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








Base de dados
Intervalo de ano de publicação
1.
Acc Chem Res ; 56(12): 1395-1405, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37071750

RESUMO

The aberrant misfolding and aggregation of peptides and proteins into amyloid aggregates occurs in over 50 largely incurable protein misfolding diseases. These pathologies include Alzheimer's and Parkinson's diseases, which are global medical emergencies owing to their prevalence in increasingly aging populations worldwide. Although the presence of mature amyloid aggregates is a hallmark of such neurodegenerative diseases, misfolded protein oligomers are increasingly recognized as of central importance in the pathogenesis of many of these maladies. These oligomers are small, diffusible species that can form as intermediates in the amyloid fibril formation process or be released by mature fibrils after they are formed. They have been closely associated with the induction of neuronal dysfunction and cell death. It has proven rather challenging to study these oligomeric species because of their short lifetimes, low concentrations, extensive structural heterogeneity, and challenges associated with producing stable, homogeneous, and reproducible populations. Despite these difficulties, investigators have developed protocols to produce kinetically, chemically, or structurally stabilized homogeneous populations of protein misfolded oligomers from several amyloidogenic peptides and proteins at experimentally ameneable concentrations. Furthermore, procedures have been established to produce morphologically similar but structurally distinct oligomers from the same protein sequence that are either toxic or nontoxic to cells. These tools offer unique opportunities to identify and investigate the structural determinants of oligomer toxicity by a close comparative inspection of their structures and the mechanisms of action through which they cause cell dysfunction.This Account reviews multidisciplinary results, including from our own groups, obtained by combining chemistry, physics, biochemistry, cell biology, and animal models for pairs of toxic and nontoxic oligomers. We describe oligomers comprised of the amyloid-ß peptide, which underlie Alzheimer's disease, and α-synuclein, which are associated with Parkinson's disease and other related neurodegenerative pathologies, collectively known as synucleinopathies. Furthermore, we also discuss oligomers formed by the 91-residue N-terminal domain of [NiFe]-hydrogenase maturation factor from E. coli, which we use as a model non-disease-related protein, and by an amyloid stretch of Sup35 prion protein from yeast. These oligomeric pairs have become highly useful experimental tools for studying the molecular determinants of toxicity characteristic of protein misfolding diseases. Key properties have been identified that differentiate toxic from nontoxic oligomers in their ability to induce cellular dysfunction. These characteristics include solvent-exposed hydrophobic regions, interactions with membranes, insertion into lipid bilayers, and disruption of plasma membrane integrity. By using these properties, it has been possible to rationalize in model systems the responses to pairs of toxic and nontoxic oligomers. Collectively, these studies provide guidance for the development of efficacious therapeutic strategies to target rationally the cytotoxicity of misfolded protein oligomers in neurodegenerative conditions.


Assuntos
Doença de Alzheimer , Deficiências na Proteostase , Animais , Escherichia coli/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Amiloide/química
2.
Methods Mol Biol ; 2551: 379-394, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36310216

RESUMO

Amyloid protein aggregation is widely involved in a number of neurodegenerative diseases for which novel therapeutic and diagnostic strategies are still needed. Owing to the complex and heterogeneous nature of the aggregated species responsible for toxicity in these disorders, a detailed characterization of the interaction of molecules of interest with the amyloid aggregates is a challenging endeavor. Here, we present the experimental and analytical steps of a protocol which combines dual-color fluorescence cross-correlation spectroscopy and dual-color single-particle fluorescence spectroscopy to quantify the binding affinity and stoichiometry of an inhibitor of α-synuclein amyloid aggregation. This approach allows studying the interaction in detail and through two independent analytical methods, thus yielding a remarkably robust tool that could be extended to investigating the interaction of molecules of interest to other pathogenic protein aggregates as well as multi-ligand/multi-receptor complexes.


Assuntos
Doenças Neurodegenerativas , Agregados Proteicos , Humanos , Espectrometria de Fluorescência , alfa-Sinucleína/metabolismo , Amiloide/química , Imagem Individual de Molécula , Doenças Neurodegenerativas/metabolismo
3.
Nat Commun ; 13(1): 4586, 2022 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-35933508

RESUMO

Amyloid aggregation of α-synuclein (αS) is the hallmark of Parkinson's disease and other synucleinopathies. Recently, Tau protein, generally associated with Alzheimer's disease, has been linked to αS pathology and observed to co-localize in αS-rich disease inclusions, although the molecular mechanisms for the co-aggregation of both proteins remain elusive. We report here that αS phase-separates into liquid condensates by electrostatic complex coacervation with positively charged polypeptides such as Tau. Condensates undergo either fast gelation or coalescence followed by slow amyloid aggregation depending on the affinity of αS for the poly-cation and the rate of valence exhaustion of the condensate network. By combining a set of advanced biophysical techniques, we have been able to characterize αS/Tau liquid-liquid phase separation and identified key factors that lead to the formation of hetero-aggregates containing both proteins in the interior of the liquid protein condensates.


Assuntos
Sinucleinopatias , alfa-Sinucleína , Amiloide/metabolismo , Humanos , Eletricidade Estática , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
4.
Cell Mol Life Sci ; 79(3): 174, 2022 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-35244787

RESUMO

Protein misfolding is a general hallmark of protein deposition diseases, such as Alzheimer's disease or Parkinson's disease, in which different types of aggregated species (oligomers, protofibrils and fibrils) are generated by the cells. Despite widespread interest, the relationship between oligomers and fibrils in the aggregation process and spreading remains elusive. A large variety of experimental evidences supported the idea that soluble oligomeric species of different proteins might be more toxic than the larger fibrillar forms. Furthermore, the lack of correlation between the presence of the typical pathological inclusions and disease sustained this debate. However, recent data show that the ß-sheet core of the α-Synuclein (αSyn) fibrils is unable to establish persistent interactions with the lipid bilayers, but they can release oligomeric species responsible for an immediate dysfunction of the recipient neurons. Reversibly, such oligomeric species could also contribute to pathogenesis via neuron-to-neuron spreading by their direct cell-to-cell transfer or by generating new fibrils, following their neuronal uptake. In this Review, we discuss the various mechanisms of cellular dysfunction caused by αSyn, including oligomer toxicity, fibril toxicity and fibril spreading.


Assuntos
Amiloide/metabolismo , Sinucleinopatias/patologia , alfa-Sinucleína/metabolismo , Amiloide/toxicidade , Humanos , Corpos de Lewy/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Agregados Proteicos , Dobramento de Proteína , Sinucleinopatias/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/genética
5.
Molecules ; 27(4)2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35209093

RESUMO

A wide variety of oligomeric structures are formed during the aggregation of proteins associated with neurodegenerative diseases. Such soluble oligomers are believed to be key toxic species in the related disorders; therefore, identification of the structural determinants of toxicity is of upmost importance. Here, we analysed toxic oligomers of α-synuclein and its pathological variants in order to identify structural features that could be related to toxicity and found a novel structural polymorphism within G51D oligomers. These G51D oligomers can adopt a variety of ß-sheet-rich structures with differing degrees of α-helical content, and the helical structural content of these oligomers correlates with the level of induced cellular dysfunction in SH-SY5Y cells. This structure-function relationship observed in α-synuclein oligomers thus presents the α-helical structure as another potential structural determinant that may be linked with cellular toxicity in amyloid-related proteins.


Assuntos
Mutação , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Multimerização Proteica , alfa-Sinucleína/química , alfa-Sinucleína/genética , Humanos , Doenças Neurodegenerativas , Agregados Proteicos , Ligação Proteica , Multimerização Proteica/genética , Análise Espectral , alfa-Sinucleína/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34462355

RESUMO

α-synuclein aggregation is present in Parkinson's disease and other neuropathologies. Among the assemblies that populate the amyloid formation process, oligomers and short fibrils are the most cytotoxic. The human Hsc70-based disaggregase system can resolve α-synuclein fibrils, but its ability to target other toxic assemblies has not been studied. Here, we show that this chaperone system preferentially disaggregates toxic oligomers and short fibrils, while its activity against large, less toxic amyloids is severely impaired. Biochemical and kinetic characterization of the disassembly process reveals that this behavior is the result of an all-or-none abrupt solubilization of individual aggregates. High-speed atomic force microscopy explicitly shows that disassembly starts with the destabilization of the tips and rapidly progresses to completion through protofilament unzipping and depolymerization without accumulation of harmful oligomeric intermediates. Our data provide molecular insights into the selective processing of toxic amyloids, which is critical to identify potential therapeutic targets against increasingly prevalent neurodegenerative disorders.


Assuntos
Amiloide/metabolismo , Chaperonas Moleculares/metabolismo , alfa-Sinucleína/metabolismo , Biopolímeros/metabolismo , Humanos , Doença de Parkinson/metabolismo , Agregados Proteicos
7.
Nat Commun ; 12(1): 3752, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145261

RESUMO

α-Synuclein aggregation is a key driver of neurodegeneration in Parkinson's disease and related syndromes. Accordingly, obtaining a molecule that targets α-synuclein toxic assemblies with high affinity is a long-pursued objective. Here, we exploit the biophysical properties of toxic oligomers and amyloid fibrils to identify a family of α-helical peptides that bind to these α-synuclein species with low nanomolar affinity, without interfering with the monomeric functional protein. This activity is translated into a high anti-aggregation potency and the ability to abrogate oligomer-induced cell damage. Using a structure-guided search we identify a human peptide expressed in the brain and the gastrointestinal tract with analogous binding, anti-aggregation, and detoxifying properties. The chemical entities we describe here may represent a therapeutic avenue for the synucleinopathies and are promising tools to assist diagnosis by discriminating between native and toxic α-synuclein species.


Assuntos
Amiloide/metabolismo , Doença de Parkinson/patologia , Agregação Patológica de Proteínas/patologia , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Trato Gastrointestinal/metabolismo , Humanos
8.
Nat Commun ; 12(1): 1814, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33753734

RESUMO

The self-assembly of α-synuclein (αS) into intraneuronal inclusion bodies is a key characteristic of Parkinson's disease. To define the nature of the species giving rise to neuronal damage, we have investigated the mechanism of action of the main αS populations that have been observed to form progressively during fibril growth. The αS fibrils release soluble prefibrillar oligomeric species with cross-ß structure and solvent-exposed hydrophobic clusters. αS prefibrillar oligomers are efficient in crossing and permeabilize neuronal membranes, causing cellular insults. Short fibrils are more neurotoxic than long fibrils due to the higher proportion of fibrillar ends, resulting in a rapid release of oligomers. The kinetics of released αS oligomers match the observed kinetics of toxicity in cellular systems. In addition to previous evidence that αS fibrils can spread in different brain areas, our in vitro results reveal that αS fibrils can also release oligomeric species responsible for an immediate dysfunction of the neurons in the vicinity of these species.


Assuntos
Amiloide/metabolismo , Corpos de Inclusão/metabolismo , Neurônios/metabolismo , alfa-Sinucleína/metabolismo , Amiloide/química , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Cinética , Microscopia Confocal , Doença de Parkinson/metabolismo , Agregação Patológica de Proteínas , Multimerização Proteica , Ratos Sprague-Dawley , alfa-Sinucleína/química
9.
Biophys Chem ; 269: 106520, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33341693

RESUMO

The understanding of the complex conformational landscape of amyloid aggregation and its modulation by relevant physicochemical and cellular factors is a prerequisite for elucidating some of the molecular basis of pathology in amyloid related diseases, and for developing and evaluating effective disease-specific therapeutics to reduce or eliminate the underlying sources of toxicity in these diseases. Interactions of proteins with solvating water have been long considered to be fundamental in mediating their function and folding; however, the relevance of water in the process of protein amyloid aggregation has been largely overlooked. Here, we provide a perspective on the role water plays in triggering primary amyloid nucleation of intrinsically disordered proteins (IDPs) based on recent experimental evidences. The initiation of amyloid aggregation likely results from the synergistic effect between both protein intermolecular interactions and the properties of the water hydration layer of the protein surface. While the self-assembly of both hydrophobic and hydrophilic IDPs would be thermodynamically favoured due to large water entropy contributions, large desolvation energy barriers are expected, particularly for the nucleation of hydrophilic IDPs. Under highly hydrating conditions, primary nucleation is slow, being facilitated by the presence of nucleation-active surfaces (heterogeneous nucleation). Under conditions of poor water activity, such as those found in the interior of protein droplets generated by liquid-liquid phase separation, however, the desolvation energy barrier is significantly reduced, and nucleation can occur very rapidly in the bulk of the solution (homogeneous nucleation), giving rise to structurally distinct amyloid polymorphs. Water, therefore, plays a key role in modulating the transition free energy of amyloid nucleation, thus governing the initiation of the process, and dictating the type of preferred primary nucleation and the type of amyloid polymorph generated, which could vary depending on the particular microenvironment that the protein molecules encounter in the cell.


Assuntos
Amiloide/química , Agregados Proteicos , Água/química , Entropia , Modelos Moleculares , Conformação Proteica
10.
Int J Mol Sci ; 21(21)2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33126694

RESUMO

α-Synuclein amyloid aggregation is a defining molecular feature of Parkinson's disease, Lewy body dementia, and multiple system atrophy, but can also be found in other neurodegenerative disorders such as Alzheimer's disease. The process of α-synuclein aggregation can be initiated through alternative nucleation mechanisms and dominated by different secondary processes giving rise to multiple amyloid polymorphs and intermediate species. Some aggregated species have more inherent abilities to induce cellular stress and toxicity, while others seem to be more potent in propagating neurodegeneration. The preference for particular types of polymorphs depends on the solution conditions and the cellular microenvironment that the protein encounters, which is likely related to the distinct cellular locations of α-synuclein inclusions in different synucleinopathies, and the existence of disease-specific amyloid polymorphs. In this review, we discuss our current understanding on the nature and structure of the various types of α-synuclein aggregated species and their possible roles in pathology. Precisely defining these distinct α-synuclein species will contribute to understanding the molecular origins of these disorders, developing accurate diagnoses, and designing effective therapeutic interventions for these highly debilitating neurodegenerative diseases.


Assuntos
Doença de Alzheimer/patologia , Amiloide/química , Atrofia de Múltiplos Sistemas/patologia , Doença de Parkinson/patologia , Agregação Patológica de Proteínas , alfa-Sinucleína/química , Doença de Alzheimer/metabolismo , Animais , Humanos , Atrofia de Múltiplos Sistemas/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo
11.
Commun Biol ; 3(1): 435, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32792544

RESUMO

The onset and progression of numerous protein misfolding diseases are associated with the presence of oligomers formed during the aberrant aggregation of several different proteins, including amyloid-ß (Aß) in Alzheimer's disease and α-synuclein (αS) in Parkinson's disease. These small, soluble aggregates are currently major targets for drug discovery. In this study, we show that trodusquemine, a naturally-occurring aminosterol, markedly reduces the cytotoxicity of αS, Aß and HypF-N oligomers to human neuroblastoma cells by displacing the oligomers from cell membranes in the absence of any substantial morphological and structural changes to the oligomers. These results indicate that the reduced toxicity results from a mechanism that is common to oligomers from different proteins, shed light on the origin of the toxicity of the most deleterious species associated with protein aggregation and suggest that aminosterols have the therapeutically-relevant potential to protect cells from the oligomer-induced cytotoxicity associated with numerous protein misfolding diseases.


Assuntos
Membrana Celular/metabolismo , Colestanos/farmacologia , Dobramento de Proteína , Multimerização Proteica , Espermina/análogos & derivados , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Fenômenos Biofísicos/efeitos dos fármacos , Carboxil e Carbamoil Transferases/química , Carboxil e Carbamoil Transferases/toxicidade , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidade , Humanos , Dobramento de Proteína/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Espermina/farmacologia , alfa-Sinucleína/química , alfa-Sinucleína/toxicidade
13.
Chem Sci ; 11(43): 11902-11914, 2020 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-33520152

RESUMO

α-Synuclein amyloid self-assembly is the hallmark of a number of neurodegenerative disorders, including Parkinson's disease, although there is still very limited understanding about the factors and mechanisms that trigger this process. Primary nucleation has been observed to be initiated in vitro at hydrophobic/hydrophilic interfaces by heterogeneous nucleation generating parallel ß-sheet aggregates, although no such interfaces have yet been identified in vivo. In this work, we have discovered that α-synuclein can self-assemble into amyloid aggregates by homogeneous nucleation, without the need of an active surface, and with a preference for an antiparallel ß-sheet arrangement. This particular structure has been previously proposed to be distinctive of stable toxic oligomers and we here demonstrate that it indeed represents the most stable structure of the preferred amyloid pathway triggered by homogeneous nucleation under limited hydration conditions, including those encountered inside α-synuclein droplets generated by liquid-liquid phase separation. In addition, our results highlight the key role that water plays not only in modulating the transition free energy of amyloid nucleation, and thus governing the initiation of the process, but also in dictating the type of preferred primary nucleation and the type of amyloid polymorph generated depending on the extent of protein hydration. These findings are particularly relevant in the context of in vivo α-synuclein aggregation where the protein can encounter a variety of hydration conditions in different cellular microenvironments, including the vicinity of lipid membranes or the interior of membraneless compartments, which could lead to the formation of remarkably different amyloid polymorphs by either heterogeneous or homogeneous nucleation.

14.
Sci Rep ; 9(1): 16947, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31740740

RESUMO

The over-expression and aggregation of α-synuclein (αSyn) are linked to the onset and pathology of Parkinson's disease. Native monomeric αSyn exists in an intrinsically disordered ensemble of interconverting conformations, which has made its therapeutic targeting by small molecules highly challenging. Nonetheless, here we successfully target the monomeric structural ensemble of αSyn and thereby identify novel drug-like small molecules that impact multiple pathogenic processes. Using a surface plasmon resonance high-throughput screen, in which monomeric αSyn is incubated with microchips arrayed with tethered compounds, we identified novel αSyn interacting drug-like compounds. Because these small molecules could impact a variety of αSyn forms present in the ensemble, we tested representative hits for impact on multiple αSyn malfunctions in vitro and in cells including aggregation and perturbation of vesicular dynamics. We thereby identified a compound that inhibits αSyn misfolding and is neuroprotective, multiple compounds that restore phagocytosis impaired by αSyn overexpression, and a compound blocking cellular transmission of αSyn. Our studies demonstrate that drug-like small molecules that interact with native αSyn can impact a variety of its pathological processes. Thus, targeting the intrinsically disordered ensemble of αSyn offers a unique approach to the development of small molecule research tools and therapeutics for Parkinson's disease.


Assuntos
Bibliotecas de Moléculas Pequenas/farmacologia , alfa-Sinucleína/metabolismo , Amiloide/antagonistas & inibidores , Amiloide/metabolismo , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência , Ensaios de Triagem em Larga Escala/métodos , Humanos , Proteínas Intrinsicamente Desordenadas/metabolismo , Fagocitose/efeitos dos fármacos , Dobramento de Proteína , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/toxicidade , Ressonância de Plasmônio de Superfície , alfa-Sinucleína/química , alfa-Sinucleína/efeitos dos fármacos
15.
J Biol Chem ; 294(27): 10392-10406, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31142553

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by fibrillar neuronal inclusions composed of aggregated α-synuclein (α-syn). These inclusions are associated with behavioral and pathological PD phenotypes. One strategy for therapeutic interventions is to prevent the formation of these inclusions to halt disease progression. α-Synuclein exists in multiple structural forms, including disordered, nonamyloid oligomers, ordered amyloid oligomers, and fibrils. It is critical to understand which conformers contribute to specific PD phenotypes. Here, we utilized a mouse model to explore the pathological effects of stable ß-amyloid-sheet oligomers compared with those of fibrillar α-synuclein. We biophysically characterized these species with transmission EM, atomic-force microscopy, CD spectroscopy, FTIR spectroscopy, analytical ultracentrifugation, and thioflavin T assays. We then injected these different α-synuclein forms into the mouse striatum to determine their ability to induce PD-related phenotypes. We found that ß-sheet oligomers produce a small but significant loss of dopamine neurons in the substantia nigra pars compacta (SNc). Injection of small ß-sheet fibril fragments, however, produced the most robust phenotypes, including reduction of striatal dopamine terminals, SNc loss of dopamine neurons, and motor-behavior defects. We conclude that although the ß-sheet oligomers cause some toxicity, the potent effects of the short fibrillar fragments can be attributed to their ability to recruit monomeric α-synuclein and spread in vivo and hence contribute to the development of PD-like phenotypes. These results suggest that strategies to reduce the formation and propagation of ß-sheet fibrillar species could be an important route for therapeutic intervention in PD and related disorders.


Assuntos
Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Masculino , Camundongos , Doença de Parkinson/metabolismo , Fenótipo , Agregados Proteicos , Conformação Proteica em Folha beta , alfa-Sinucleína/química , alfa-Sinucleína/farmacologia
16.
ACS Chem Biol ; 13(8): 2308-2319, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-29953201

RESUMO

The aggregation of α-synuclein, an intrinsically disordered protein that is highly abundant in neurons, is closely associated with the onset and progression of Parkinson's disease. We have shown previously that the aminosterol squalamine can inhibit the lipid induced initiation process in the aggregation of α-synuclein, and we report here that the related compound trodusquemine is capable of inhibiting not only this process but also the fibril-dependent secondary pathways in the aggregation reaction. We further demonstrate that trodusquemine can effectively suppress the toxicity of α-synuclein oligomers in neuronal cells, and that its administration, even after the initial growth phase, leads to a dramatic reduction in the number of α-synuclein inclusions in a Caenorhabditis elegans model of Parkinson's disease, eliminates the related muscle paralysis, and increases lifespan. On the basis of these findings, we show that trodusquemine is able to inhibit multiple events in the aggregation process of α-synuclein and hence to provide important information about the link between such events and neurodegeneration, as it is initiated and progresses. Particularly in the light of the previously reported ability of trodusquemine to cross the blood-brain barrier and to promote tissue regeneration, the present results suggest that this compound has the potential to be an important therapeutic candidate for Parkinson's disease and related disorders.


Assuntos
Colestanos/farmacologia , Doença de Parkinson/tratamento farmacológico , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/prevenção & controle , Espermina/análogos & derivados , alfa-Sinucleína/metabolismo , Animais , Caenorhabditis elegans/fisiologia , Linhagem Celular , Colestanos/uso terapêutico , Modelos Animais de Doenças , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Agregação Patológica de Proteínas/metabolismo , Espermina/farmacologia , Espermina/uso terapêutico
17.
Methods Mol Biol ; 1779: 45-60, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29886526

RESUMO

Amyloid assemblies of certain proteins, including the Parkinson disease-related protein α-synuclein, are commonly associated with the development and spreading of neurodegenerative diseases, although the nature of the most toxic forms and the mechanisms by which they trigger neurodegeneration remain largely unknown. This is at least in part due to the inherent challenges involved in the preparation of stable and structurally homogeneous samples of amyloid assemblies that could be used in toxicity experiments. Here, we describe the preparation of two different types of stable α-synuclein amyloid assemblies, namely a kinetically trapped oligomeric species and a propagating-competent fibrillar polymorph. The degree of heterogeneity in the samples has been defined and carefully minimized, thus allowing for meaningful structure-toxicity relationships in different α-synuclein amyloid assemblies to be established.


Assuntos
Doença de Parkinson/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/isolamento & purificação , Amiloide/química , Amiloide/isolamento & purificação , Microscopia Crioeletrônica , Humanos , Modelos Moleculares , Multimerização Proteica , Relação Estrutura-Atividade , alfa-Sinucleína/toxicidade
18.
J Parkinsons Dis ; 8(2): 303-322, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29400668

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disease, affecting approximately one-percent of the population over the age of sixty. Although many animal models have been developed to study this disease, each model presents its own advantages and caveats. A unique model has arisen to study the role of alpha-synuclein (aSyn) in the pathogenesis of PD. This model involves the conversion of recombinant monomeric aSyn protein to a fibrillar form-the aSyn pre-formed fibril (aSyn PFF)-which is then injected into the brain or introduced to the media in culture. Although many groups have successfully adopted and replicated the aSyn PFF model, issues with generating consistent pathology have been reported by investigators. To improve the replicability of this model and diminish these issues, The Michael J. Fox Foundation for Parkinson's Research (MJFF) has enlisted the help of field leaders who performed key experiments to establish the aSyn PFF model to provide the research community with guidelines and practical tips for improving the robustness and success of this model. Specifically, we identify key pitfalls and suggestions for avoiding these mistakes as they relate to generating the aSyn PFFs from monomeric protein, validating the formation of pathogenic aSyn PFFs, and using the aSyn PFFs in vivo or in vitro to model PD. With this additional information, adoption and use of the aSyn PFF model should present fewer challenges, resulting in a robust and widely available model of PD.


Assuntos
Encéfalo/patologia , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Doença de Parkinson/metabolismo , Roedores
19.
Neurobiol Dis ; 109(Pt B): 178-190, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28709995

RESUMO

Many neurodegenerative disorders, including Alzheimer's, Parkinson's and the prion diseases, are characterized by a conformational conversion of normally soluble proteins or peptides into pathological species, by a process of misfolding and self-assembly that leads ultimately to the formation of amyloid fibrils. Recent studies support the idea that multiple intermediate species with a wide variety of degrees of neuronal toxicity are generated during such processes. The development of a high level of knowledge of the nature and structure of the pathogenic amyloid species would significantly enhance efforts to underline the molecular origins of these disorders and also to develop both accurate diagnoses and effective therapeutic interventions for these types of conditions. In this review, we discuss recent biophysical and structural information concerning different types of amyloid aggregates and the way in which such information can guide rational therapeutic approaches designed to target specific pathogenic events that occur during the development of these highly debilitating and increasingly common diseases.


Assuntos
Amiloide/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/terapia , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/terapia , Amiloide/efeitos dos fármacos , Animais , Fenômenos Biofísicos , Humanos , Dobramento de Proteína
20.
Science ; 358(6369): 1440-1443, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29242346

RESUMO

Oligomeric species populated during the aggregation process of α-synuclein have been linked to neuronal impairment in Parkinson's disease and related neurodegenerative disorders. By using solution and solid-state nuclear magnetic resonance techniques in conjunction with other structural methods, we identified the fundamental characteristics that enable toxic α-synuclein oligomers to perturb biological membranes and disrupt cellular function; these include a highly lipophilic element that promotes strong membrane interactions and a structured region that inserts into lipid bilayers and disrupts their integrity. In support of these conclusions, mutations that target the region that promotes strong membrane interactions by α-synuclein oligomers suppressed their toxicity in neuroblastoma cells and primary cortical neurons.


Assuntos
Doença de Parkinson/metabolismo , Agregação Patológica de Proteínas/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/toxicidade , Linhagem Celular Tumoral , Membrana Celular/química , Córtex Cerebral/química , Humanos , Bicamadas Lipídicas , Mutação , Neurônios/química , Neurônios/efeitos dos fármacos , Ressonância Magnética Nuclear Biomolecular , alfa-Sinucleína/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA