) can self-associate into oligomers and amyloid fibrils. Several lines of evidence suggest that oligomeric αSN is cytotoxic, making it important to devise strategies to either prevent oligomer formation and/or inhibit the ensuing toxicity. (-)-epigallocatechin gallate (EGCG) has emerged as a molecular modulator of αSN self-assembly, as it reduces the flexibility of the C-terminal region of αSN in the oligomer and inhibits the oligomer's ability to perturb phospholipid membranes and induce cell death. However, a detailed structural and kinetic characterization of this interaction is still lacking. Here, we use liquid-state NMR spectroscopy to investigate how EGCG interacts with monomeric and oligomeric forms of αSN. We find that EGCG can bind to all parts of monomeric αSN but exhibits highest affinity for the N-terminal region. Monomeric αSN binds ∼54 molecules of EGCG in total during oligomerization. Furthermore, kinetic data suggest that EGCG dimerization is coupled with the αSN association reaction. In contrast, preformed oligomers only bind ∼7 EGCG molecules per protomer, in agreement with the more compact nature of the oligomer compared with the natively unfolded monomer. In previously conducted cell assays, as little as 0.36 EGCG per αSN reduce oligomer toxicity by 50%. Our study thus demonstrates that αSN cytotoxicity can be inhibited by small molecules at concentrations at least an order of magnitude below full binding capacity. We speculate this is due to cooperative binding of protein-stabilized EGCG dimers, which in turn implies synergy between protein association and EGCG dimerization.


Asunto(s)
Catequina/análogos & derivados , alfa-Sinucleína/metabolismo , Catequina/farmacología , Humanos , Agregado de Proteínas/efectos de los fármacos , Unión Proteica , Conformación Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , alfa-Sinucleína/química , alfa-Sinucleína/ultraestructura
10.
J Biol Chem ; 296: 100737, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33933456

RESUMEN

Synucleinopathies are neurodegenerative diseases characterized by the presence of intracellular deposits containing the protein alpha-synuclein (aSYN) within patients' brains. It has been shown that aSYN can form structurally distinct fibrillar assemblies, also termed polymorphs. We previously showed that distinct aSYN polymorphs assembled in vitro, named fibrils, ribbons, and fibrils 91, differentially bind to and seed the aggregation of endogenous aSYN in neuronal cells, which suggests that distinct synucleinopathies may arise from aSYN polymorphs. In order to better understand the differential interactions of aSYN polymorphs with their partner proteins, we mapped aSYN polymorphs surfaces. We used limited proteolysis, hydrogen-deuterium exchange, and differential antibody accessibility to identify amino acids on their surfaces. We showed that the aSYN C-terminal region spanning residues 94 to 140 exhibited similarly high solvent accessibility in these three polymorphs. However, the N-terminal amino acid residues 1 to 38 of fibrils were exposed to the solvent, while only residues 1 to 18 within fibrils 91 were exposed, and no N-terminal residues within ribbons were solvent-exposed. It is likely that these differences in surface accessibility contribute to the differential binding of distinct aSYN polymorphs to partner proteins. We thus posit that the polypeptides exposed on the surface of distinct aSYN fibrillar polymorphs are comparable to fingerprints. Our findings have diagnostic and therapeutic potential, particularly in the prion-like propagation of fibrillar aSYN, as they can facilitate the design of ligands that specifically bind and distinguish between fibrillar polymorphs.


Asunto(s)
Sinucleinopatías/metabolismo , alfa-Sinucleína/metabolismo , Humanos , Conformación Proteica , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Solventes/metabolismo , Sinucleinopatías/patología , alfa-Sinucleína/química , alfa-Sinucleína/ultraestructura
11.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33903234

RESUMEN

Amyloid fibril formation of α-synuclein (αS) is associated with multiple neurodegenerative diseases, including Parkinson's disease (PD). Growing evidence suggests that progression of PD is linked to cell-to-cell propagation of αS fibrils, which leads to seeding of endogenous intrinsically disordered monomer via templated elongation and secondary nucleation. A molecular understanding of the seeding mechanism and driving interactions is crucial to inhibit progression of amyloid formation. Here, using relaxation-based solution NMR experiments designed to probe large complexes, we probe weak interactions of intrinsically disordered acetylated-αS (Ac-αS) monomers with seeding-competent Ac-αS fibrils and seeding-incompetent off-pathway oligomers to identify Ac-αS monomer residues at the binding interface. Under conditions that favor fibril elongation, we determine that the first 11 N-terminal residues on the monomer form a common binding site for both fibrils and off-pathway oligomers. Additionally, the presence of off-pathway oligomers within a fibril seeding environment suppresses seeded amyloid formation, as observed through thioflavin-T fluorescence experiments. This highlights that off-pathway αS oligomers can act as an auto-inhibitor against αS fibril elongation. Based on these data taken together with previous results, we propose a model in which Ac-αS monomer recruitment to the fibril is driven by interactions between the intrinsically disordered monomer N terminus and the intrinsically disordered flanking regions (IDR) on the fibril surface. We suggest that this monomer recruitment may play a role in the elongation of amyloid fibrils and highlight the potential of the IDRs of the fibril as important therapeutic targets against seeded amyloid formation.


Asunto(s)
Amiloide/ultraestructura , Proteínas Intrínsecamente Desordenadas/ultraestructura , Enfermedad de Parkinson/genética , alfa-Sinucleína/ultraestructura , Amiloide/química , Amiloide/genética , Benzotiazoles/química , Benzotiazoles/metabolismo , Sitios de Unión/genética , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/genética , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Resonancia Magnética Nuclear Biomolecular , Enfermedad de Parkinson/patología , alfa-Sinucleína/química , alfa-Sinucleína/genética
12.
Cells ; 10(1)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445653

RESUMEN

We investigated α-synuclein's (αSyn) seeding activity in tissue from the brain and enteric nervous system. Specifically, we assessed the seeding propensity of pathogenic αSyn in formalin-fixed tissue from the gastric cardia and five brain regions of 29 individuals (12 Parkinson's disease, 8 incidental Lewy body disease, 9 controls) using a protein misfolding cyclic amplification assay. The structural characteristics of the resultant αSyn assemblies were determined by limited proteolysis and transmission electron microscopy. We show that fixed tissue from Parkinson's disease (PD) and incidental Lewy body disease (ILBD) seeds the aggregation of monomeric αSyn into fibrillar assemblies. Significant variations in the characteristics of fibrillar assemblies derived from different regions even within the same individual were observed. This finding suggests that fixation stabilizes seeds with an otherwise limited seeding propensity, that yield assemblies with different intrinsic structures (i.e., strains). The lag phase preceding fibril assembly for patients ≥80 was significantly shorter than in other age groups, suggesting the existence of increased numbers of seeds or a higher seeding potential of pathogenic αSyn with time. Seeding activity did not diminish in late-stage disease. No statistically significant difference in the seeding efficiency of specific regions was found, nor was there a relationship between seeding efficiency and the load of pathogenic αSyn in a particular region at a given neuropathological stage.


Asunto(s)
Tronco Encefálico/patología , Sistema Nervioso Entérico/patología , Bulbo Olfatorio/patología , Enfermedad de Parkinson/patología , Fijación del Tejido , alfa-Sinucleína/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Formaldehído , Humanos , Enfermedad por Cuerpos de Lewy/patología , Masculino , Persona de Mediana Edad , Neuritas/metabolismo , Neuritas/patología , Pliegue de Proteína , Proteolisis , alfa-Sinucleína/ultraestructura
13.
Int J Mol Sci ; 22(2)2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33477465

RESUMEN

A number of neurodegenerative diseases including prion diseases, tauopathies and synucleinopathies exhibit multiple clinical phenotypes. A diversity of clinical phenotypes has been attributed to the ability of amyloidogenic proteins associated with a particular disease to acquire multiple, conformationally distinct, self-replicating states referred to as strains. Structural diversity of strains formed by tau, α-synuclein or prion proteins has been well documented. However, the question how different strains formed by the same protein elicit different clinical phenotypes remains poorly understood. The current article reviews emerging evidence suggesting that posttranslational modifications are important players in defining strain-specific structures and disease phenotypes. This article put forward a new hypothesis referred to as substrate selection hypothesis, according to which individual strains selectively recruit protein isoforms with a subset of posttranslational modifications that fit into strain-specific structures. Moreover, it is proposed that as a result of selective recruitment, strain-specific patterns of posttranslational modifications are formed, giving rise to unique disease phenotypes. Future studies should define whether cell-, region- and age-specific differences in metabolism of posttranslational modifications play a causative role in dictating strain identity and structural diversity of strains of sporadic origin.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Proteínas Priónicas/ultraestructura , alfa-Sinucleína/ultraestructura , Proteínas tau/ultraestructura , Humanos , Enfermedades Neurodegenerativas/patología , Fenotipo , Proteínas Priónicas/genética , Conformación Proteica , Procesamiento Proteico-Postraduccional/genética , Especificidad por Sustrato , Sinucleinopatías/genética , Sinucleinopatías/patología , Tauopatías/genética , Tauopatías/patología , alfa-Sinucleína/genética , Proteínas tau/genética
14.
Cells ; 11(1)2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-35011649

RESUMEN

Parkinson's disease (PD) and multiple system atrophy (MSA) are caused by two distinct strains of disease-associated α-synuclein (αSynD). Recently, we have shown that olfactory mucosa (OM) samples of patients with PD and MSA can seed the aggregation of recombinant α-synuclein by means of Real-Time Quaking-Induced Conversion (αSyn_RT-QuIC). Remarkably, the biochemical and morphological properties of the final α-synuclein aggregates significantly differed between PD and MSA seeded samples. Here, these aggregates were given to neuron-like differentiated SH-SY5Y cells and distinct inflammatory responses were observed. To deepen whether the morphological features of α-synuclein aggregates were responsible for this variable SH-SY5Y inflammatory response, we generated three biochemically and morphologically distinct α-synuclein aggregates starting from recombinant α-synuclein that were used to seed αSyn_RT-QuIC reaction; the final reaction products were used to stimulate SH-SY5Y cells. Our study showed that, in contrast to OM samples of PD and MSA patients, the artificial aggregates did not transfer their distinctive features to the αSyn_RT-QuIC products and the latter induced analogous inflammatory responses in cells. Thus, the natural composition of the αSynD strains but also other specific factors in OM tissue can substantially modulate the biochemical, morphological and inflammatory features of the αSyn_RT-QuIC products.


Asunto(s)
Inflamación/patología , Atrofia de Múltiples Sistemas/metabolismo , Atrofia de Múltiples Sistemas/patología , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , Neuroblastoma/patología , Agregado de Proteínas , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/ultraestructura
15.
Mol Neurobiol ; 58(2): 867-876, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33048264

RESUMEN

Cell-to-cell transfer of α-synuclein (αS) is increasingly thought to play an important role in propagation of αS pathology, but mechanisms responsible for formation of initial αS seeds and factors facilitating their propagation remain unclear. We previously demonstrated that αS aggregates are formed rapidly in apoptotic neurons and that interaction between cytoplasmic αS and proaggregant nuclear factors generates seed-competent αS. We also provided initial evidence that histones have proaggregant properties. Since histones are released from cells undergoing apoptosis or cell stress, we hypothesized that internalization of histones into αS expressing cells could lead to intracellular αS aggregation. Here using mCherry-tagged histone, we show that nuclear extracts from apoptotic cells can induce intracellular αS inclusions after uptake into susceptible cells, while extracts from non-apoptotic cells did not. We also demonstrate that nuclear extracts from apoptotic cells contained histone-immunoreactive amyloid fibrils. Moreover, recombinant histone-derived amyloid fibrils are able to induce αS aggregation in cellular and animal models. Induction of αS aggregation by histone amyloid fibrils is associated with endocytosis-mediated rupture of lysosomes, and this effect can be enhanced in cells with chemically induced lysosomal membrane defects. These studies provide initial descriptions of the contribution of histone amyloid fibrils to αS aggregation.


Asunto(s)
Amiloide/metabolismo , Apoptosis , Histonas/metabolismo , Neuronas/metabolismo , Agregado de Proteínas , alfa-Sinucleína/metabolismo , Animales , Extractos Celulares , Línea Celular , Núcleo Celular/metabolismo , Endocitosis , Femenino , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Neuronas/ultraestructura , Solubilidad , alfa-Sinucleína/ultraestructura
16.
J Mol Biol ; 432(24): 166689, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33211011

RESUMEN

Heterogeneous aggregates of the human protein α-synuclein (αSyn) are abundantly found in Lewy body inclusions of Parkinson's disease patients. While structural information on classical αSyn amyloid fibrils is available, little is known about the conformational properties of disease-relevant, non-canonical aggregates. Here, we analyze the structural and dynamic properties of megadalton-sized dityrosine adducts of αSyn that form in the presence of reactive oxygen species and cytochrome c, a proapoptotic peroxidase that is released from mitochondria during sustained oxidative stress. In contrast to canonical cross-ß amyloids, these aggregates retain high degrees of internal dynamics, which enables their characterization by solution-state NMR spectroscopy. We find that intermolecular dityrosine crosslinks restrict αSyn motions only locally whereas large segments of concatenated molecules remain flexible and disordered. Indistinguishable aggregates form in crowded in vitro solutions and in complex environments of mammalian cell lysates, where relative amounts of free reactive oxygen species, rather than cytochrome c, are rate limiting. We further establish that dityrosine adducts inhibit classical amyloid formation by maintaining αSyn in its monomeric form and that they are non-cytotoxic despite retaining basic membrane-binding properties. Our results suggest that oxidative αSyn aggregation scavenges cytochrome c's activity into the formation of amorphous, high molecular-weight structures that may contribute to the structural diversity of Lewy body deposits.


Asunto(s)
Amiloide/genética , Enfermedad de Parkinson/genética , Tirosina/análogos & derivados , alfa-Sinucleína/genética , Amiloide/química , Amiloide/ultraestructura , Péptidos beta-Amiloides/genética , Citocromos c/genética , Humanos , Espectroscopía de Resonancia Magnética , Mitocondrias/genética , Mitocondrias/metabolismo , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Estrés Oxidativo/genética , Enfermedad de Parkinson/patología , Agregado de Proteínas/genética , Conformación Proteica , Especies Reactivas de Oxígeno/metabolismo , Tirosina/química , Tirosina/genética , alfa-Sinucleína/ultraestructura
17.
Nature ; 585(7825): 464-469, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32461689

RESUMEN

Synucleinopathies, which include multiple system atrophy (MSA), Parkinson's disease, Parkinson's disease with dementia and dementia with Lewy bodies (DLB), are human neurodegenerative diseases1. Existing treatments are at best symptomatic. These diseases are characterized by the presence of, and believed to be caused by the formation of, filamentous inclusions of α-synuclein in brain cells2,3. However, the structures of α-synuclein filaments from the human brain are unknown. Here, using cryo-electron microscopy, we show that α-synuclein inclusions from the brains of individuals with MSA are made of two types of filament, each of which consists of two different protofilaments. In each type of filament, non-proteinaceous molecules are present at the interface of the two protofilaments. Using two-dimensional class averaging, we show that α-synuclein filaments from the brains of individuals with MSA differ from those of individuals with DLB, which suggests that distinct conformers or strains characterize specific synucleinopathies. As is the case with tau assemblies4-9, the structures of α-synuclein filaments extracted from the brains of individuals with MSA differ from those formed in vitro using recombinant proteins, which has implications for understanding the mechanisms of aggregate propagation and neurodegeneration in the human brain. These findings have diagnostic and potential therapeutic relevance, especially because of the unmet clinical need to be able to image filamentous α-synuclein inclusions in the human brain.


Asunto(s)
Encéfalo/metabolismo , Microscopía por Crioelectrón , Cuerpos de Inclusión/química , Cuerpos de Inclusión/ultraestructura , Atrofia de Múltiples Sistemas/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/ultraestructura , Encéfalo/patología , Encéfalo/ultraestructura , Humanos , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Modelos Moleculares , Atrofia de Múltiples Sistemas/diagnóstico , Atrofia de Múltiples Sistemas/patología , Atrofia de Múltiples Sistemas/terapia , Pliegue de Proteína , Putamen/metabolismo , Putamen/ultraestructura , alfa-Sinucleína/metabolismo
18.
Nat Commun ; 11(1): 2643, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457390

RESUMEN

Amyloid aggregation of α-synuclein (α-syn) is closely associated with Parkinson's disease (PD) and other synucleinopathies. Several single amino-acid mutations (e.g. E46K) of α-syn have been identified causative to the early onset of familial PD. Here, we report the cryo-EM structure of an α-syn fibril formed by N-terminally acetylated E46K mutant α-syn (Ac-E46K). The fibril structure represents a distinct fold of α-syn, which demonstrates that the E46K mutation breaks the electrostatic interactions in the wild type (WT) α-syn fibril and thus triggers the rearrangement of the overall structure. Furthermore, we show that the Ac-E46K fibril is less resistant to harsh conditions and protease cleavage, and more prone to be fragmented with an enhanced seeding capability than that of the WT fibril. Our work provides a structural view to the severe pathology of the PD familial mutation E46K of α-syn and highlights the importance of electrostatic interactions in defining the fibril polymorphs.


Asunto(s)
Proteínas Mutantes/química , Proteínas Mutantes/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética , Acetilación , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Amiloide/química , Amiloide/genética , Amiloide/ultraestructura , Microscopía por Crioelectrón , Humanos , Microscopía de Fuerza Atómica , Modelos Moleculares , Proteínas Mutantes/ultraestructura , Mutación Missense , Conformación Proteica , Estabilidad Proteica , Electricidad Estática , alfa-Sinucleína/ultraestructura
19.
J Mol Biol ; 432(2): 585-596, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31518613

RESUMEN

Aggregation of amyloidogenic proteins is an abnormal biological process implicated in neurodegenerative disorders. Whereas the aggregation process of amyloid-forming proteins has been studied extensively, the mechanism of aggregate removal is poorly understood. We recently demonstrated that proteasomes could fragment filamentous aggregates into smaller entities, restricting aggregate size [1]. Here, we show in vitro that UBE2W can modify the N-terminus of both α-synuclein and a tau tetra-repeat domain with a single ubiquitin. We demonstrate that an engineered N-terminal ubiquitin modification changes the aggregation process of both proteins, resulting in the formation of structurally distinct aggregates. Single-molecule approaches further reveal that the proteasome can target soluble oligomers assembled from ubiquitin-modified proteins independently of its peptidase activity, consistent with our recently reported fibril-fragmenting activity. Based on these results, we propose that proteasomes are able to target oligomers assembled from N-terminally ubiquitinated proteins. Our data suggest a possible disassembly mechanism by which N-terminal ubiquitination and the proteasome may together impede aggregate formation.


Asunto(s)
Proteínas Amiloidogénicas/genética , Enfermedades Neurodegenerativas/genética , Enzimas Ubiquitina-Conjugadoras/genética , alfa-Sinucleína/genética , Proteínas tau/genética , Proteínas Amiloidogénicas/ultraestructura , Citoplasma/genética , Citoplasma/ultraestructura , Holoenzimas/genética , Holoenzimas/ultraestructura , Humanos , Enfermedades Neurodegenerativas/patología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/ultraestructura , Agregación Patológica de Proteínas/genética , Dominios Proteicos , Multimerización de Proteína , Ubiquitina/genética , Enzimas Ubiquitina-Conjugadoras/ultraestructura , Ubiquitinación/genética , alfa-Sinucleína/ultraestructura , Proteínas tau/ultraestructura
20.
Biochim Biophys Acta Proteins Proteom ; 1868(1): 140298, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676453

RESUMEN

The misfolding and aggregation of alpha-synuclein (aSyn) are thought to be central events in synucleinopathies. The physiological function of aSyn has been related to vesicle binding and trafficking, but the precise molecular mechanisms leading to aSyn pathogenicity are still obscure. In cell models, aSyn does not readily aggregate, even upon overexpression. Therefore, cellular models that enable the study of aSyn aggregation are essential tools for our understanding of the molecular mechanisms that govern such processes. Here, we investigated the structural features of SynT, an artificial variant of aSyn that has been widely used as a model of aggregation in mammalian cell systems, since it is more prone to aggregation than aSyn. Using Nuclear Magnetic Resonance (NMR) spectroscopy we performed a detailed structural characterization of SynT through a systematic comparison with normal, unmodified aSyn. Interestingly, we found that the conformations adopted by SynT resemble those described for the unmodified protein, demonstrating the usefulness of SynT as a model for aSyn aggregation. However, subtle differences were observed at the N-terminal region involving transient intra and/or intermolecular interactions that are known to regulate aSyn aggregation. Importantly, our results indicate that disturbances in the N-terminal region of SynT, and the consequent decrease in membrane binding of the modified protein, might contribute to the observed aggregation behavior of aSyn, and validate the use of SynT, one of the few models of aSyn aggregation in cultured cells.


Asunto(s)
Sinucleinopatías , alfa-Sinucleína/química , Línea Celular Tumoral , Escherichia coli/genética , Humanos , Microscopía Electrónica de Transmisión , Agregación Patológica de Proteínas , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , alfa-Sinucleína/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...