Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 19732, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34611196

RESUMEN

Aggregation of proteins is a prominent hallmark of virtually all neurodegenerative disorders including Alzheimer's, Parkinson's and Huntington's diseases. Little progress has been made in their treatment to slow or prevent the formation of aggregates by post-translational modification and regulation of cellular responses to misfolded proteins. Here, we introduce a label-free, laser-based photothermal treatment of polyglutamine (polyQ) aggregates in a C. elegans nematode model of huntingtin-like polyQ aggregation. As a proof of principle, we demonstrated that nanosecond laser pulse-induced local photothermal heating can directly disrupt the aggregates so as to delay their accumulation, maintain motility, and extend the lifespan of treated nematodes. These beneficial effects were validated by confocal photothermal, fluorescence, and video imaging. The results obtained demonstrate that our theranostics platform, integrating photothermal therapy without drugs or other chemicals, combined with advanced imaging to monitor photothermal ablation of aggregates, initiates systemic recovery and thus validates the concept of aggregate-disruption treatments for neurodegenerative diseases in humans.


Asunto(s)
Enfermedad de Huntington/etiología , Enfermedad de Huntington/metabolismo , Agregado de Proteínas/efectos de la radiación , Agregación Patológica de Proteínas/metabolismo , Animales , Caenorhabditis elegans , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/patología , Enfermedad de Huntington/terapia , Rayos Láser , Terapia por Luz de Baja Intensidad , Péptidos/metabolismo , Terapia Fototérmica , Agregación Patológica de Proteínas/terapia , Proteínas Recombinantes de Fusión/metabolismo
2.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166234, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34339840

RESUMEN

TAR DNA-binding protein-43 (TDP-43) pathology, including fibrillar aggregates and mutations, develops in amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD) and limbic-predominant age-related TDP-43 encephalopathy (LATE). Hyperphosphorylation and aggregation of TDP-43 contribute to pathology and are viable therapeutic targets for ALS. In vivo inhibition of TDP-43 aggregation was evaluated using anti-TDP-43 antibodies with promising outcomes. However, the exact mechanism of antibody-based inhibition targeting TDP-43 is not well understood but may lead to the identification of viable immunotherapies. Herein, the mechanism of in vitro aggregation of phosphorylated TDP-43 was explored, and the anti-TDP-43 antibodies tested for their inhibitor efficacies. Specifically, the aggregation of phosphorylated full-length TDP-43 protein (pS410) was monitored by transmission electron microscopy (TEM), turbidity absorbance, and thioflavin (ThT) spectroscopy. The protein aggregates were insoluble, ThT-positive and characterized with heterogeneous morphologies (fibers, amorphous structures). Antibodies specific to epitopes 178-393 and 256-269, within the RRM2-CTD domain, reduced the formation of ß-sheets and insoluble aggregates, at low antibody loading (antibody: protein ratio = 1 µg/mL: 45 µg/mL). Inhibition outcomes were highly dependent on the type and loading of antibodies, indicating dual functionality of such inhibitors, as aggregation inhibitors or aggregation promoters. Anti-SOD1 and anti-tau antibodies were not effective inhibitors against TDP-43 aggregation, indicating selective inhibition.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Anticuerpos Antiidiotipos/inmunología , Encefalopatías/genética , Proteínas de Unión al ADN/genética , Degeneración Lobar Frontotemporal/genética , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/terapia , Encefalopatías/inmunología , Encefalopatías/patología , Encefalopatías/terapia , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/inmunología , Epítopos/inmunología , Degeneración Lobar Frontotemporal/inmunología , Degeneración Lobar Frontotemporal/patología , Degeneración Lobar Frontotemporal/terapia , Humanos , Microscopía Electrónica de Transmisión , Fosforilación/genética , Agregado de Proteínas/genética , Agregado de Proteínas/inmunología , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/inmunología , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/terapia , Conformación Proteica en Lámina beta/genética , Superóxido Dismutasa-1/antagonistas & inhibidores , Superóxido Dismutasa-1/inmunología , Proteínas tau/antagonistas & inhibidores , Proteínas tau/inmunología
3.
Molecules ; 26(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34299575

RESUMEN

The aberrant aggregation of amyloid-ß (Aß) peptides in the brain has been recognized as the major hallmark of Alzheimer's disease (AD). Thus, the inhibition and dissociation of Aß aggregation are believed to be effective therapeutic strategiesforthe prevention and treatment of AD. When integrated with traditional agents and biomolecules, nanomaterials can overcome their intrinsic shortcomings and boost their efficiency via synergistic effects. This article provides an overview of recent efforts to utilize nanomaterials with superior properties to propose effective platforms for AD treatment. The underlying mechanismsthat are involved in modulating Aß aggregation are discussed. The summary of nanomaterials-based modulation of Aß aggregation may help researchers to understand the critical roles in therapeutic agents and provide new insight into the exploration of more promising anti-amyloid agents and tactics in AD theranostics.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Nanoestructuras/uso terapéutico , Agregación Patológica de Proteínas/terapia , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Animales , Humanos , Nanomedicina , Nanoestructuras/química , Terapia Fototérmica , Agregación Patológica de Proteínas/metabolismo
4.
Mol Neurobiol ; 58(7): 3095-3118, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33629274

RESUMEN

Spinocerebellar ataxia type 3 (SCA3) is the most common type of SCA worldwide caused by abnormal polyglutamine expansion in the coding region of the ataxin-3 gene. Ataxin-3 is a multi-faceted protein involved in various cellular processes such as deubiquitination, cytoskeletal organisation, and transcriptional regulation. The presence of an expanded poly(Q) stretch leads to altered processing and misfolding of the protein culminating in the production of insoluble protein aggregates in the cell. Various post-translational modifications affect ataxin-3 fibrillation and aggregation. This review provides an exhaustive assessment of the various pathogenic mechanisms undertaken by the mutant ataxin-3-containing aggregates (MATAGGs) for disease induction and neurodegeneration. This includes in-depth discussion on MATAGG dynamics including their formation, role in neuronal pathogenesis, and the debate over the toxic v/s protective nature of the MATAGGs in disease progression. Additionally, the currently available therapeutic strategies against SCA3 have been reviewed. The shift in the focus of such strategies, from targeting the steps that lead to or reduce aggregate formation to targeting the expression of mutant ataxin-3 itself via RNA-based therapeutics, has also been presented. We also discuss the intriguing promise that various growth and neurotrophic factors, especially the insulin pathway, hold in the modulation of SCA3 progression. These emerging areas show the newer directions through which SCA3 can be targeted including various preclinical and clinical trials. All these advances made in the last three decades since the discovery of the ataxin-3 gene have been critically reviewed here.


Asunto(s)
Ataxina-3/genética , Ataxina-3/metabolismo , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/metabolismo , Mutación/fisiología , Animales , Daño del ADN/fisiología , Terapia Genética/métodos , Terapia Genética/tendencias , Humanos , Enfermedad de Machado-Joseph/terapia , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/terapia
5.
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
6.
J Biol Chem ; 295(30): 10224-10244, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32424039

RESUMEN

α-Synuclein (αsyn) is an abundant brain neuronal protein that can misfold and polymerize to form toxic fibrils coalescing into pathologic inclusions in neurodegenerative diseases, including Parkinson's disease, Lewy body dementia, and multiple system atrophy. These fibrils may induce further αsyn misfolding and propagation of pathologic fibrils in a prion-like process. It is unclear why αsyn initially misfolds, but a growing body of literature suggests a critical role of partial proteolytic processing resulting in various truncations of the highly charged and flexible carboxyl-terminal region. This review aims to 1) summarize recent evidence that disease-specific proteolytic truncations of αsyn occur in Parkinson's disease, Lewy body dementia, and multiple system atrophy and animal disease models; 2) provide mechanistic insights on how truncation of the amino and carboxyl regions of αsyn may modulate the propensity of αsyn to pathologically misfold; 3) compare experiments evaluating the prion-like properties of truncated forms of αsyn in various models with implications for disease progression; 4) assess uniquely toxic properties imparted to αsyn upon truncation; and 5) discuss pathways through which truncated αsyn forms and therapies targeted to interrupt them. Cumulatively, it is evident that truncation of αsyn, particularly carboxyl truncation that can be augmented by dysfunctional proteostasis, dramatically potentiates the propensity of αsyn to pathologically misfold into uniquely toxic fibrils with modulated prion-like seeding activity. Therapeutic strategies and experimental paradigms should operate under the assumption that truncation of αsyn is likely occurring in both initial and progressive disease stages, and preventing truncation may be an effective preventative strategy against pathologic inclusion formation.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Agregación Patológica de Proteínas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/terapia , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/terapia , alfa-Sinucleína/genética
7.
Cell Mol Neurobiol ; 40(3): 313-345, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31584139

RESUMEN

Neurodegeneration entails progressive loss of neuronal structure as well as function leading to cognitive failure, apathy, anxiety, irregular body movements, mood swing and ageing. Proteomic dysregulation is considered the key factor for neurodegeneration. Mechanisms involving deregulated processing of proteins such as amyloid beta (Aß) oligomerization; tau hyperphosphorylation, prion misfolding; α-synuclein accumulation/lewy body formation, chaperone deregulation, acetylcholine depletion, adenosine 2A (A2A) receptor hyperactivation, secretase deregulation, leucine-rich repeat kinase 2 (LRRK2) mutation and mitochondrial proteinopathies have deeper implications in neurodegenerative disorders. Better understanding of such pathological mechanisms is pivotal for exploring crucial drug targets. Herein, we provide a comprehensive outlook about the diverse proteomic irregularities in Alzheimer's, Parkinson's and Creutzfeldt Jakob disease (CJD). We explicate the role of key neuroproteomic drug targets notably Aß, tau, alpha synuclein, prions, secretases, acetylcholinesterase (AchE), LRRK2, molecular chaperones, A2A receptors, muscarinic acetylcholine receptors (mAchR), N-methyl-D-aspartate receptor (NMDAR), glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) and mitochondrial/oxidative stress-related proteins for combating neurodegeneration and associated cognitive and motor impairment. Cross talk between amyloidopathy, synucleinopathy, tauopathy and several other proteinopathies pinpoints the need to develop safe therapeutics with ability to strike multiple targets in the aetiology of the neurodegenerative disorders. Therapeutics like microtubule stabilisers, chaperones, kinase inhibitors, anti-aggregation agents and antibodies could serve promising regimens for treating neurodegeneration. However, drugs should be target specific, safe and able to penetrate blood-brain barrier.


Asunto(s)
Terapia Molecular Dirigida , Degeneración Nerviosa/metabolismo , Agregación Patológica de Proteínas/metabolismo , Proteoma/análisis , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Síndrome de Creutzfeldt-Jakob/metabolismo , Síndrome de Creutzfeldt-Jakob/patología , Síndrome de Creutzfeldt-Jakob/fisiopatología , Humanos , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/fisiopatología , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/fisiopatología , Agregación Patológica de Proteínas/terapia , Proteoma/metabolismo , Proteómica , Transducción de Señal/fisiología
8.
Neurobiol Dis ; 132: 104543, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31351173

RESUMEN

α-Synuclein (αS) and tau have a lot in common. Dyshomeostasis and aggregation of both proteins are central in the pathogenesis of neurodegenerative diseases: Parkinson's disease, dementia with Lewy bodies, multi-system atrophy and other 'synucleinopathies' in the case of αS; Alzheimer's disease, frontotemporal dementia, progressive supranuclear palsy and other 'tauopathies' in the case of tau. The aggregated states of αS and tau are found to be (hyper)phosphorylated, but the relevance of the phosphorylation in health or disease is not well understood. Both tau and αS are typically characterized as 'intrinsically disordered' proteins, while both engage in transient interactions with cellular components, thereby undergoing structural changes and context-specific folding. αS transiently binds to (synaptic) vesicles forming a membrane-induced amphipathic helix; tau transiently interacts with microtubules forming an 'extended structure'. The regulation and exact nature of the interactions are not fully understood. Here we review recent and previous insights into the dynamic, transient nature of αS and tau with regard to the mode of interaction with their targets, the dwell-time while bound, and the cis and trans factors underlying the frequent switching between bound and unbound states. These aspects are intimately linked to hypotheses on how subtle changes in the transient behaviors may trigger the earliest steps in the pathogenesis of the respective brain diseases. Based on a deeper understanding of transient αS and tau conformations in the cellular context, new therapeutic strategies may emerge, and it may become clearer why existing approaches have failed or how they could be optimized.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Humanos , Enfermedades Neurodegenerativas/terapia , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/terapia , Pliegue de Proteína
9.
ACS Chem Neurosci ; 10(5): 2250-2262, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30707008

RESUMEN

The aggregation of ß-amyloid peptides is a key event in the formative stages of Alzheimer's disease. Promoting folding and inhibiting aggregation was reported as an effective strategy in reducing Aß-elicited toxicity. This study experimentally investigates the influence of the external electric field (EF) and magnetic field (MF) of varying strengths on the in vitro fibrillogenesis of hydrophobic core sequence, Aß16-22, and its parent peptide, Aß1-42. Biophysical methods such as ThT fluorescence, static light scattering, circular dichroism, and infrared spectroscopy suggest that EF has a stabilizing effect on the secondary structure, initiating a conformational switch of Aß16-22 and Aß1-42 from ß to non-ß conformation. This observation was further corroborated by dynamic light scattering and transmission electron microscopic studies. To mimic in vivo conditions, we repeated ThT fluorescence assay with Aß1-42 in human cerebrospinal fluid to verify EF-mediated modulation. The self-seeding of Aß1-42 and cross-seeding with Aß1-40 to verify that the autocatalytic amplification of self-assembly as a result of secondary nucleation also yields comparable results in EF-exposed and unexposed samples. Aß-elicited toxicity of EF-treated samples in two neuroblastoma cell lines (SH-SY5Y and IMR-32) and human embryonic kidney cell line (HEK293) were found to be 15-38% less toxic than the EF untreated ones under identical conditions. Experiments with fluorescent labeled Aß1-42 to correlate reduced cytotoxicity and cell internalization suggest a comparatively smaller uptake of the EF-treated peptides. Our results provide a scientific roadmap for future noninvasive, therapeutic solutions for the treatment of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Agregación Patológica de Proteínas/terapia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Línea Celular Tumoral , Dicroismo Circular , Células HEK293 , Humanos , Neuronas/metabolismo , Neuronas/patología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología
10.
Nano Lett ; 19(2): 674-683, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30444372

RESUMEN

Alzheimer's disease (AD) is a progressive and irreversible brain disorder. Recent studies revealed the pivotal role of ß-amyloid (Aß) in AD. However, there is no conclusive indication that the existing therapeutic strategies exerted any effect on the mitigation of Aß-induced neurotoxicity and the elimination of Aß aggregates simultaneously in vivo. Herein, we developed a novel nanocomposite that can eliminate toxic Aß aggregates and mitigate Aß-induced neurotoxicity in AD mice. This nanocomposite was designed to be a small-sized particle (14 ± 4 nm) with Aß-binding peptides (KLVFF) integrated on the surface. The nanocomposite was prepared by wrapping a protein molecule with a cross-linked KLVFF-containing polymer layer synthesized by in situ polymerization. The presence of the nanocomposite remarkably changed the morphology of Aß aggregates, which led to the formation of Aß/nanocomposite coassembled nanoclusters instead of Aß oligomers. With the reduction of the pathological Aß oligomers, the nanocomposites attenuated the Aß-induced neuron damages, regained endocranial microglia's capability to phagocytose Aß, and eventually protected hippocampal neurons against apoptosis. Thus, we anticipate that the small-sized nanocomposite will potentially offer a feasible strategy in the development of novel AD treatments.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Nanocompuestos/uso terapéutico , Nanomedicina/métodos , Péptidos/uso terapéutico , Agregación Patológica de Proteínas/terapia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/aislamiento & purificación , Animales , Modelos Animales de Enfermedad , Ratones , Modelos Moleculares , Nanocompuestos/química , Nanocompuestos/ultraestructura , Péptidos/química , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología
11.
J Chem Inf Model ; 59(5): 1909-1918, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-30575391

RESUMEN

The specific properties of carbon nanoparticles (NPs) have attracted great attention in applications in biotechnology and biomedicine, e.g., in the field of amyloidosis. To date, it is still indefinable whether carbon NPs would promote or inhibit the fibril formation of amyloid proteins. Here, to uncover the effects of carbon nanoparticles (NPs) including graphene and carbon nanotubes on the aggregation of prion proteins, whose misfolding and aggregation will lead to prion diseases, a ThT fluorescence assay and a molecular dynamics (MD) simulation were performed. The ThT fluorescence assay reveals that both graphene and carbon nanotubes can inhibit the fibril formation of prion proteins, especially graphene. Further MD simulation of the PrP127-147 tetramer with or without carbon NPs suggests that the interactions between prion proteins and carbon NPs reduce the aggregation tendency of PrP127-147 by decreasing the interpeptide interactions and thus inhibiting ß-sheet formation. Meanwhile, aromatic residues greatly contribute to the inhibition effects of carbon NPs by a π-π stacking interaction. The obtained results can increase our understanding on the interaction between nanoparticles and amyloid-related proteins.


Asunto(s)
Carbono/farmacología , Nanopartículas , Nanotubos de Carbono , Proteínas Priónicas/metabolismo , Agregado de Proteínas , Carbono/química , Grafito/química , Grafito/farmacología , Humanos , Simulación de Dinámica Molecular , Nanomedicina , Nanopartículas/química , Nanotubos de Carbono/química , Enfermedades por Prión/metabolismo , Enfermedades por Prión/terapia , Proteínas Priónicas/química , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/terapia
12.
Acta Neuropathol Commun ; 6(1): 59, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-30001207

RESUMEN

Aggregation of tau protein and spreading of tau aggregates are pivotal pathological processes in a range of neurological disorders. Accumulating evidence suggests that immunotherapy targeting tau may be a viable therapeutic strategy. We have previously described the isolation of antibody CBTAU-22.1 from the memory B-cell repertoire of healthy human donors. CBTAU-22.1 was shown to specifically bind a disease-associated phosphorylated epitope in the C-terminus of tau (Ser422) and to be able to inhibit the spreading of pathological tau aggregates from P301S spinal cord lysates in vitro, albeit with limited potency. Using a combination of rational design and random mutagenesis we have derived a variant antibody with improved affinity while maintaining the specificity of the parental antibody. This affinity improved antibody showed greatly enhanced potency in a cell-based immunodepletion assay using paired helical filaments (PHFs) derived from human Alzheimer's disease (AD) brain tissue. Moreover, the affinity improved antibody limits the in vitro aggregation propensity of full length tau species specifically phosphorylated at position 422 produced by employing a native chemical ligation approach. Together, these results indicate that in addition to being able to inhibit the spreading of pathological tau aggregates, the matured antibody can potentially also interfere with the nucleation of tau which is believed to be the first step of the pathogenic process. Finally, the functionality in a P301L transgenic mice co-injection model highlights the therapeutic potential of human antibody dmCBTAU-22.1.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Anticuerpos/farmacología , Encéfalo/metabolismo , Serina/metabolismo , Proteínas tau/inmunología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Animales , Afinidad de Anticuerpos/efectos de los fármacos , Autopsia , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Epítopos/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Microscopía de Fuerza Atómica , Persona de Mediana Edad , Modelos Moleculares , Mutagénesis , Mutación/genética , Fosforilación/fisiología , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/terapia
13.
Neurobiol Dis ; 109(Pt B): 209-218, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28751258

RESUMEN

The deposition of misfolded ß-sheet enriched amyloid protein is a shared feature of many neurodegenerative diseases. Recent studies demonstrated the existence of conformationally diverse strains as a common property for multiple amyloidogenic proteins including α-Synuclein (α-Syn). α-Syn is misfolded and aggregated in a group of neurodegenerative diseases collectively known as α-Synucleinopathies, which include Parkinson's disease (PD), dementia with Lewy body, multiple system atrophy and also a subset of Alzheimer's disease patients with concomitant PD-like Lewy bodies and neurites. While sharing the same pathological protein, different α-Synucleinopathies demonstrate distinct clinical and pathological phenotypes, which could result from the existence of diverse pathological α-Syn strains in patients. In this review, we summarized the characteristics of different α-Synucleinopathies and α-Syn strains generated with recombinant α-Syn monomers. We also make predictions of α-Syn strains that could potentially exist in patients based on the knowledge from other amyloid proteins and the clinical and pathological features of different α-Synucleinopathies.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/terapia , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/terapia , Conformación Proteica , alfa-Sinucleína/química , alfa-Sinucleína/genética
15.
Neurobiol Dis ; 109(Pt B): 178-190, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28709995

RESUMEN

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


Asunto(s)
Amiloide/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/terapia , Amiloide/efectos de los fármacos , Animales , Fenómenos Biofísicos , Humanos , Pliegue de Proteína
16.
Pediatr Blood Cancer ; 65(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29286575

RESUMEN

MYH9-related disease is a rare cause of thrombocytopenia. We report an infant girl who presented with severe thrombocytopenia at birth and was initially diagnosed with and treated for neonatal alloimmune thrombocytopenia. However, persistent thrombocytopenia led to the suspicion of congenital thrombocytopenia and subsequent identification of a novel variant in MYH9 (E1421K). In silico analysis strongly predicts that this is a disruptive substitution. Immunofluorescent analysis of neutrophils demonstrates abnormal aggregates of MYH9 protein. This case also suggests that a very high immature platelet fraction (≥40%) may be useful for rapidly differentiating MYH9-related disease from other causes of neonatal thrombocytopenia.


Asunto(s)
Proteínas Motoras Moleculares , Mutación Missense , Cadenas Pesadas de Miosina , Agregación Patológica de Proteínas , Trombocitopenia Neonatal Aloinmune , Sustitución de Aminoácidos , Plaquetas/metabolismo , Plaquetas/patología , Femenino , Enfermedades Genéticas Congénitas/sangre , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Enfermedades Genéticas Congénitas/terapia , Humanos , Recién Nacido , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patología , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/terapia , Trombocitopenia Neonatal Aloinmune/sangre , Trombocitopenia Neonatal Aloinmune/genética , Trombocitopenia Neonatal Aloinmune/patología , Trombocitopenia Neonatal Aloinmune/terapia
17.
Prog Neuropsychopharmacol Biol Psychiatry ; 79(Pt B): 452-461, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28779908

RESUMEN

BACKGROUND: Tau is a microtubule-associated protein that becomes pathological when it undergoes hyperphosphorylation and aggregation as seen in Alzheimer's disease (AD). AD is mostly sporadic, with environmental, biological and/or genetic risks factors, interacting together to promote the disease. In the past decade, reports have suggested that obesity in midlife could be one of these risk factors. On the other hand, caloric restriction and physical exercise have been reported to reduce the incidence and outcome of obesity as well as AD. METHODS: We evaluated the impact of voluntary physical exercise and caloric restriction on tau pathology during 2months in hTau mice under high caloric diet in order to evaluate if these strategies could prevent AD-like pathology in obese conditions. RESULTS: We found no effects of obesity induced by Western diet on both Tau phosphorylation and aggregation compared to controls. However, exercise reduced tau phosphorylation while caloric restriction exacerbated its aggregation in the brains of obese hTau mice. We then examined the mechanisms underlying changes in tau phosphorylation and aggregation by exploring major tau kinases and phosphatases and key proteins involved in autophagy. However, there were no significant effects of voluntary exercise and caloric restriction on these proteins in hTau mice that could explain our results. CONCLUSION: In this study, we report differential effects of voluntary treadmill exercise and caloric restriction on tau pathogenesis in our obese mice, namely beneficial effect of exercise on tau phosphorylation and deleterious effect of caloric restriction on tau aggregation. Our results suggest that lifestyle strategies used to reduce metabolic disorders and AD must be selected and studied carefully to avoid exacerbation of pathologies.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Restricción Calórica/efectos adversos , Dieta Occidental/efectos adversos , Actividad Motora/fisiología , Obesidad/fisiopatología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/complicaciones , Obesidad/patología , Obesidad/terapia , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/fisiopatología , Agregación Patológica de Proteínas/terapia , Volición , Proteínas tau/deficiencia , Proteínas tau/genética
18.
Acta Neuropathol ; 134(6): 819-838, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28803412

RESUMEN

Alpha-synuclein is a protein implicated in Parkinson's disease and thought to be one of the main pathological drivers in the disease, although it remains unclear how this protein elicits its neurotoxic effects. Recent findings indicate that the assembly of toxic oligomeric species of alpha-synuclein may be one of the key processes for the pathology and spread of the disease. The absence of a sensitive in situ detection method has hindered the study of these oligomeric species and the role they play in the human brain until recently. In this review, we assess the evidence for the toxicity and prion-like activity of oligomeric forms of alpha-synuclein and discuss the advances in our understanding of the role of alpha-synuclein in Parkinson's disease that may be brought about by the specific and sensitive detection of distinct oligomeric species in post-mortem patient brain. Finally, we discuss current approaches being taken to therapeutically target alpha-synuclein oligomers and their implications.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , alfa-Sinucleína/metabolismo , Animales , Antiparkinsonianos/farmacología , Biomarcadores/metabolismo , Humanos , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/terapia , alfa-Sinucleína/toxicidad
19.
Nature ; 544(7650): 367-371, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28405022

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease that is characterized by motor neuron loss and that leads to paralysis and death 2-5 years after disease onset. Nearly all patients with ALS have aggregates of the RNA-binding protein TDP-43 in their brains and spinal cords, and rare mutations in the gene encoding TDP-43 can cause ALS. There are no effective TDP-43-directed therapies for ALS or related TDP-43 proteinopathies, such as frontotemporal dementia. Antisense oligonucleotides (ASOs) and RNA-interference approaches are emerging as attractive therapeutic strategies in neurological diseases. Indeed, treatment of a rat model of inherited ALS (caused by a mutation in Sod1) with ASOs against Sod1 has been shown to substantially slow disease progression. However, as SOD1 mutations account for only around 2-5% of ALS cases, additional therapeutic strategies are needed. Silencing TDP-43 itself is probably not appropriate, given its critical cellular functions. Here we present a promising alternative therapeutic strategy for ALS that involves targeting ataxin-2. A decrease in ataxin-2 suppresses TDP-43 toxicity in yeast and flies, and intermediate-length polyglutamine expansions in the ataxin-2 gene increase risk of ALS. We used two independent approaches to test whether decreasing ataxin-2 levels could mitigate disease in a mouse model of TDP-43 proteinopathy. First, we crossed ataxin-2 knockout mice with TDP-43 (also known as TARDBP) transgenic mice. The decrease in ataxin-2 reduced aggregation of TDP-43, markedly increased survival and improved motor function. Second, in a more therapeutically applicable approach, we administered ASOs targeting ataxin-2 to the central nervous system of TDP-43 transgenic mice. This single treatment markedly extended survival. Because TDP-43 aggregation is a component of nearly all cases of ALS, targeting ataxin-2 could represent a broadly effective therapeutic strategy.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Ataxina-2/deficiencia , Proteínas de Unión al ADN/metabolismo , Longevidad , Oligonucleótidos Antisentido/uso terapéutico , Agregación Patológica de Proteínas/terapia , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Ataxina-2/genética , Sistema Nervioso Central/metabolismo , Gránulos Citoplasmáticos/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Destreza Motora/fisiología , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/genética , Agregación Patológica de Proteínas/genética , Estrés Fisiológico , Análisis de Supervivencia
20.
EMBO Mol Med ; 9(5): 687-702, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28351931

RESUMEN

Cell-to-cell transmission of protein aggregates is an emerging theme in neurodegenerative disease. Here, we analyze the dipeptide repeat (DPR) proteins that form neuronal inclusions in patients with hexanucleotide repeat expansion C9orf72, the most common known cause of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Sense and antisense transcripts of the (G4C2)n repeat are translated by repeat-associated non-ATG (RAN) translation in all reading frames into five aggregating DPR proteins. We show that the hydrophobic DPR proteins poly-GA, poly-GP, and poly-PA are transmitted between cells using co-culture assays and cell extracts. Moreover, uptake or expression of poly-GA induces nuclear RNA foci in (G4C2)80-expressing cells and patient fibroblasts, suggesting an unexpected positive feedback loop. Exposure to recombinant poly-GA and cerebellar extracts of C9orf72 patients increases repeat RNA levels and seeds aggregation of all DPR proteins in receiver cells expressing (G4C2)80 Treatment with anti-GA antibodies inhibits intracellular poly-GA aggregation and blocks the seeding activity of C9orf72 brain extracts. Poly-GA-directed immunotherapy may thus reduce DPR aggregation and disease progression in C9orf72 ALS/FTD.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Anticuerpos/uso terapéutico , Proteína C9orf72/genética , Inmunoterapia , Agregación Patológica de Proteínas/terapia , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Células HEK293 , Humanos , Inmunoterapia/métodos , Neuronas/metabolismo , Neuronas/patología , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...