Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(7): e2220075121, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38335256

RESUMEN

Self-replication of amyloid fibrils via secondary nucleation is an intriguing physicochemical phenomenon in which existing fibrils catalyze the formation of their own copies. The molecular events behind this fibril surface-mediated process remain largely inaccessible to current structural and imaging techniques. Using statistical mechanics, computer modeling, and chemical kinetics, we show that the catalytic structure of the fibril surface can be inferred from the aggregation behavior in the presence and absence of a fibril-binding inhibitor. We apply our approach to the case of Alzheimer's A[Formula: see text] amyloid fibrils formed in the presence of proSP-C Brichos inhibitors. We find that self-replication of A[Formula: see text] fibrils occurs on small catalytic sites on the fibril surface, which are far apart from each other, and each of which can be covered by a single Brichos inhibitor.


Asunto(s)
Péptidos beta-Amiloides , Amiloide , Péptidos beta-Amiloides/química , Amiloide/química , Simulación por Computador , Fragmentos de Péptidos/química , Cinética
2.
Proc Natl Acad Sci U S A ; 121(18): e2313107121, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38652742

RESUMEN

Full understanding of proteostasis and energy utilization in cells will require knowledge of the fraction of cell proteins being degraded with different half-lives and their rates of synthesis. We therefore developed a method to determine such information that combines mathematical analysis of protein degradation kinetics obtained in pulse-chase experiments with Bayesian data fitting using the maximum entropy principle. This approach will enable rapid analyses of whole-cell protein dynamics in different cell types, physiological states, and neurodegenerative disease. Using it, we obtained surprising insights about protein stabilities in cultured cells normally and upon activation of proteolysis by mTOR inhibition and increasing cAMP or cGMP. It revealed that >90% of protein content in dividing mammalian cell lines is long-lived, with half-lives of 24 to 200 h, and therefore comprises much of the proteins in daughter cells. The well-studied short-lived proteins (half-lives < 10 h) together comprise <2% of cell protein mass, but surprisingly account for 10 to 20% of measurable newly synthesized protein mass. Evolution thus appears to have minimized intracellular proteolysis except to rapidly eliminate misfolded and regulatory proteins.


Asunto(s)
Entropía , Proteolisis , Proteoma , Proteoma/metabolismo , Humanos , Animales , Teorema de Bayes , Proteostasis , Cinética , AMP Cíclico/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , GMP Cíclico/metabolismo
3.
Nat Chem Biol ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38503834

RESUMEN

Segments of proteins with high ß-strand propensity can self-associate to form amyloid fibrils implicated in many diseases. We describe a general approach to bind such segments in ß-strand and ß-hairpin conformations using de novo designed scaffolds that contain deep peptide-binding clefts. The designs bind their cognate peptides in vitro with nanomolar affinities. The crystal structure of a designed protein-peptide complex is close to the design model, and NMR characterization reveals how the peptide-binding cleft is protected in the apo state. We use the approach to design binders to the amyloid-forming proteins transthyretin, tau, serum amyloid A1 and amyloid ß1-42 (Aß42). The Aß binders block the assembly of Aß fibrils as effectively as the most potent of the clinically tested antibodies to date and protect cells from toxic Aß42 species.

4.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836595

RESUMEN

Protein aggregation is associated with a wide range of degenerative human diseases with devastating consequences, as exemplified by Alzheimer's, Parkinson's, and Huntington's diseases. In vitro kinetic studies have provided a mechanistic understanding of the aggregation process at the molecular level. However, it has so far remained largely unclear to what extent the biophysical principles of amyloid formation learned in vitro translate to the complex environment of living organisms. Here, we take advantage of the unique properties of a Caenorhabditis elegans model expressing a fluorescently tagged polyglutamine (polyQ) protein, which aggregates into discrete micrometer-sized inclusions that can be directly visualized in real time. We provide a quantitative analysis of protein aggregation in this system and show that the data are described by a molecular model where stochastic nucleation occurs independently in each cell, followed by rapid aggregate growth. Global fitting of the image-based aggregation kinetics reveals a nucleation rate corresponding to 0.01 h-1 per cell at 1 mM intracellular protein concentration, and shows that the intrinsic molecular stochasticity of nucleation accounts for a significant fraction of the observed animal-to-animal variation. Our results highlight how independent, stochastic nucleation events in individual cells control the overall progression of polyQ aggregation in a living animal. The key finding that the biophysical principles associated with protein aggregation in small volumes remain the governing factors, even in the complex environment of a living organism, will be critical for the interpretation of in vivo data from a wide range of protein aggregation diseases.


Asunto(s)
Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Amiloide/metabolismo , Animales , Caenorhabditis elegans , Cinética , Modelos Moleculares , Células Musculares/metabolismo , Agregado de Proteínas
5.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34518228

RESUMEN

Molecular chaperones are key components of the cellular proteostasis network whose role includes the suppression of the formation and proliferation of pathogenic aggregates associated with neurodegenerative diseases. The molecular principles that allow chaperones to recognize misfolded and aggregated proteins remain, however, incompletely understood. To address this challenge, here we probe the thermodynamics and kinetics of the interactions between chaperones and protein aggregates under native solution conditions using a microfluidic platform. We focus on the binding between amyloid fibrils of α-synuclein, associated with Parkinson's disease, to the small heat-shock protein αB-crystallin, a chaperone widely involved in the cellular stress response. We find that αB-crystallin binds to α-synuclein fibrils with high nanomolar affinity and that the binding is driven by entropy rather than enthalpy. Measurements of the change in heat capacity indicate significant entropic gain originates from the disassembly of the oligomeric chaperones that function as an entropic buffer system. These results shed light on the functional roles of chaperone oligomerization and show that chaperones are stored as inactive complexes which are capable of releasing active subunits to target aberrant misfolded species.


Asunto(s)
Amiloide/metabolismo , Proteínas de Choque Térmico Pequeñas/metabolismo , Cadena B de alfa-Cristalina/metabolismo , alfa-Sinucleína/metabolismo , Entropía , Humanos , Enfermedad de Parkinson/metabolismo , Agregado de Proteínas/fisiología , Proteostasis/fisiología
6.
Biochemistry ; 62(21): 3036-3040, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37788367

RESUMEN

Human annexin A7, a calcium- and phospholipid-binding protein, governs calcium homeostasis, plasma membrane repair, apoptosis, and tumor progression. A7 contains an N-terminal proline-rich domain (PRD; 180 residues, ∼24% prolines) that determines its functional specificity. Using microscopy and dye-binding assays, we show that recombinant A7 and its isolated PRD spontaneously phase separate into spherical condensates, which subsequently transform into ß-sheet-rich fibrils. We demonstrate that fibrillization of A7-PRD proceeds via primary nucleation and fibril-catalyzed secondary nucleation processes, as determined by chemical kinetics, providing a mechanistic basis for its amyloid assembly. This study confirms and highlights a subclass of eukaryotic PRDs prone to forming aggregates with important physiological and pathological implications.


Asunto(s)
Anexina A7 , Calcio , Humanos , Anexina A7/química , Anexina A7/metabolismo , Calcio/metabolismo , Dominios Proteicos , Amiloide/química , Prolina/química
7.
Proc Natl Acad Sci U S A ; 117(41): 25272-25283, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33004626

RESUMEN

Crystals, nanoparticles, and fibrils catalyze the generation of new aggregates on their surface from the same type of monomeric building blocks as the parent assemblies. This secondary nucleation process can be many orders of magnitude faster than primary nucleation. In the case of amyloid fibrils associated with Alzheimer's disease, this process leads to the multiplication and propagation of aggregates, whereby short-lived oligomeric intermediates cause neurotoxicity. Understanding the catalytic activity is a fundamental goal in elucidating the molecular mechanisms of Alzheimer's and associated diseases. Here we explore the role of fibril structure and hydrophobicity by asking whether the V18, A21, V40, and A42 side chains which are exposed on the Aß42 fibril surface as continuous hydrophobic patches play a role in secondary nucleation. Single, double, and quadruple serine substitutions were made. Kinetic analyses of aggregation data at multiple monomer concentrations reveal that all seven mutants retain the dominance of secondary nucleation as the main mechanism of fibril proliferation. This finding highlights the generality of secondary nucleation and its independence of the detailed molecular structure. Cryo-electron micrographs reveal that the V18S substitution causes fibrils to adopt a distinct morphology with longer twist distance than variants lacking this substitution. Self- and cross-seeding data show that surface catalysis is only efficient between peptides of identical morphology, indicating a templating role of secondary nucleation with structural conversion at the fibril surface. Our findings thus provide clear evidence that the propagation of amyloid fibril strains is possible even in systems dominated by secondary nucleation rather than fragmentation.


Asunto(s)
Amiloide/química , Microscopía por Crioelectrón , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformación Proteica
8.
Proc Natl Acad Sci U S A ; 117(22): 12087-12094, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32414930

RESUMEN

The spontaneous assembly of proteins into amyloid fibrils is a phenomenon central to many increasingly common and currently incurable human disorders, including Alzheimer's and Parkinson's diseases. Oligomeric species form transiently during this process and not only act as essential intermediates in the assembly of new filaments but also represent major pathogenic agents in these diseases. While amyloid fibrils possess a common, defining set of physicochemical features, oligomers, by contrast, appear much more diverse, and their commonalities and differences have hitherto remained largely unexplored. Here, we use the framework of chemical kinetics to investigate their dynamical properties. By fitting experimental data for several unrelated amyloidogenic systems to newly derived mechanistic models, we find that oligomers present with a remarkably wide range of kinetic and thermodynamic stabilities but that they possess two properties that are generic: they are overwhelmingly nonfibrillar, and they predominantly dissociate back to monomers rather than maturing into fibrillar species. These discoveries change our understanding of the relationship between amyloid oligomers and amyloid fibrils and have important implications for the nature of their cellular toxicity.


Asunto(s)
Amiloide/química , Proteínas Amiloidogénicas/química , Cinética , Enfermedad de Alzheimer , Péptidos beta-Amiloides/química , Amiloidosis , Modelos Teóricos , Agregado de Proteínas , Termodinámica
9.
Proc Natl Acad Sci U S A ; 117(21): 11265-11273, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32439711

RESUMEN

The nucleation of Alzheimer-associated Aß peptide monomers can be catalyzed by preexisting Aß fibrils. This leads to autocatalytic amplification of aggregate mass and underlies self-replication and generation of toxic oligomers associated with several neurodegenerative diseases. However, the nature of the interactions between the monomeric species and the fibrils during this key process, and indeed the ultrastructural localization of the interaction sites have remained elusive. Here we used NMR and optical spectroscopy to identify conditions that enable the capture of transient species during the aggregation and secondary nucleation of the Aß42 peptide. Cryo-electron microscopy (cryo-EM) images show that new aggregates protrude from the entire length of the progenitor fibril. These protrusions are morphologically distinct from the well-ordered fibrils dominating at the end of the aggregation process. The data provide direct evidence that self-replication through secondary nucleation occurs along the sides of fibrils, which become heavily decorated under the current solution conditions (14 µM Aß42, 20 mM sodium phosphate, 200 µM EDTA, pH 6.8).


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/ultraestructura , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/ultraestructura , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Amiloide/ultraestructura , Péptidos beta-Amiloides/química , Benzotiazoles/química , Benzotiazoles/metabolismo , Microscopía por Crioelectrón , Humanos , Concentración de Iones de Hidrógeno , Cinética , Espectroscopía de Resonancia Magnética , Fragmentos de Péptidos/química , Imagen de Lapso de Tiempo
10.
Proc Natl Acad Sci U S A ; 117(39): 24251-24257, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32929030

RESUMEN

Understanding the mechanism of action of compounds capable of inhibiting amyloid-fibril formation is critical to the development of potential therapeutics against protein-misfolding diseases. A fundamental challenge for progress is the range of possible target species and the disparate timescales involved, since the aggregating proteins are simultaneously the reactants, products, intermediates, and catalysts of the reaction. It is a complex problem, therefore, to choose the states of the aggregating proteins that should be bound by the compounds to achieve the most potent inhibition. We present here a comprehensive kinetic theory of amyloid-aggregation inhibition that reveals the fundamental thermodynamic and kinetic signatures characterizing effective inhibitors by identifying quantitative relationships between the aggregation and binding rate constants. These results provide general physical laws to guide the design and optimization of inhibitors of amyloid-fibril formation, revealing in particular the important role of on-rates in the binding of the inhibitors.


Asunto(s)
Amiloide/química , Modelos Químicos , Agregación Patológica de Proteínas/tratamiento farmacológico , Diseño de Fármacos , Cinética , Terapia Molecular Dirigida , Termodinámica
11.
Biophys J ; 121(22): 4280-4298, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36230002

RESUMEN

Mutations in the TP53 gene are common in cancer with the R248Q missense mutation conferring an increased propensity to aggregate. Previous p53 aggregation studies showed that, at micromolar concentrations, protein unfolding to produce aggregation-prone species is the rate-determining step. Here we show that, at physiological concentrations, aggregation kinetics of insect cell-derived full-length wild-type p53 and p53R248Q are determined by a nucleation-growth model, rather than formation of aggregation-prone monomeric species. Self-seeding, but not cross-seeding, increases aggregation rate, confirming the aggregation process as rate determining. p53R248Q displays enhanced aggregation propensity due to decreased solubility and increased aggregation rate, forming greater numbers of larger amorphous aggregates that disrupt lipid bilayers and invokes an inflammatory response. These results suggest that p53 aggregation can occur under physiological conditions, a rate enhanced by R248Q mutation, and that aggregates formed can cause membrane damage and inflammation that may influence tumorigenesis.


Asunto(s)
Genes p53 , Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Cinética , Mutación , Desplegamiento Proteico , Agregado de Proteínas
12.
Molecules ; 27(4)2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35209093

RESUMEN

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.


Asunto(s)
Mutación , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Multimerización de Proteína , alfa-Sinucleína/química , alfa-Sinucleína/genética , Humanos , Enfermedades Neurodegenerativas , Agregado de Proteínas , Unión Proteica , Multimerización de Proteína/genética , Análisis Espectral , alfa-Sinucleína/metabolismo
13.
J Am Chem Soc ; 143(40): 16621-16629, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34582216

RESUMEN

The formation of amyloid fibrils and oligomers is a hallmark of several neurodegenerative disorders, including Alzheimer's disease (AD), and contributes to the disease pathway. To progress our understanding of these diseases at a molecular level, it is crucial to determine the mechanisms and rates of amyloid formation and replication. In the context of AD, the self-replication of aggregates of the Aß42 peptide by secondary nucleation, leading to the formation of new aggregates on the surfaces of existing ones, is a major source of both new fibrils and smaller toxic oligomeric species. However, the core mechanistic determinants, including the presence of intermediates, as well as the role of heterogeneities in the fibril population, are challenging to determine from bulk aggregation measurements. Here, we obtain such information by monitoring directly the time evolution of individual fibrils by TIRF microscopy. Crucially, essentially all aggregates have the ability to self-replicate via secondary nucleation, and the amplification of the aggregate concentration cannot be explained by a small fraction of "superspreader" fibrils. We observe that secondary nucleation is a catalytic multistep process involving the attachment of soluble species to the fibril surface, followed by conversion/detachment to yield a new fibril in solution. Furthermore, we find that fibrils formed by secondary nucleation resemble the parent fibril population. This detailed level of mechanistic insights into aggregate self-replication is key in the rational design of potential inhibitors of this process.


Asunto(s)
Amiloide
14.
Chembiochem ; 22(19): 2867-2871, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34383993

RESUMEN

The aggregation of α-synuclein into small soluble aggregates and then fibrils is important in the development and spreading of aggregates through the brain in Parkinson's disease. Fibrillar aggregates can grow by monomer addition and then break into fragments that could spread into neighboring cells. The rate constants for fibril elongation and fragmentation have been measured but it is not known how large an aggregate needs to be before fibril formation is thermodynamically favorable. This critical size is an important parameter controlling at what stage in an aggregation reaction fibrils can form and replicate. We determined this value to be approximately 70 monomers using super-resolution and atomic force microscopy imaging of individual α-synuclein aggregates formed in solution over long time periods. This represents the minimum size for a stable α-synuclein fibril and we hypothesis the formation of aggregates of this size in a cell represents a tipping point at which rapid replication occurs.


Asunto(s)
Amiloide/metabolismo , Encéfalo/metabolismo , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Humanos , Microscopía de Fuerza Atómica , Tamaño de la Partícula , Agregado de Proteínas , Termodinámica , alfa-Sinucleína/análisis
15.
Proc Natl Acad Sci U S A ; 115(26): E5849-E5858, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29895690

RESUMEN

The aggregation of the amyloid-ß (Aß) peptide is linked to the pathogenesis of Alzheimer's disease (AD). In particular, some point mutations within Aß are associated with early-onset familial Alzheimer's disease. Here we set out to explore how the physical properties of the altered side chains, including their sizes and charges, affect the molecular mechanisms of aggregation. We focus on Aß42 with familial mutations-A21G (Flemish), E22K (Italian), E22G (Arctic), E22Q (Dutch), and D23N (Iowa)-which lead to similar or identical pathology with sporadic AD or severe cerebral amyloid angiopathy. Through global kinetic analysis, we find that for the E22K, E22G, E22Q, and D23N mutations, the acceleration of the overall aggregation originates primarily from the modulation of the nucleation processes, in particular secondary nucleation on the surface of existing fibrils, whereas the elongation process is not significantly affected. Remarkably, the D23 position appears to be responsible for most of the charge effects during nucleation, while the size of the side chain at the E22 position plays a more significant role than its charge. Thus, we have developed a kinetic approach to determine the nature and the magnitude of the contribution of specific residues to the rate of individual steps of the aggregation reaction, through targeted mutations and variations in ionic strength. This strategy can help rationalize the effect of some disease-related mutations as well as yield insights into the mechanism of aggregation and the transition states of the wild-type protein.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Mutación Missense , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Agregación Patológica de Proteínas/genética , Enfermedad de Alzheimer/metabolismo , Sustitución de Aminoácidos , Péptidos beta-Amiloides/metabolismo , Humanos , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo
16.
Biomacromolecules ; 21(3): 1112-1125, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32011129

RESUMEN

Alzheimer's disease is associated with the deposition of the amyloid-ß peptide (Aß) into extracellular senile plaques in the brain. In vitro and in vivo observations have indicated that transthyretin (TTR) acts as an Aß scavenger in the brain, but the mechanism has not been fully resolved. We have monitored the aggregation process of Aß40 by thioflavin T fluorescence, in the presence or absence of different concentrations of preformed seed aggregates of Aß40, of wild-type tetrameric TTR (WT-TTR), and of a variant engineered to be stable as a monomer (M-TTR). Both WT-TTR and M-TTR were found to inhibit specific steps of the process of Aß40 fibril formation, which are primary and secondary nucleations, without affecting the elongation of the resulting fibrils. Moreover, the analysis shows that both WT-TTR and M-TTR bind to Aß40 oligomers formed in the aggregation reaction and inhibit their conversion into the shortest fibrils able to elongate. Using biophysical methods, TTR was found to change some aspects of its overall structure following such interactions with Aß40 oligomers, as well as with oligomers of Aß42, while maintaining its overall topology. Hence, it is likely that the predominant mechanism by which TTR exerts its protective role lies in the binding of TTR to the Aß oligomers and in inhibiting primary and secondary nucleation processes, which limits both the toxicity of Aß oligomers and the ability of the fibrils to proliferate.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/toxicidad , Humanos , Sustancias Macromoleculares , Fragmentos de Péptidos , Placa Amiloide , Prealbúmina/genética
17.
J Chem Phys ; 152(4): 045101, 2020 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-32007046

RESUMEN

The formation of amyloid fibrils from soluble peptide is a hallmark of many neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Characterization of the microscopic reaction processes that underlie these phenomena have yielded insights into the progression of such diseases and may inform rational approaches for the design of drugs to halt them. Experimental evidence suggests that most of these reaction processes are intrinsically catalytic in nature and may display enzymelike saturation effects under conditions typical of biological systems, yet a unified modeling framework accounting for these saturation effects is still lacking. In this paper, we therefore present a universal kinetic model for biofilament formation in which every fundamental process in the reaction network can be catalytic. The single closed-form expression derived is capable of describing with high accuracy a wide range of mechanisms of biofilament formation and providing the first integrated rate law of a system in which multiple reaction processes are saturated. Moreover, its unprecedented mathematical simplicity permits us to very clearly interpret the effects of increasing saturation on the overall kinetics. The effectiveness of the model is illustrated by fitting it to the data of in vitro Aß40 aggregation. Remarkably, we find that primary nucleation becomes saturated, demonstrating that it must be heterogeneous, occurring at interfaces and not in solution.


Asunto(s)
Amiloide/química , Modelos Químicos , Agregación Patológica de Proteínas , Amiloide/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Catálisis , Humanos , Cinética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo
18.
Proc Natl Acad Sci U S A ; 114(25): 6444-6449, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28584111

RESUMEN

The aggregation of the amyloid ß peptide (Aß) into amyloid fibrils is a defining characteristic of Alzheimer's disease. Because of the complexity of this aggregation process, effective therapeutic inhibitors will need to target the specific microscopic steps that lead to the production of neurotoxic species. We introduce a strategy for generating fibril-specific antibodies that selectively suppress fibril-dependent secondary nucleation of the 42-residue form of Aß (Aß42). We target this step because it has been shown to produce the majority of neurotoxic species during aggregation of Aß42. Starting from large phage display libraries of single-chain antibody fragments (scFvs), the three-stage approach that we describe includes (i) selection of scFvs with high affinity for Aß42 fibrils after removal of scFvs that bind Aß42 in its monomeric form; (ii) ranking, by surface plasmon resonance affinity measurements, of the resulting candidate scFvs that bind to the Aß42 fibrils; and (iii) kinetic screening and analysis to find the scFvs that inhibit selectively the fibril-catalyzed secondary nucleation process in Aß42 aggregation. By applying this approach, we have identified four scFvs that inhibit specifically the fibril-dependent secondary nucleation process. Our method also makes it possible to discard antibodies that inhibit elongation, an important factor because the suppression of elongation does not target directly the production of toxic oligomers and may even lead to its increase. On the basis of our results, we suggest that the method described here could form the basis for rationally designed immunotherapy strategies to combat Alzheimer's and related neurodegenerative diseases.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Anticuerpos/metabolismo , Bacteriófagos/metabolismo , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Proteínas Amiloidogénicas/metabolismo , Amiloidosis/metabolismo , Humanos , Cinética , Fragmentos de Péptidos/metabolismo , Biblioteca de Péptidos
19.
Proc Natl Acad Sci U S A ; 114(6): E1009-E1017, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28096355

RESUMEN

The self-assembly of α-synuclein is closely associated with Parkinson's disease and related syndromes. We show that squalamine, a natural product with known anticancer and antiviral activity, dramatically affects α-synuclein aggregation in vitro and in vivo. We elucidate the mechanism of action of squalamine by investigating its interaction with lipid vesicles, which are known to stimulate nucleation, and find that this compound displaces α-synuclein from the surfaces of such vesicles, thereby blocking the first steps in its aggregation process. We also show that squalamine almost completely suppresses the toxicity of α-synuclein oligomers in human neuroblastoma cells by inhibiting their interactions with lipid membranes. We further examine the effects of squalamine in a Caenorhabditis elegans strain overexpressing α-synuclein, observing a dramatic reduction of α-synuclein aggregation and an almost complete elimination of muscle paralysis. These findings suggest that squalamine could be a means of therapeutic intervention in Parkinson's disease and related conditions.


Asunto(s)
Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/prevención & control , alfa-Sinucleína/química , Algoritmos , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Productos Biológicos/química , Productos Biológicos/farmacología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Línea Celular Tumoral , Colestanoles/química , Colestanoles/farmacología , Humanos , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Estructura Molecular , Neuroblastoma/metabolismo , Neuroblastoma/patología , Paresia/genética , Paresia/metabolismo , Paresia/prevención & control , Enfermedad de Parkinson/metabolismo , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
20.
Q Rev Biophys ; 50: e6, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-29233218

RESUMEN

Parkinson's disease (PD) is characterized by proteinaceous aggregates named Lewy Bodies and Lewy Neurites containing α-synuclein fibrils. The underlying aggregation mechanism of this protein is dominated by a secondary process at mildly acidic pH, as in endosomes and other organelles. This effect manifests as a strong acceleration of the aggregation in the presence of seeds and a weak dependence of the aggregation rate on monomer concentration. The molecular mechanism underlying this process could be nucleation of monomers on fibril surfaces or fibril fragmentation. Here, we aim to distinguish between these mechanisms. The nature of the secondary processes was investigated using differential sedimentation analysis, trap and seed experiments, quartz crystal microbalance experiments and super-resolution microscopy. The results identify secondary nucleation of monomers on the fibril surface as the dominant secondary process leading to rapid generation of new aggregates, while no significant contribution from fragmentation was found. The newly generated oligomeric species quickly elongate to further serve as templates for secondary nucleation and this may have important implications in the spreading of PD.


Asunto(s)
Amiloide/química , Biocatálisis , Multimerización de Proteína , alfa-Sinucleína/química , Cinética , Imagen Molecular , Estructura Secundaria de Proteína , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA