Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochemistry ; 61(17): 1743-1756, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35944093

RESUMEN

Parkinson's disease is associated with the aberrant aggregation of α-synuclein. Although the causes of this process are still unclear, post-translational modifications of α-synuclein are likely to play a modulatory role. Since α-synuclein is constitutively N-terminally acetylated, we investigated how this post-translational modification alters the aggregation behavior of this protein. By applying a three-pronged aggregation kinetics approach, we observed that N-terminal acetylation results in a reduced rate of lipid-induced aggregation and slows down both elongation and fibril-catalyzed aggregate proliferation. An analysis of the amyloid fibrils produced by the aggregation process revealed different morphologies for the acetylated and non-acetylated forms in both lipid-induced aggregation and seed-induced aggregation assays. In addition, we found that fibrils formed by acetylated α-synuclein exhibit a lower ß-sheet content. These findings indicate that N-terminal acetylation of α-synuclein alters its lipid-dependent aggregation behavior, reduces its rate of in vitro aggregation, and affects the structural properties of its fibrillar aggregates.


Asunto(s)
Amiloide , alfa-Sinucleína , Acetilación , Amiloide/química , Lípidos , Agregado de Proteínas , Procesamiento Proteico-Postraduccional , alfa-Sinucleína/química
2.
RSC Chem Biol ; 2(4): 1232-1238, 2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34458836

RESUMEN

We demonstrate a solution method that allows both elongation rate and average fibril length of assembling amyloid fibrils to be estimated. The approach involves acquisition of real-time neutron scattering data during the initial stages of seeded growth, using contrast matched buffer to make the seeds effectively invisible to neutrons. As deuterated monomers add on to the seeds, the labelled growing ends give rise to scattering patterns that we model as cylinders whose increase in length with time gives an elongation rate. In addition, the absolute intensity of the signal can be used to determine the number of growing ends per unit volume, which in turn provides an estimate of seed length. The number of ends did not change significantly during elongation, demonstrating that any spontaneous or secondary nucleation was not significant compared with growth on the ends of pre-existing fibrils, and in addition providing a method of internal validation for the technique. Our experiments on initial growth of alpha synuclein fibrils using 1.2 mg ml-1 seeds in 2.5 mg ml-1 deuterated monomer at room temperature gave an elongation rate of 6.3 ± 0.5 Å min-1, and an average seed length estimate of 4.2 ± 1.3 µm.

3.
Front Neurosci ; 15: 680026, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220435

RESUMEN

The aberrant aggregation of proteins is a key molecular event in the development and progression of a wide range of neurodegenerative disorders. We have shown previously that squalamine and trodusquemine, two natural products in the aminosterol class, can modulate the aggregation of the amyloid-ß peptide (Aß) and of α-synuclein (αS), which are associated with Alzheimer's and Parkinson's diseases. In this work, we expand our previous analyses to two squalamine derivatives, des-squalamine and α-squalamine, obtaining further insights into the mechanism by which aminosterols modulate Aß and αS aggregation. We then characterize the ability of these small molecules to alter the physicochemical properties of stabilized oligomeric species in vitro and to suppress the toxicity of these aggregates to varying degrees toward human neuroblastoma cells. We found that, despite the fact that these aminosterols exert opposing effects on Aß and αS aggregation under the conditions that we tested, the modifications that they induced to the toxicity of oligomers were similar. Our results indicate that the suppression of toxicity is mediated by the displacement of toxic oligomeric species from cellular membranes by the aminosterols. This study, thus, provides evidence that aminosterols could be rationally optimized in drug discovery programs to target oligomer toxicity in Alzheimer's and Parkinson's diseases.

4.
Front Cell Dev Biol ; 9: 552549, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33829010

RESUMEN

The aggregation of α-synuclein is a hallmark of Parkinson's disease (PD) and a variety of related neurological disorders. A number of mutations in this protein, including A30P and A53T, are associated with familial forms of the disease. Patients carrying the A30P mutation typically exhibit a similar age of onset and symptoms as sporadic PD, while those carrying the A53T mutation generally have an earlier age of onset and an accelerated progression. We report two C. elegans models of PD (PDA30P and PDA53T), which express these mutational variants in the muscle cells, and probed their behavior relative to animals expressing the wild-type protein (PDWT). PDA30P worms showed a reduced speed of movement and an increased paralysis rate, control worms, but no change in the frequency of body bends. By contrast, in PDA53T worms both speed and frequency of body bends were significantly decreased, and paralysis rate was increased. α-Synuclein was also observed to be less well localized into aggregates in PDA30P worms compared to PDA53T and PDWT worms, and amyloid-like features were evident later in the life of the animals, despite comparable levels of expression of α-synuclein. Furthermore, squalamine, a natural product currently in clinical trials for treating symptomatic aspects of PD, was found to reduce significantly the aggregation of α-synuclein and its associated toxicity in PDA53T and PDWT worms, but had less marked effects in PDA30P. In addition, using an antibody that targets the N-terminal region of α-synuclein, we observed a suppression of toxicity in PDA30P, PDA53T and PDWT worms. These results illustrate the use of these two C. elegans models in fundamental and applied PD research.

5.
J Biol Chem ; 296: 100631, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33823153

RESUMEN

TREM2 is a pattern recognition receptor, expressed on microglia and myeloid cells, detecting lipids and Aß and inducing an innate immune response. Missense mutations (e.g., R47H) of TREM2 increase risk of Alzheimer's disease (AD). The soluble ectodomain of wild-type TREM2 (sTREM2) has been shown to protect against AD in vivo, but the underlying mechanisms are unclear. We show that Aß oligomers bind to cellular TREM2, inducing shedding of the sTREM2 domain. Wild-type sTREM2 bound to Aß oligomers (measured by single-molecule imaging, dot blots, and Bio-Layer Interferometry) inhibited Aß oligomerization and disaggregated preformed Aß oligomers and protofibrils (measured by transmission electron microscopy, dot blots, and size-exclusion chromatography). Wild-type sTREM2 also inhibited Aß fibrillization (measured by imaging and thioflavin T fluorescence) and blocked Aß-induced neurotoxicity (measured by permeabilization of artificial membranes and by loss of neurons in primary neuronal-glial cocultures). In contrast, the R47H AD-risk variant of sTREM2 is less able to bind and disaggregate oligomeric Aß but rather promotes Aß protofibril formation and neurotoxicity. Thus, in addition to inducing an immune response, wild-type TREM2 may protect against amyloid pathology by the Aß-induced release of sTREM2, which blocks Aß aggregation and neurotoxicity. In contrast, R47H sTREM2 promotes Aß aggregation into protofibril that may be toxic to neurons. These findings may explain how wild-type sTREM2 apparently protects against AD in vivo and why a single copy of the R47H variant gene is associated with increased AD risk.


Asunto(s)
Péptidos beta-Amiloides/química , Amiloide/química , Glicoproteínas de Membrana/fisiología , Proteínas Mutantes/metabolismo , Mutación , Neuronas/patología , Síndromes de Neurotoxicidad/patología , Receptores Inmunológicos/fisiología , Enfermedad de Alzheimer , Amiloide/metabolismo , Animales , Ratones , Ratones Noqueados , Proteínas Mutantes/genética , Neuronas/metabolismo , Síndromes de Neurotoxicidad/etiología
6.
Nat Struct Mol Biol ; 27(10): 886-891, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32778821

RESUMEN

The formation of amyloid deposits in human tissues is a defining feature of more than 50 medical disorders, including Alzheimer's disease. Strong genetic and histological evidence links these conditions to the process of protein aggregation, yet it has remained challenging to identify a definitive connection between aggregation and pathogenicity. Using time-resolved fluorescence microscopy of individual synthetic vesicles, we show for the Aß42 peptide implicated in Alzheimer's disease that the disruption of lipid bilayers correlates linearly with the time course of the levels of transient oligomers generated through secondary nucleation. These findings indicate a specific role of oligomers generated through the catalytic action of fibrillar species during the protein aggregation process in driving deleterious biological function and establish a direct causative connection between amyloid formation and its pathological effects.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/toxicidad , Calcio/metabolismo , Permeabilidad de la Membrana Celular , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Cinética , Membrana Dobles de Lípidos , Microscopía Fluorescente , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Imagen Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/toxicidad
7.
ACS Nano ; 14(5): 5213-5222, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32159944

RESUMEN

Proteinaceous deposits of α-synuclein amyloid fibrils are a hallmark of human disorders including Parkinson's disease. The onset of this disease is also associated with five familial mutations of the gene encoding the protein. However, the mechanistic link between single point mutations and the kinetics of aggregation, biophysical properties of the resulting amyloid fibrils, and an increased risk of disease is still elusive. Here, we demonstrate that the disease-associated mutations of α-synuclein generate different amyloid fibril polymorphs compared to the wild type protein. Remarkably, the α-synuclein variants forming amyloid fibrils of a comparable structure, morphology, and heterogeneity show similar microscopic steps defining the aggregation kinetics. These results demonstrate that a single point mutation can significantly alter the distribution of fibrillar polymorphs in α-synuclein, suggesting that differences in the clinical phenotypes of familial Parkinson's disease could be associated with differences in the mechanism of formation and the structural characteristics of the aggregates.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Amiloide/genética , Biofisica , Humanos , Mutación , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética
8.
Commun Chem ; 3(1): 191, 2020 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-36703335

RESUMEN

The aggregation of α-synuclein is a central event in Parkinsons's disease and related synucleinopathies. Since pharmacologically targeting this process, however, has not yet resulted in approved disease-modifying treatments, there is an unmet need of developing novel methods of drug discovery. In this context, the use of chemical kinetics has recently enabled accurate quantifications of the microscopic steps leading to the proliferation of protein misfolded oligomers. As these species are highly neurotoxic, effective therapeutic strategies may be aimed at reducing their numbers. Here, we exploit this quantitative approach to develop a screening strategy that uses the reactive flux toward α-synuclein oligomers as a selection parameter. Using this approach, we evaluate the efficacy of a library of flavone derivatives, identifying apigenin as a compound that simultaneously delays and reduces the formation of α-synuclein oligomers. These results demonstrate a compound selection strategy based on the inhibition of the formation of α-synuclein oligomers, which may be key in identifying small molecules in drug discovery pipelines for diseases associated with α-synuclein aggregation.

9.
Elife ; 82019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31389332

RESUMEN

Removing or preventing the formation of [Formula: see text]-synuclein aggregates is a plausible strategy against Parkinson's disease. To this end, we have engineered the [Formula: see text]-wrapin AS69 to bind monomeric [Formula: see text]-synuclein with high affinity. In cultured cells, AS69 reduced the self-interaction of [Formula: see text]-synuclein and formation of visible [Formula: see text]-synuclein aggregates. In flies, AS69 reduced [Formula: see text]-synuclein aggregates and the locomotor deficit resulting from [Formula: see text]-synuclein expression in neuronal cells. In biophysical experiments in vitro, AS69 highly sub-stoichiometrically inhibited both primary and autocatalytic secondary nucleation processes, even in the presence of a large excess of monomer. We present evidence that the AS69-[Formula: see text]-synuclein complex, rather than the free AS69, is the inhibitory species responsible for sub-stoichiometric inhibition of secondary nucleation. These results represent a new paradigm that high affinity monomer binders can lead to strongly sub-stoichiometric inhibition of nucleation processes.


Asunto(s)
Amiloide/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , alfa-Sinucleína/metabolismo , Células HEK293 , Humanos , Agregación Patológica de Proteínas , Multimerización de Proteína/efectos de los fármacos , Proteínas Recombinantes/genética
10.
Nat Commun ; 10(1): 1541, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30948723

RESUMEN

Protein aggregation is a complex process resulting in the formation of heterogeneous mixtures of aggregate populations that are closely linked to neurodegenerative conditions, such as Alzheimer's disease. Here, we find that soluble aggregates formed at different stages of the aggregation process of amyloid beta (Aß42) induce the disruption of lipid bilayers and an inflammatory response to different extents. Further, by using gradient ultracentrifugation assay, we show that the smaller aggregates are those most potent at inducing membrane permeability and most effectively inhibited by antibodies binding to the C-terminal region of Aß42. By contrast, we find that the larger soluble aggregates are those most effective at causing an inflammatory response in microglia cells and more effectively inhibited by antibodies targeting the N-terminal region of Aß42. These findings suggest that different toxic mechanisms driven by different soluble aggregated species of Aß42 may contribute to the onset and progression of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Membrana Dobles de Lípidos/metabolismo , Agregación Patológica de Proteínas , Péptidos beta-Amiloides/metabolismo , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Ultracentrifugación
11.
ACS Chem Neurosci ; 10(5): 2374-2384, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30793584

RESUMEN

Aggregation of the amyloid-ß (Aß) peptide into plaques is believed to play a crucial role in Alzheimer's disease. Amyloid plaques consist of fibrils of full length Aß peptides as well as N-terminally truncated species. ß-Site amyloid precursor protein-cleaving enzyme (BACE1) cleaves amyloid precursor protein in the first step in Aß peptide production and is an attractive therapeutic target to limit Aß generation. Inhibition of BACE1, however, induces a unique pattern of Aß peptides with increased levels of N-terminally truncated Aß peptides starting at position 5 (Aß5-X), indicating that these peptides are generated through a BACE1-independent pathway. Here we elucidate the aggregation mechanism of Aß5-42 and its influence on full-length Aß42. We find that, compared to Aß42, Aß5-42 is more aggregation prone and displays enhanced nucleation rates. Aß5-42 oligomers cause nonspecific membrane disruption to similar extent as Aß42 but appear at earlier time points in the aggregation reaction. Noteworthy, this implies similar toxicity of Aß42 and Aß5-42 and the toxic species are generated faster by Aß5-42. The increased rate of secondary nucleation on the surface of existing fibrils originates from a higher affinity of Aß5-42 monomers for fibrils, as compared to Aß42: an effect that may be related to the reduced net charge of Aß5-42. Moreover, Aß5-42 and Aß42 peptides coaggregate into heteromolecular fibrils and either species can elongate existing Aß42 or Aß5-42 fibrils but Aß42 fibrils are more catalytic than Aß5-42 fibrils. Our findings highlight the importance of the N-terminus for surface-catalyzed nucleation and thus the production of toxic oligomers.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Humanos , Cinética , Nanopartículas/metabolismo
12.
Nano Lett ; 18(12): 7494-7501, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30380895

RESUMEN

Proteins fold into a single structural ensemble but can also misfold into many diverse structures including small aggregates and fibrils, which differ in their toxicity. The aggregate surface properties play an important role in how they interact with the plasma membrane and cellular organelles, potentially inducing cellular toxicity, however, these properties have not been measured to date due to the lack of suitable methods. Here, we used a spectrally resolved, super-resolution imaging method combined with an environmentally sensitive fluorescent dye to measure the surface hydrophobicity of individual aggregates formed by the protein α-synuclein (αS), whose aggregation is associated with Parkinson's disease. We show that the surface of soluble oligomers is more hydrophobic than fibrils and populates a diverse range of coexisting states. Overall, our data show that the conversion of oligomers to fibril-like aggregates and ultimately to fibrils results in a reduction in both hydrophobicity and the variation in hydrophobicity. This funneling characteristic of the energy landscape explains many of the observed properties of αS aggregates and may be a common feature of aggregating proteins.


Asunto(s)
Agregado de Proteínas , alfa-Sinucleína/química , Colorantes Fluorescentes/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Imagen Óptica , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/metabolismo , Multimerización de Proteína , Solubilidad , alfa-Sinucleína/metabolismo
13.
ACS Nano ; 12(11): 10855-10866, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30371053

RESUMEN

Small oligomers of the protein α-synuclein (αS) are highly cytotoxic species associated with Parkinson's disease (PD). In addition, αS can form co-aggregates with its mutational variants and with other proteins such as amyloid-ß (Aß) and tau, which are implicated in Alzheimer's disease. The processes of self-oligomerization and co-oligomerization of αS are, however, challenging to study quantitatively. Here, we have utilized single-molecule techniques to measure the equilibrium populations of oligomers formed in vitro by mixtures of wild-type αS with its mutational variants and with Aß40, Aß42, and a fragment of tau. Using a statistical mechanical model, we find that co-oligomer formation is generally more favorable than self-oligomer formation at equilibrium. Furthermore, self-oligomers more potently disrupt lipid membranes than do co-oligomers. However, this difference is sometimes outweighed by the greater formation propensity of co-oligomers when multiple proteins coexist. Our results suggest that co-oligomer formation may be important in PD and related neurodegenerative diseases.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , alfa-Sinucleína/metabolismo , Proteínas tau/biosíntesis , Péptidos beta-Amiloides/química , Humanos , Modelos Moleculares , Termodinámica , alfa-Sinucleína/química , Proteínas tau/química
14.
Chembiochem ; 19(19): 2033-2038, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30051958

RESUMEN

The aberrant misfolding and subsequent conversion of monomeric protein into amyloid aggregates characterises many neurodegenerative disorders, including Parkinson's and Alzheimer's diseases. These aggregates are highly heterogeneous in structure, generally of low abundance and typically smaller than the diffraction limit of light (≈250 nm). To overcome the challenges these characteristics pose to the study of endogenous aggregates formed in cells, we have developed a method to characterise them at the nanometre scale without the need for a conjugated fluorophore. Using a combination of DNA PAINT and an amyloid-specific aptamer, we demonstrate that this technique is able to detect and super-resolve a range of aggregated species, including those formed by α-synuclein and amyloid-ß. Additionally, this method enables endogenous protein aggregates within cells to be characterised. We found that neuronal cells derived from patients with Parkinson's disease contain a larger number of protein aggregates than those from healthy controls.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Neuronas/patología , Enfermedad de Parkinson/patología , Agregado de Proteínas , alfa-Sinucleína/química , Péptidos beta-Amiloides/metabolismo , Aptámeros de Péptidos/química , Humanos , Agregación Patológica de Proteínas , alfa-Sinucleína/metabolismo
15.
ACS Chem Biol ; 13(8): 2308-2319, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29953201

RESUMEN

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.


Asunto(s)
Colestanos/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/prevención & control , Espermina/análogos & derivados , alfa-Sinucleína/metabolismo , Animales , Caenorhabditis elegans/fisiología , Línea Celular , Colestanos/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/metabolismo , Espermina/farmacología , Espermina/uso terapéutico
16.
Cell Rep ; 23(12): 3492-3500, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29924993

RESUMEN

The aberrant aggregation of α-synuclein is associated with several human diseases, collectively termed the α-synucleinopathies, which includes Parkinson's disease. The progression of these diseases is, in part, mediated by extracellular α-synuclein oligomers that may exert effects through several mechanisms, including prion-like transfer, direct cytotoxicity, and pro-inflammatory actions. In this study, we show that two abundant extracellular chaperones, clusterin and α2-macroglobulin, directly bind to exposed hydrophobic regions on the surface of α-synuclein oligomers. Using single-molecule fluorescence techniques, we found that clusterin, unlike α2-macroglobulin, exhibits differential binding to α-synuclein oligomers that may be related to structural differences between two previously described forms of αS oligomers. The binding of both chaperones reduces the ability of the oligomers to permeabilize lipid membranes and prevents an oligomer-induced increase in ROS production in cultured neuronal cells. Taken together, these data suggest a neuroprotective role for extracellular chaperones in suppressing the toxicity associated with α-synuclein oligomers.


Asunto(s)
Espacio Extracelular/metabolismo , Multimerización de Proteína , alfa-Sinucleína/química , alfa-Sinucleína/toxicidad , Interacciones Hidrofóbicas e Hidrofílicas , Chaperonas Moleculares/metabolismo , Unión Proteica
17.
Angew Chem Int Ed Engl ; 57(18): 4886-4890, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29342318

RESUMEN

Small aggregates of misfolded proteins play a key role in neurodegenerative disorders. Such species have proved difficult to study due to the lack of suitable methods capable of resolving these heterogeneous aggregates, which are smaller than the optical diffraction limit. We demonstrate here an all-optical fluorescence microscopy method to characterise the structure of individual protein aggregates based on the fluorescence anisotropy of dyes such as thioflavin-T, and show that this technology is capable of studying oligomers in human biofluids such as cerebrospinal fluid. We first investigated in vitro the structural changes in individual oligomers formed during the aggregation of recombinant α-synuclein. By studying the diffraction-limited aggregates we directly evaluated their structural conversion and correlated this with the potential of aggregates to disrupt lipid bilayers. We finally characterised the structural features of aggregates present in cerebrospinal fluid of Parkinson's disease patients and age-matched healthy controls.


Asunto(s)
Imagen Óptica , alfa-Sinucleína/análisis , alfa-Sinucleína/química , Humanos , Agregado de Proteínas , Conformación Proteica
18.
ACS Chem Biol ; 13(3): 636-646, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29300447

RESUMEN

As a key player of the protein quality control network of the cell, the molecular chaperone Hsp70 inhibits the aggregation of the amyloid protein tau. To date, the mechanism of this inhibition and the tau species targeted by Hsp70 remain unknown. This is partly due to the inherent difficulty of studying amyloid aggregates because of their heterogeneous and transient nature. Here, we used ensemble and single-molecule fluorescence measurements to dissect how Hsp70 counteracts the self-assembly process of the K18 ΔK280 tau variant. We found that Hsp70 blocks the early stages of tau aggregation by suppressing the formation of tau nuclei. Additionally, Hsp70 sequesters oligomers and mature tau fibrils with nanomolar affinity into a protective complex, efficiently neutralizing their ability to damage membranes and seed further tau aggregation. Our results provide novel insights into the molecular mechanisms by which the chaperone Hsp70 counteracts the formation, propagation, and toxicity of tau aggregates.


Asunto(s)
Proteínas HSP70 de Choque Térmico/farmacología , Agregación Patológica de Proteínas/tratamiento farmacológico , Proteínas tau/antagonistas & inhibidores , Amiloide/efectos de los fármacos , Fluorescencia , Humanos , Imagen Individual de Molécula
19.
Cell Rep ; 21(11): 3310-3316, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29241555

RESUMEN

One potential therapeutic strategy for Alzheimer's disease (AD) is to use antibodies that bind to small soluble protein aggregates to reduce their toxic effects. However, these therapies are rarely tested in human CSF before clinical trials because of the lack of sensitive methods that enable the measurement of aggregate-induced toxicity at low concentrations. We have developed highly sensitive single vesicle and single-cell-based assays that detect the Ca2+ influx caused by the CSF of individuals affected with AD and healthy controls, and we have found comparable effects for both types of samples. We also show that an extracellular chaperone clusterin; a nanobody specific to the amyloid-ß peptide (Aß); and bapineuzumab, a humanized monoclonal antibody raised against Aß, could all reduce the Ca2+ influx caused by synthetic Aß oligomers but are less effective in CSF. These assays could be used to characterize potential therapeutic agents in CSF before clinical trials.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Bioensayo , Calcio/metabolismo , Líquido Cefalorraquídeo/química , Vesículas Citoplasmáticas/efectos de los fármacos , Fragmentos de Péptidos/antagonistas & inhibidores , Agregado de Proteínas/efectos de los fármacos , Anciano , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Clusterina/farmacología , Medios de Cultivo/farmacología , Vesículas Citoplasmáticas/metabolismo , Femenino , Humanos , Transporte Iónico/efectos de los fármacos , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Cultivo Primario de Células , Ratas , Anticuerpos de Dominio Único/farmacología
20.
Proc Natl Acad Sci U S A ; 114(30): 8005-8010, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28698377

RESUMEN

The coaggregation of the amyloid-ß peptide (Aß) and α-synuclein is commonly observed in a range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. The complex interplay between Aß and α-synuclein has led to seemingly contradictory results on whether α-synuclein promotes or inhibits Aß aggregation. Here, we show how these conflicts can be rationalized and resolved by demonstrating that different structural forms of α-synuclein exert different effects on Aß aggregation. Our results demonstrate that whereas monomeric α-synuclein blocks the autocatalytic proliferation of Aß42 (the 42-residue form of Aß) fibrils, fibrillar α-synuclein catalyses the heterogeneous nucleation of Aß42 aggregates. It is thus the specific balance between the concentrations of monomeric and fibrillar α-synuclein that determines the outcome of the Aß42 aggregation reaction.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , alfa-Sinucleína/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...