Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Chem Phys ; 156(16): 164904, 2022 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-35490011

RESUMEN

Protein self-assembly into amyloid fibrils underlies several neurodegenerative conditions, including Alzheimer's and Parkinson's diseases. It has become apparent that the small oligomers formed during this process constitute neurotoxic molecular species associated with amyloid aggregation. Targeting the formation of oligomers represents, therefore, a possible therapeutic avenue to combat these diseases. However, it remains challenging to establish which microscopic steps should be targeted to suppress most effectively the generation of oligomeric aggregates. Recently, we have developed a kinetic model of oligomer dynamics during amyloid aggregation. Here, we use this approach to derive explicit scaling relationships that reveal how key features of the time evolution of oligomers, including oligomer peak concentration and lifetime, are controlled by the different rate parameters. We discuss the therapeutic implications of our framework by predicting changes in oligomer concentrations when the rates of the individual microscopic events are varied. Our results identify the kinetic parameters that control most effectively the generation of oligomers, thus opening a new path for the systematic rational design of therapeutic strategies against amyloid-related diseases.


Asunto(s)
Amiloide , Enfermedades Neurodegenerativas , Amiloide/metabolismo , Proteínas Amiloidogénicas , Humanos , Cinética
2.
ACS Chem Neurosci ; 12(23): 4406-4415, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34783519

RESUMEN

The self-assembly of the protein tau into neurofibrillary tangles is one of the hallmarks of Alzheimer's disease and related tauopathies. Still, the molecular mechanism of tau aggregation is largely unknown. This problem may be addressed by systematically obtaining reproducible in vitro kinetics measurements under quiescent conditions in the absence of triggering substances. Here, we implement this strategy by developing protocols for obtaining an ultrapure tau fragment (residues 304-380 of tau441) and for performing spontaneous aggregation assays with reproducible kinetics under quiescent conditions. We are thus able to identify the mechanism of fibril formation of the tau 304-380 fragment at physiological pH using fluorescence spectroscopy and mass spectrometry. We find that primary nucleation is slow, and that secondary processes dominate the aggregation process once the initial aggregates are formed. Moreover, our results further show that secondary nucleation of monomers on fibril surfaces dominates over fragmentation of fibrils. Using separate isotopes in monomers and fibrils, through mass spectroscopy measurements, we verify the isotope composition of the intermediate oligomeric species, which reveals that these small aggregates are generated from monomer through secondary nucleation. Our results provide a framework for understanding the processes leading to tau aggregation in disease and for selecting possible tau forms as targets in the development of therapeutic interventions in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Proteínas tau , Proliferación Celular , Humanos , Cinética , Ovillos Neurofibrilares/metabolismo , Agregado de Proteínas , Proteínas tau/metabolismo
3.
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.

4.
Nat Struct Mol Biol ; 28(4): 365-372, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33767451

RESUMEN

Prions consist of pathological aggregates of cellular prion protein and have the ability to replicate, causing neurodegenerative diseases, a phenomenon mirrored in many other diseases connected to protein aggregation, including Alzheimer's and Parkinson's diseases. However, despite their key importance in disease, the individual processes governing this formation of pathogenic aggregates, as well as their rates, have remained challenging to elucidate in vivo. Here we bring together a mathematical framework with kinetics of the accumulation of prions in mice and microfluidic measurements of aggregate size to dissect the overall aggregation reaction into its constituent processes and quantify the reaction rates in mice. Taken together, the data show that multiplication of prions in vivo is slower than in in vitro experiments, but efficient when compared with other amyloid systems, and displays scaling behavior characteristic of aggregate fragmentation. These results provide a framework for the determination of the mechanisms of disease-associated aggregation processes within living organisms.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Parkinson/genética , Priones/genética , Agregación Patológica de Proteínas/genética , Enfermedad de Alzheimer/patología , Amiloide/genética , Animales , Humanos , Ratones , Modelos Teóricos , Enfermedad de Parkinson/patología
5.
Nat Struct Mol Biol ; 27(12): 1125-1133, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32989305

RESUMEN

The amyloid cascade hypothesis, according to which the self-assembly of amyloid-ß peptide (Aß) is a causative process in Alzheimer's disease, has driven many therapeutic efforts for the past 20 years. Failures of clinical trials investigating Aß-targeted therapies have been interpreted as evidence against this hypothesis, irrespective of the characteristics and mechanisms of action of the therapeutic agents, which are highly challenging to assess. Here, we combine kinetic analyses with quantitative binding measurements to address the mechanism of action of four clinical stage anti-Aß antibodies, aducanumab, gantenerumab, bapineuzumab and solanezumab. We quantify the influence of these antibodies on the aggregation kinetics and on the production of oligomeric aggregates and link these effects to the affinity and stoichiometry of each antibody for monomeric and fibrillar forms of Aß. Our results reveal that, uniquely among these four antibodies, aducanumab dramatically reduces the flux of Aß oligomers.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/química , Anticuerpos Monoclonales Humanizados/química , Humanos , Cinética , Modelos Biológicos , Modelos Moleculares , Fármacos Neuroprotectores/química , Fragmentos de Péptidos/química , Mapeo Peptídico/métodos , Agregado de Proteínas/efectos de los fármacos , Conformación Proteica , Relación Estructura-Actividad
6.
Nat Chem ; 12(5): 445-451, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284577

RESUMEN

Oligomeric species populated during the aggregation of the Aß42 peptide have been identified as potent cytotoxins linked to Alzheimer's disease, but the fundamental molecular pathways that control their dynamics have yet to be elucidated. By developing a general approach that combines theory, experiment and simulation, we reveal, in molecular detail, the mechanisms of Aß42 oligomer dynamics during amyloid fibril formation. Even though all mature amyloid fibrils must originate as oligomers, we found that most Aß42 oligomers dissociate into their monomeric precursors without forming new fibrils. Only a minority of oligomers converts into fibrillar structures. Moreover, the heterogeneous ensemble of oligomeric species interconverts on timescales comparable to those of aggregation. Our results identify fundamentally new steps that could be targeted by therapeutic interventions designed to combat protein misfolding diseases.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Simulación por Computador , Humanos , Cinética , Modelos Moleculares , Fragmentos de Péptidos/química , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína
7.
Nat Chem ; 12(5): 497, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32303714

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Sci Adv ; 5(4): eaau3112, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31001578

RESUMEN

The aggregates of the Aß peptide associated with Alzheimer's disease are able to both grow in size as well as generate, through secondary nucleation, new small oligomeric species, that are major cytotoxins associated with neuronal death. Despite the importance of these amyloid fibril-dependent processes, their structural and molecular underpinnings have remained challenging to elucidate. Here, we consider two molecular chaperones: the Brichos domain, which suppresses specifically secondary nucleation processes, and clusterin which our results show is capable of inhibiting, specifically, the elongation of Aß fibrils at remarkably low substoichiometric ratios. Microfluidic diffusional sizing measurements demonstrate that this inhibition originates from interactions of clusterin with fibril ends with high affinity. Kinetic experiments in the presence of both molecular chaperones reveal that their inhibitory effects are additive and noncooperative, thereby indicating that the reactive sites associated with the formation of new aggregates and the growth of existing aggregates are distinct.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Agregado de Proteínas/fisiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Clusterina/metabolismo , Humanos , Cinética , Microfluídica , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química
9.
Nat Commun ; 10(1): 225, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30644384

RESUMEN

Transient oligomeric species formed during the aggregation process of the 42-residue form of the amyloid-ß peptide (Aß42) are key pathogenic agents in Alzheimer's disease (AD). To investigate the relationship between Aß42 aggregation and its cytotoxicity and the influence of a potential drug on both phenomena, we have studied the effects of trodusquemine. This aminosterol enhances the rate of aggregation by promoting monomer-dependent secondary nucleation, but significantly reduces the toxicity of the resulting oligomers to neuroblastoma cells by inhibiting their binding to the cellular membranes. When administered to a C. elegans model of AD, we again observe an increase in aggregate formation alongside the suppression of Aß42-induced toxicity. In addition to oligomer displacement, the reduced toxicity could also point towards an increased rate of conversion of oligomers to less toxic fibrils. The ability of a small molecule to reduce the toxicity of oligomeric species represents a potential therapeutic strategy against AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Colestanos/uso terapéutico , Fragmentos de Péptidos/metabolismo , Espermina/análogos & derivados , Péptidos beta-Amiloides/efectos de los fármacos , Animales , Caenorhabditis elegans , Línea Celular Tumoral , Colestanos/farmacología , Evaluación Preclínica de Medicamentos , Fragmentos de Péptidos/efectos de los fármacos , Espermina/farmacología , Espermina/uso terapéutico
10.
Proc Natl Acad Sci U S A ; 115(41): 10245-10250, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30257937

RESUMEN

To develop effective therapeutic strategies for protein misfolding diseases, a promising route is to identify compounds that inhibit the formation of protein oligomers. To achieve this goal, we report a structure-activity relationship (SAR) approach based on chemical kinetics to estimate quantitatively how small molecules modify the reactive flux toward oligomers. We use this estimate to derive chemical rules in the case of the amyloid beta peptide (Aß), which we then exploit to optimize starting compounds to curtail Aß oligomer formation. We demonstrate this approach by converting an inactive rhodanine compound into an effective inhibitor of Aß oligomer formation by generating chemical derivatives in a systematic manner. These results provide an initial demonstration of the potential of drug discovery strategies based on targeting directly the production of protein oligomers.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Descubrimiento de Drogas/métodos , Fragmentos de Péptidos/metabolismo , Relación Estructura-Actividad , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Humanos , Cinética , Fragmentos de Péptidos/genética , Multimerización de Proteína/efectos de los fármacos , Deficiencias en la Proteostasis/tratamiento farmacológico , Rodanina/química , Rodanina/farmacología
11.
Chem Commun (Camb) ; 54(63): 8667-8684, 2018 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-29978862

RESUMEN

Nucleation of new peptide and protein aggregates on the surfaces of amyloid fibrils of the same peptide or protein has emerged in the past two decades as a major pathway for both the generation of molecular species responsible for cellular toxicity and for the autocatalytic proliferation of peptide and protein aggregates. A key question in current research is the molecular mechanism and driving forces governing such processes, known as secondary nucleation. In this context, the analogies with other self-assembling systems for which monomer-dependent secondary nucleation has been studied for more than a century provide a valuable source of inspiration. Here, we present a short overview of this background and then review recent results regarding secondary nucleation of amyloid-forming peptides and proteins, focusing in particular on the amyloid ß peptide (Aß) from Alzheimer's disease, with some examples regarding α-synuclein from Parkinson's disease. Monomer-dependent secondary nucleation of Aß was discovered using a combination of kinetic experiments, global analysis, seeding experiments and selective isotope-enrichment, which pinpoint the monomer as the origin of new aggregates in a fibril-catalyzed reaction. Insights into driving forces are gained from variations of solution conditions, temperature and peptide sequence. Selective inhibition of secondary nucleation is explored as an effective means to limit oligomer production and toxicity. We also review experiments aimed at finding interaction partners of oligomers generated by secondary nucleation in an ongoing aggregation process. At the end of this feature article we bring forward outstanding questions and testable mechanistic hypotheses regarding monomer-dependent secondary nucleation in amyloid formation.


Asunto(s)
Amiloide/química , Amiloide/metabolismo , Humanos , Modelos Moleculares , Estructura Secundaria de Proteína
12.
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
13.
Nat Chem ; 10(5): 523-531, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29581486

RESUMEN

Mapping free-energy landscapes has proved to be a powerful tool for studying reaction mechanisms. Many complex biomolecular assembly processes, however, have remained challenging to access using this approach, including the aggregation of peptides and proteins into amyloid fibrils implicated in a range of disorders. Here, we generalize the strategy used to probe free-energy landscapes in protein folding to determine the activation energies and entropies that characterize each of the molecular steps in the aggregation of the amyloid-ß peptide (Aß42), which is associated with Alzheimer's disease. Our results reveal that interactions between monomeric Aß42 and amyloid fibrils during fibril-dependent secondary nucleation fundamentally reverse the thermodynamic signature of this process relative to primary nucleation, even though both processes generate aggregates from soluble peptides. By mapping the energetic and entropic contributions along the reaction trajectories, we show that the catalytic efficiency of Aß42 fibril surfaces results from the enthalpic stabilization of adsorbing peptides in conformations amenable to nucleation, resulting in a dramatic lowering of the activation energy for nucleation.


Asunto(s)
Péptidos beta-Amiloides/química , Biopolímeros/química , Termodinámica , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Humanos , Cinética , Pliegue de Proteína
14.
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
15.
Proc Natl Acad Sci U S A ; 114(2): E200-E208, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28011763

RESUMEN

The aggregation of the 42-residue form of the amyloid-ß peptide (Aß42) is a pivotal event in Alzheimer's disease (AD). The use of chemical kinetics has recently enabled highly accurate quantifications of the effects of small molecules on specific microscopic steps in Aß42 aggregation. Here, we exploit this approach to develop a rational drug discovery strategy against Aß42 aggregation that uses as a read-out the changes in the nucleation and elongation rate constants caused by candidate small molecules. We thus identify a pool of compounds that target specific microscopic steps in Aß42 aggregation. We then test further these small molecules in human cerebrospinal fluid and in a Caenorhabditis elegans model of AD. Our results show that this strategy represents a powerful approach to identify systematically small molecule lead compounds, thus offering an appealing opportunity to reduce the attrition problem in drug discovery.


Asunto(s)
Péptidos beta-Amiloides/química , Descubrimiento de Drogas , Fragmentos de Péptidos/química , Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Animales , Caenorhabditis elegans , Líquido Cefalorraquídeo/química , Humanos , Fragmentos de Péptidos/metabolismo , Bibliotecas de Moléculas Pequeñas
16.
Sci Adv ; 2(2): e1501244, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26933687

RESUMEN

The conversion of the ß-amyloid (Aß) peptide into pathogenic aggregates is linked to the onset and progression of Alzheimer's disease. Although this observation has prompted an extensive search for therapeutic agents to modulate the concentration of Aß or inhibit its aggregation, all clinical trials with these objectives have so far failed, at least in part because of a lack of understanding of the molecular mechanisms underlying the process of aggregation and its inhibition. To address this problem, we describe a chemical kinetics approach for rational drug discovery, in which the effects of small molecules on the rates of specific microscopic steps in the self-assembly of Aß42, the most aggregation-prone variant of Aß, are analyzed quantitatively. By applying this approach, we report that bexarotene, an anticancer drug approved by the U.S. Food and Drug Administration, selectively targets the primary nucleation step in Aß42 aggregation, delays the formation of toxic species in neuroblastoma cells, and completely suppresses Aß42 deposition and its consequences in a Caenorhabditis elegans model of Aß42-mediated toxicity. These results suggest that the prevention of the primary nucleation of Aß42 by compounds such as bexarotene could potentially reduce the risk of onset of Alzheimer's disease and, more generally, that our strategy provides a general framework for the rational identification of a range of candidate drugs directed against neurodegenerative disorders.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Antineoplásicos/farmacología , Fragmentos de Péptidos/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Tetrahidronaftalenos/farmacología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Animales , Animales Modificados Genéticamente , Bexaroteno , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Descubrimiento de Drogas/métodos , Humanos , Técnicas In Vitro , Cinética , Modelos Animales , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Multimerización de Proteína/efectos de los fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Taurina/análogos & derivados , Taurina/farmacología
17.
Phys Rev Lett ; 116(3): 038101, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26849615

RESUMEN

We establish the Hamiltonian structure of the rate equations describing the formation of protein filaments. We then show that this formalism provides a unified view of the behavior of a range of biological self-assembling systems as diverse as actin, prions, and amyloidogenic polypeptides. We further demonstrate that the time-translation symmetry of the resulting Hamiltonian leads to previously unsuggested conservation laws that connect the number and mass concentrations of fibrils and allow linear growth phenomena to be equated with autocatalytic growth processes. We finally show how these results reveal simple rate laws that provide the basis for interpreting experimental data in terms of specific mechanisms controlling the proliferation of fibrils.


Asunto(s)
Modelos Biológicos , Modelos Químicos , Proteínas/química , Amiloide/química , Amiloide/metabolismo , Dinámicas no Lineales , Proteínas/metabolismo , Termodinámica
18.
ACS Nano ; 10(1): 333-41, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26678709

RESUMEN

Characterizing the sizes and interactions of macromolecules under native conditions is a challenging problem in many areas of molecular sciences, which fundamentally arises from the polydisperse nature of biomolecular mixtures. Here, we describe a microfluidic platform for diffusional sizing based on monitoring micron-scale mass transport simultaneously in space and time. We show that the global analysis of such combined space-time data enables the hydrodynamic radii of individual species within mixtures to be determined directly by deconvoluting average signals into the contributions from the individual species. We demonstrate that the ability to perform rapid noninvasive sizing allows this method to be used to characterize interactions between biomolecules under native conditions. We illustrate the potential of the technique by implementing a single-step quantitative immunoassay that operates on a time scale of seconds and detects specific interactions between biomolecules within complex mixtures.


Asunto(s)
Inmunoensayo , Técnicas Analíticas Microfluídicas , Microfluídica/métodos , Coloración y Etiquetado/métodos , Animales , Bovinos , Difusión , Colorantes Fluorescentes/química , Glucagón/química , Proteínas HSP70 de Choque Térmico/química , Humanos , Hidrodinámica , Microfluídica/instrumentación , Peso Molecular , Albúmina Sérica Bovina/química , Anticuerpos de Dominio Único/química , Soluciones , Agua/química , alfa-Sinucleína/química , o-Ftalaldehído/química
19.
Nat Struct Mol Biol ; 22(3): 207-213, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25686087

RESUMEN

Alzheimer's disease is an increasingly prevalent neurodegenerative disorder whose pathogenesis has been associated with aggregation of the amyloid-ß peptide (Aß42). Recent studies have revealed that once Aß42 fibrils are generated, their surfaces effectively catalyze the formation of neurotoxic oligomers. Here we show that a molecular chaperone, a human Brichos domain, can specifically inhibit this catalytic cycle and limit human Aß42 toxicity. We demonstrate in vitro that Brichos achieves this inhibition by binding to the surfaces of fibrils, thereby redirecting the aggregation reaction to a pathway that involves minimal formation of toxic oligomeric intermediates. We verify that this mechanism occurs in living mouse brain tissue by cytotoxicity and electrophysiology experiments. These results reveal that molecular chaperones can help maintain protein homeostasis by selectively suppressing critical microscopic steps within the complex reaction pathways responsible for the toxic effects of protein misfolding and aggregation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Chaperonas Moleculares/fisiología , Agregación Patológica de Proteínas , Enfermedad de Alzheimer/metabolismo , Animales , Microscopía por Crioelectrón , Electrofisiología , Femenino , Hipocampo/metabolismo , Hipocampo/fisiología , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Pliegue de Proteína , Estructura Terciaria de Proteína
20.
Health Aff (Millwood) ; 33(9): 1507-15, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25201654

RESUMEN

Accountable care--a way to align health care payments with patient-focused reform goals--is currently being pursued in the United States, but its principles are also being applied in many other countries. In this article we review experiences with such reforms to offer a globally applicable definition of an accountable care system and propose a conceptual framework for characterizing and assessing accountable care reforms. The framework consists of five components: population, outcomes, metrics and learning, payments and incentives, and coordinated delivery. We describe how the framework applies to accountable care reforms that are already being implemented in Spain and Singapore. We also describe how it can be used to map progress through increasingly sophisticated levels of reforms. We recommend that policy makers pursuing accountable care reforms emphasize the following steps: highlight population health and wellness instead of just treating illness; pay for outcomes instead of activities; create a more favorable environment for collaboration and coordinated care; and promote interoperable data systems.


Asunto(s)
Organizaciones Responsables por la Atención/organización & administración , Salud Global , Reforma de la Atención de Salud/organización & administración , Organizaciones Responsables por la Atención/economía , Reforma de la Atención de Salud/economía , Política de Salud , Humanos , Modelos Organizacionales , Calidad de la Atención de Salud
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA