Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
ACS Chem Neurosci ; 15(6): 1125-1134, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38416693

RESUMEN

Oligomeric assemblies of the amyloid ß peptide (Aß) have been investigated for over two decades as possible neurotoxic agents in Alzheimer's disease. However, due to their heterogeneous and transient nature, it is not yet fully established which of the structural features of these oligomers may generate cellular damage. Here, we study distinct oligomer species formed by Aß40 (the 40-residue form of Aß) in the presence of four different metal ions (Al3+, Cu2+, Fe2+, and Zn2+) and show that they differ in their structure and toxicity in human neuroblastoma cells. We then describe a correlation between the size of the oligomers and their neurotoxic activity, which provides a type of structure-toxicity relationship for these Aß40 oligomer species. These results provide insight into the possible role of metal ions in Alzheimer's disease by the stabilization of Aß oligomers.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Péptidos beta-Amiloides/química , Metales , Iones , Fragmentos de Péptidos/química
3.
Mol Neurodegener ; 19(1): 20, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378578

RESUMEN

The conversion of native peptides and proteins into amyloid aggregates is a hallmark of over 50 human disorders, including Alzheimer's and Parkinson's diseases. Increasing evidence implicates misfolded protein oligomers produced during the amyloid formation process as the primary cytotoxic agents in many of these devastating conditions. In this review, we analyze the processes by which oligomers are formed, their structures, physicochemical properties, population dynamics, and the mechanisms of their cytotoxicity. We then focus on drug discovery strategies that target the formation of oligomers and their ability to disrupt cell physiology and trigger degenerative processes.


Asunto(s)
Enfermedad de Parkinson , Deficiencias en la Proteostasis , Humanos , Amiloide/metabolismo , Enfermedad de Parkinson/metabolismo , Péptidos beta-Amiloides
4.
J Med Chem ; 66(14): 9519-9536, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37433124

RESUMEN

Natural aminosterols are promising drug candidates against neurodegenerative diseases, like Alzheimer and Parkinson, and one relevant protective mechanism occurs via their binding to biological membranes and displacement or binding inhibition of amyloidogenic proteins and their cytotoxic oligomers. We compared three chemically different aminosterols, finding that they exhibited different (i) binding affinities, (ii) charge neutralizations, (iii) mechanical reinforcements, and (iv) key lipid redistributions within membranes of reconstituted liposomes. They also had different potencies (EC50) in protecting cultured cell membranes against amyloid-ß oligomers. A global fitting analysis led to an analytical equation describing quantitatively the protective effects of aminosterols as a function of their concentration and relevant membrane effects. The analysis correlates aminosterol-mediated protection with well-defined chemical moieties, including the polyamine group inducing a partial membrane-neutralizing effect (79 ± 7%) and the cholestane-like tail causing lipid redistribution and bilayer mechanical resistance (21 ± 7%), linking quantitatively their chemistry to their protective effects on biological membranes.


Asunto(s)
Enfermedades Neurodegenerativas , Agregado de Proteínas , Humanos , Membrana Celular/metabolismo , Proteínas Amiloidogénicas/química , Enfermedades Neurodegenerativas/metabolismo , Lípidos , Membrana Dobles de Lípidos/metabolismo , Péptidos beta-Amiloides/metabolismo
5.
Acc Chem Res ; 56(12): 1395-1405, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37071750

RESUMEN

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


Asunto(s)
Enfermedad de Alzheimer , Deficiencias en la Proteostasis , Animales , Escherichia coli/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Amiloide/química
6.
ACS Chem Neurosci ; 14(4): 657-666, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36728544

RESUMEN

Alzheimer's disease is characterized by the presence in the brain of amyloid plaques formed by the aberrant deposition of the amyloid-ß peptide (Aß). Since many vitamins are dysregulated in this disease, we explored whether these molecules contribute to the protein homeostasis system by modulating Aß aggregation. By screening 18 fat-soluble and water-soluble vitamin metabolites, we found that retinoic acid and α-tocopherol, two metabolites of vitamin A and vitamin E, respectively, affect Aß aggregation both in vitro and in a Caenorhabditis elegans model of Aß toxicity. We then show that the effects of these two vitamin metabolites in specific combinations cancel each other out, consistent with the "resilience in complexity" hypothesis, according to which the complex composition of the cellular environment could have an overall protective role against protein aggregation through the simultaneous presence of aggregation promoters and inhibitors. Taken together, these results indicate that vitamins can be added to the list of components of the protein homeostasis system that regulate protein aggregation.


Asunto(s)
Enfermedad de Alzheimer , Vitamina A , Animales , Vitamina E/farmacología , Vitamina E/metabolismo , Agregado de Proteínas , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/metabolismo , Vitaminas/farmacología , Vitaminas/metabolismo , Vitamina K/metabolismo , Caenorhabditis elegans
7.
Chem Biol Interact ; 371: 110307, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36535315

RESUMEN

Natural proteinaceous pore-forming agents can bind and permeabilize cell membranes, leading to ion dyshomeostasis and cell death. In the search for antidotes that can protect cells from peptide toxins, we discovered that the polyphenol epigallocatechin gallate (EGCG) interacts directly with melittin from honeybee venom, resulting in the elimination of its binding to the cell membrane and toxicity by markedly lowering the extent of its solvent-exposed hydrophobicity and promoting its oligomerization into larger species. These physicochemical parameters have also been shown to play a key role in the binding to cells of misfolded protein oligomers in a host of neurodegenerative diseases, where oligomer-membrane binding and associated toxicity have been shown to correlate negatively with oligomer size and positively with solvent-exposed hydrophobicity. For melittin, which is not an amyloid-forming protein and has a very distinct mechanism of toxicity compared to misfolded oligomers, we find that the size-hydrophobicity-toxicity relationship also rationalizes the pharmacological attenuation of melittin toxicity by EGCG. These results highlight the importance of the physicochemical properties of pore forming agents in mediating their interactions with cell membranes and suggest a possible therapeutic approach based on compounds with a similar mechanism of action as EGCG.


Asunto(s)
Catequina , Meliteno , Catequina/farmacología , Catequina/química , Interacciones Hidrofóbicas e Hidrofílicas , Meliteno/farmacología , Solventes , Venenos de Abeja , Animales
8.
ACS Chem Neurosci ; 13(8): 1219-1231, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35404569

RESUMEN

The molecular composition of the plasma membrane plays a key role in mediating the susceptibility of cells to perturbations induced by toxic molecules. The pharmacological regulation of the properties of the cell membrane has therefore the potential to enhance cellular resilience to a wide variety of chemical and biological compounds. In this study, we investigate the ability of claramine, a blood-brain barrier permeable small molecule in the aminosterol class, to neutralize the toxicity of acute biological threat agents, including melittin from honeybee venom and α-hemolysin from Staphylococcus aureus. Our results show that claramine neutralizes the toxicity of these pore-forming agents by preventing their interactions with cell membranes without perturbing their structures in a detectable manner. We thus demonstrate that the exogenous administration of an aminosterol can tune the properties of lipid membranes and protect cells from diverse biotoxins, including not just misfolded protein oligomers as previously shown but also biological protein-based toxins. Our results indicate that the investigation of regulators of the physicochemical properties of cell membranes offers novel opportunities to develop countermeasures against an extensive set of cytotoxic effects associated with cell membrane disruption.


Asunto(s)
Encéfalo , Transporte Biológico , Membrana Celular
9.
Nat Prod Rep ; 39(4): 742-753, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-34698757

RESUMEN

Covering: 1993 to 2021 (mainly 2017-2021)Alzheimer's and Parkinson's diseases are neurodegenerative conditions affecting over 50 million people worldwide. Since these disorders are still largely intractable pharmacologically, discovering effective treatments is of great urgency and importance. These conditions are characteristically associated with the aberrant deposition of proteinaceous aggregates in the brain, and with the formation of metastable intermediates known as protein misfolded oligomers that play a central role in their aetiology. In this Highlight article, we review the evidence at the physicochemical, cellular, animal model and clinical levels on how the natural products squalamine and trodusquemine offer promising opportunities for chronic treatments for these progressive conditions by preventing both the formation of neurotoxic oligomers and their interaction with cell membranes.


Asunto(s)
Enfermedad de Alzheimer , Productos Biológicos , Enfermedades Neurodegenerativas , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Productos Biológicos/farmacología , Química Física , Colestanos , Colestanoles , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Espermina/análogos & derivados
10.
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.

11.
Commun Biol ; 4(1): 843, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234268

RESUMEN

Age-related changes in cellular metabolism can affect brain homeostasis, creating conditions that are permissive to the onset and progression of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Although the roles of metabolites have been extensively studied with regard to cellular signaling pathways, their effects on protein aggregation remain relatively unexplored. By computationally analysing the Human Metabolome Database, we identified two endogenous metabolites, carnosine and kynurenic acid, that inhibit the aggregation of the amyloid beta peptide (Aß) and rescue a C. elegans model of Alzheimer's disease. We found that these metabolites act by triggering a cytosolic unfolded protein response through the transcription factor HSF-1 and downstream chaperones HSP40/J-proteins DNJ-12 and DNJ-19. These results help rationalise previous observations regarding the possible anti-ageing benefits of these metabolites by providing a mechanism for their action. Taken together, our findings provide a link between metabolite homeostasis and protein homeostasis, which could inspire preventative interventions against neurodegenerative disorders.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Caenorhabditis elegans/metabolismo , Carnosina/metabolismo , Modelos Animales de Enfermedad , Ácido Quinurénico/metabolismo , Respuesta de Proteína Desplegada/fisiología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Carnosina/farmacología , Citosol/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Ácido Quinurénico/farmacología , Agregado de Proteínas , Agregación Patológica de Proteínas/prevención & control , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
13.
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.

14.
Sci Adv ; 6(45)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33148639

RESUMEN

Disordered proteins are challenging therapeutic targets, and no drug is currently in clinical use that modifies the properties of their monomeric states. Here, we identify a small molecule (10074-G5) capable of binding and sequestering the intrinsically disordered amyloid-ß (Aß) peptide in its monomeric, soluble state. Our analysis reveals that this compound interacts with Aß and inhibits both the primary and secondary nucleation pathways in its aggregation process. We characterize this interaction using biophysical experiments and integrative structural ensemble determination methods. We observe that this molecule increases the conformational entropy of monomeric Aß while decreasing its hydrophobic surface area. We also show that it rescues a Caenorhabditis elegans model of Aß-associated toxicity, consistent with the mechanism of action identified from the in silico and in vitro studies. These results illustrate the strategy of stabilizing the monomeric states of disordered proteins with small molecules to alter their behavior for therapeutic purposes.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Descubrimiento de Drogas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Fragmentos de Péptidos/metabolismo
15.
Int J Mol Sci ; 21(22)2020 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-33212787

RESUMEN

The aberrant aggregation of proteins is implicated in the onset and pathogenesis of a wide range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Mounting evidence indicates that misfolded protein oligomers produced as intermediates in the aggregation process are potent neurotoxic agents in these diseases. Because of the transient and heterogeneous nature of these elusive aggregates, however, it has proven challenging to develop therapeutics that can effectively target them. Here, we review approaches aimed at reducing oligomer toxicity, including (1) modulating the oligomer populations (e.g., by altering the kinetics of aggregation by inhibiting, enhancing, or redirecting the process), (2) modulating the oligomer properties (e.g., through the size-hydrophobicity-toxicity relationship), (3) modulating the oligomer interactions (e.g., by protecting cell membranes by displacing oligomers), and (4) reducing oligomer toxicity by potentiating the protein homeostasis system. We analyze examples of these complementary approaches, which may lead to the development of compounds capable of preventing or treating neurodegenerative disorders associated with protein aggregation.


Asunto(s)
Agregación Patológica de Proteínas/terapia , Multimerización de Proteína , Deficiencias en la Proteostasis/terapia , Animales , Humanos , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Deficiencias en la Proteostasis/metabolismo , Deficiencias en la Proteostasis/patología
16.
Mil Med ; 185(11-12): e2104-e2109, 2020 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-32870979

RESUMEN

INTRODUCTION: Military installations are at increased risk for the transmission of infectious disease. Personnel who live and train on military installations live and train near one another facilitating disease transmission. An understanding of historical sanitation and hygiene can inform modern practices. This is especially pertinent considering the continuing rise of variants of infectious diseases, such as the recent pandemic of the 2019 severe acute respiratory syndrome coronavirus 2. In this article, we review the rise and decline of infectious disease at the United States Military Academy (USMA) during the period spanning 1890 through 1910, and the public health interventions used to combat disease spread. MATERIALS AND METHODS: Primary data regarding cadet illness were acquired from the historical archives of the USMA. These included annual reports, clinical admission records, casualty ledgers, and sanitation reports. Unpublished documents from the medical history of USMA provide periodic trends of health among cadets because of infectious disease. RESULTS: Between 1890 and 1910, the USMA at West Point was confronted with cases of influenza, measles, mumps, scarlet fever, smallpox, typhus, and malaria. In response, a series of non-pharmaceutical interventions (NPIs) were instituted to curb the spread of infectious disease. These interventions most likely proved effective in suppressing the transmission of communicable diseases. The most common and arguably the most effective NPI was the physical separation of the sick from the well. CONCLUSIONS: The USMA experience mirrored what was occurring in the larger U.S. Army in the early 20th century and may serve as a model for the application of NPIs in response to modern infectious diseases resulting from novel or unknown etiologies.


Asunto(s)
Academias e Institutos/estadística & datos numéricos , Higiene Militar/normas , Medicina Militar/métodos , Academias e Institutos/historia , Academias e Institutos/organización & administración , Historia del Siglo XIX , Historia del Siglo XX , Humanos , Gripe Humana/epidemiología , Gripe Humana/historia , Malaria/epidemiología , Malaria/historia , Sarampión/epidemiología , Sarampión/historia , Higiene Militar/historia , Personal Militar/educación , Personal Militar/historia , Personal Militar/estadística & datos numéricos , Paperas/epidemiología , Paperas/historia , Escarlatina/epidemiología , Escarlatina/historia , Viruela/epidemiología , Viruela/historia , Tifus Epidémico Transmitido por Piojos/epidemiología , Tifus Epidémico Transmitido por Piojos/historia , Estados Unidos/epidemiología
17.
Sci Rep ; 10(1): 15280, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943652

RESUMEN

Bicyclic peptides have great therapeutic potential since they can bridge the gap between small molecules and antibodies by combining a low molecular weight of about 2 kDa with an antibody-like binding specificity. Here we apply a recently developed in silico rational design strategy to produce a bicyclic peptide to target the C-terminal region (residues 31-42) of the 42-residue form of the amyloid ß peptide (Aß42), a protein fragment whose aggregation into amyloid plaques is linked with Alzheimer's disease. We show that this bicyclic peptide is able to remodel the aggregation process of Aß42 in vitro and to reduce its associated toxicity in vivo in a C. elegans worm model expressing Aß42. These results provide an initial example of a computational approach to design bicyclic peptides to target specific epitopes on disordered proteins.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Caenorhabditis elegans/metabolismo , Agregación Patológica de Proteínas/metabolismo , Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Fragmentos de Péptidos , Placa Amiloide/metabolismo
18.
Commun Biol ; 3(1): 435, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792544

RESUMEN

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


Asunto(s)
Membrana Celular/metabolismo , Colestanos/farmacología , Pliegue de Proteína , Multimerización de Proteína , Espermina/análogos & derivados , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Fenómenos Biofísicos/efectos de los fármacos , Transferasas de Carboxilo y Carbamoilo/química , Transferasas de Carboxilo y Carbamoilo/toxicidad , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidad , Humanos , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Espermina/farmacología , alfa-Sinucleína/química , alfa-Sinucleína/toxicidad
19.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630615

RESUMEN

Alzheimer's disease is associated with the aggregation of the amyloid-ß peptide (Aß), resulting in the deposition of amyloid plaques in brain tissue. Recent scrutiny of the mechanisms by which Aß aggregates induce neuronal dysfunction has highlighted the importance of the Aß oligomers of this protein fragment. Because of the transient and heterogeneous nature of these oligomers, however, it has been challenging to investigate the detailed mechanisms by which these species exert cytotoxicity. To address this problem, we demonstrate here the use of rationally designed single-domain antibodies (DesAbs) to characterize the structure-toxicity relationship of Aß oligomers. For this purpose, we use Zn2+-stabilized oligomers of the 40-residue form of Aß (Aß40) as models of brain Aß oligomers and two single-domain antibodies (DesAb18-24 and DesAb34-40), designed to bind to epitopes at residues 18-24 and 34-40 of Aß40, respectively. We found that the DesAbs induce a change in structure of the Zn2+-stabilized Aß40 oligomers, generating a simultaneous increase in their size and solvent-exposed hydrophobicity. We then observed that these increments in both the size and hydrophobicity of the oligomers neutralize each other in terms of their effects on cytotoxicity, as predicted by a recently proposed general structure-toxicity relationship, and observed experimentally. These results illustrate the use of the DesAbs as research tools to investigate the biophysical and cytotoxicity properties of Aß oligomers.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Anticuerpos/inmunología , Anticuerpos/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Formación de Anticuerpos/inmunología , Encéfalo/metabolismo , Diseño de Fármacos , Humanos , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Agregado de Proteínas/fisiología , Ingeniería de Proteínas/métodos , Relación Estructura-Actividad
20.
Proc Natl Acad Sci U S A ; 117(24): 13509-13518, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32493749

RESUMEN

Protein misfolding and aggregation is the hallmark of numerous human disorders, including Alzheimer's disease. This process involves the formation of transient and heterogeneous soluble oligomers, some of which are highly cytotoxic. A major challenge for the development of effective diagnostic and therapeutic tools is thus the detection and quantification of these elusive oligomers. Here, to address this problem, we develop a two-step rational design method for the discovery of oligomer-specific antibodies. The first step consists of an "antigen scanning" phase in which an initial panel of antibodies is designed to bind different epitopes covering the entire sequence of a target protein. This procedure enables the determination through in vitro assays of the regions exposed in the oligomers but not in the fibrillar deposits. The second step involves an "epitope mining" phase, in which a second panel of antibodies is designed to specifically target the regions identified during the scanning step. We illustrate this method in the case of the amyloid ß (Aß) peptide, whose oligomers are associated with Alzheimer's disease. Our results show that this approach enables the accurate detection and quantification of Aß oligomers in vitro, and in Caenorhabditis elegans and mouse hippocampal tissues.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Anticuerpos/inmunología , Agregado de Proteínas , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Animales , Anticuerpos/química , Anticuerpos/metabolismo , Especificidad de Anticuerpos , Caenorhabditis elegans , Modelos Animales de Enfermedad , Epítopos , Hipocampo/metabolismo , Ratones , Unión Proteica , Conformación Proteica , Anticuerpos de Dominio Único
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...