Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Acta Neuropathol Commun ; 5(1): 81, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29115989

RESUMO

It is now widely accepted in the field that the normally secreted chaperone clusterin is redirected to the cytosol during endoplasmic reticulum (ER) stress, although the physiological function(s) of this physical relocation remain unknown. We have examined in this study whether or not increased expression of clusterin is able to protect neuronal cells against intracellular protein aggregation and cytotoxicity, characteristics that are strongly implicated in a range of neurodegenerative diseases. We used the amyotrophic lateral sclerosis-associated protein TDP-43 as a primary model to investigate the effects of clusterin on protein aggregation and neurotoxicity in complementary in vitro, neuronal cell and Drosophila systems. We have shown that clusterin directly interacts with TDP-43 in vitro and potently inhibits its aggregation, and observed that in ER stressed neuronal cells, clusterin co-localized with TDP-43 and specifically reduced the numbers of cytoplasmic inclusions. We further showed that the expression of TDP-43 in transgenic Drosophila neurons induced ER stress and that co-expression of clusterin resulted in a dramatic clearance of mislocalized TDP-43 from motor neuron axons, partially rescued locomotor activity and significantly extended lifespan. We also showed that in Drosophila photoreceptor cells, clusterin co-expression gave ER stress-dependent protection against proteotoxicity arising from both Huntingtin-Q128 and mutant (R406W) human tau. We therefore conclude that increased expression of clusterin can provide an important defense against intracellular proteotoxicity under conditions that mimic specific features of neurodegenerative disease.


Assuntos
Clusterina/metabolismo , Clusterina/farmacologia , Proteínas de Ligação a DNA/metabolismo , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Clusterina/genética , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Estresse do Retículo Endoplasmático/genética , Olho/metabolismo , Olho/ultraestrutura , Hemolinfa/citologia , Humanos , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Larva , Atividade Motora/genética , Atividade Motora/fisiologia , Neurônios Motores/ultraestrutura , Neuroblastoma/patologia , Síndromes Neurotóxicas/genética , Síndromes Neurotóxicas/patologia , Agregados Proteicos/efeitos dos fármacos , Agregados Proteicos/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
2.
Chem Biol ; 21(6): 732-42, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24856820

RESUMO

Insight into how amyloid ß (Aß) aggregation occurs in vivo is vital for understanding the molecular pathways that underlie Alzheimer's disease and requires new techniques that provide detailed kinetic and mechanistic information. Using noninvasive fluorescence lifetime recordings, we imaged the formation of Aß(1-40) and Aß(1-42) aggregates in live cells. For both peptides, the cellular uptake via endocytosis is rapid and spontaneous. They are then retained in lysosomes, where their accumulation leads to aggregation. The kinetics of Aß(1-42) aggregation are considerably faster than those of Aß(1-40) and, unlike those of the latter peptide, show no detectable lag phase. We used superresolution fluorescence imaging to examine the resulting aggregates and could observe compact amyloid structures, likely because of spatial confinement within cellular compartments. Taken together, these findings provide clues as to how Aß aggregation may occur within neurons.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos , Agregação Patológica de Proteínas , Peptídeos beta-Amiloides/biossíntese , Sobrevivência Celular , Humanos , Cinética , Fragmentos de Peptídeos/biossíntese , Células Tumorais Cultivadas
3.
Acta Neuropathol Commun ; 2: 43, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24725347

RESUMO

INTRODUCTION: The self-assembly of Aß peptides into a range of conformationally heterogeneous amyloid states represents a fundamental event in Alzheimer's disease. Within these structures oligomeric intermediates are considered to be particularly pathogenic. To test this hypothesis we have used a conformational targeting approach where particular conformational states, such as oligomers or fibrils, are recognized in vivo by state-specific antibody fragments. RESULTS: We show that oligomer targeting with the KW1 antibody fragment, but not fibril targeting with the B10 antibody fragment, affects toxicity in Aß-expressing Drosophila melanogaster. The effect of KW1 is observed to occur selectively with flies expressing Aß(1-40) and not with those expressing Aß(1-42) or the arctic variant of Aß(1-42) This finding is consistent with the binding preference of KW1 for Aß(1-40) oligomers that has been established in vitro. Strikingly, and in contrast to the previously demonstrated in vitro ability of this antibody fragment to block oligomeric toxicity in long-term potentiation measurements, KW1 promotes toxicity in the flies rather than preventing it. This result shows the crucial importance of the environment in determining the influence of antibody binding on the nature and consequences of the protein misfolding and aggregation. CONCLUSIONS: While our data support to the pathological relevance of oligomers, they highlight the issues to be addressed when developing inhibitory strategies that aim to neutralize these states by means of antagonistic binding agents.


Assuntos
Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Anticorpos/uso terapêutico , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Animais , Animais Geneticamente Modificados , Anticorpos/química , Anticorpos/genética , Anticorpos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster , Olho/metabolismo , Olho/ultraestrutura , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/patologia , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/fisiopatologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Agregação Patológica de Proteínas , Ligação Proteica/efeitos dos fármacos , Conformação Proteica
4.
Proc Natl Acad Sci U S A ; 110(24): 9758-63, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23703910

RESUMO

The generation of toxic oligomers during the aggregation of the amyloid-ß (Aß) peptide Aß42 into amyloid fibrils and plaques has emerged as a central feature of the onset and progression of Alzheimer's disease, but the molecular pathways that control pathological aggregation have proved challenging to identify. Here, we use a combination of kinetic studies, selective radiolabeling experiments, and cell viability assays to detect directly the rates of formation of both fibrils and oligomers and the resulting cytotoxic effects. Our results show that once a small but critical concentration of amyloid fibrils has accumulated, the toxic oligomeric species are predominantly formed from monomeric peptide molecules through a fibril-catalyzed secondary nucleation reaction, rather than through a classical mechanism of homogeneous primary nucleation. This catalytic mechanism couples together the growth of insoluble amyloid fibrils and the generation of diffusible oligomeric aggregates that are implicated as neurotoxic agents in Alzheimer's disease. These results reveal that the aggregation of Aß42 is promoted by a positive feedback loop that originates from the interactions between the monomeric and fibrillar forms of this peptide. Our findings bring together the main molecular species implicated in the Aß aggregation cascade and suggest that perturbation of the secondary nucleation pathway identified in this study could be an effective strategy to control the proliferation of neurotoxic Aß42 oligomers.


Assuntos
Peptídeos beta-Amiloides/química , Amiloide/química , Fragmentos de Peptídeos/química , Placa Amiloide/química , Multimerização Proteica , Algoritmos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Marcação por Isótopo , Cinética , Modelos Químicos , Modelos Moleculares , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Placa Amiloide/metabolismo , Polimerização , Conformação Proteica
5.
ACS Nano ; 6(6): 4740-7, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22631869

RESUMO

The aggregation of misfolded proteins is a common feature underlying a wide range of age-related degenerative disorders, including Alzheimer's and Parkinson's diseases. A key aspect of understanding the molecular origins of these conditions is to define the manner in which specific types of protein aggregates influence disease pathogenesis through their interactions with cells. We demonstrate how selenium-enhanced electron microscopy (SE-EM), combined with tomographic reconstruction methods, can be used to image, here at a resolution of 5-10 nm, the interaction with human macrophage cells of amyloid aggregates formed from Aß(25-36), a fragment of the Aß peptide whose self-assembly is associated with Alzheimer's disease. We find that prefibrillar aggregates and mature fibrils are distributed into distinct subcellular compartments and undergo varying degrees of morphological change over time, observations that shed new light on the origins of their differential toxicity and the mechanisms of their clearance. In addition, the results show that SE-EM provides a powerful and potentially widely applicable means to define the nature and location of protein assemblies in situ and to provide detailed and specific information about their partitioning and processing.


Assuntos
Peptídeos beta-Amiloides/química , Aumento da Imagem/métodos , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Microscopia Eletrônica/métodos , Complexos Multiproteicos/ultraestrutura , Selênio , Células Cultivadas , Meios de Contraste , Humanos , Ligação Proteica , Dobramento de Proteína
6.
FASEB J ; 26(1): 192-202, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21965601

RESUMO

We have created a Drosophila model of lysozyme amyloidosis to investigate the in vivo behavior of disease-associated variants. To achieve this objective, wild-type (WT) protein and the amyloidogenic variants F57I and D67H were expressed in Drosophila melanogaster using the UAS-gal4 system and both the ubiquitous and retinal expression drivers Act5C-gal4 and gmr-gal4. The nontransgenic w(1118) Drosophila line was used as a control throughout. We utilized ELISA experiments to probe lysozyme protein levels, scanning electron microscopy for eye phenotype classification, and immunohistochemistry to detect the unfolded protein response (UPR) activation. We observed that expressing the destabilized F57I and D67H lysozymes triggers UPR activation, resulting in degradation of these variants, whereas the WT lysozyme is secreted into the fly hemolymph. Indeed, the level of WT was up to 17 times more abundant than the variant proteins. In addition, the F57I variant gave rise to a significant disruption of the eye development, and this correlated to pronounced UPR activation. These results support the concept that the onset of familial amyloid disease is linked to an inability of the UPR to degrade completely the amyloidogenic lysozymes prior to secretion, resulting in secretion of these destabilized variants, thereby leading to deposition and associated organ damage.


Assuntos
Amiloidose/enzimologia , Anormalidades do Olho/enzimologia , Muramidase/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Amiloidose/patologia , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster , Estresse do Retículo Endoplasmático/fisiologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Feminino , Proteínas de Fluorescência Verde/genética , Hemolinfa/enzimologia , Humanos , Masculino , Metamorfose Biológica/fisiologia , Microscopia Eletrônica de Varredura , Muramidase/genética , Células Fotorreceptoras de Invertebrados/enzimologia , Células Fotorreceptoras de Invertebrados/patologia , Células Fotorreceptoras de Invertebrados/ultraestrutura , Solubilidade
7.
Proc Natl Acad Sci U S A ; 107(35): 15595-600, 2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20713699

RESUMO

Soluble oligomeric aggregates of the amyloid-beta peptide (Abeta) have been implicated in the pathogenesis of Alzheimer's disease (AD). Although the conformation adopted by Abeta within these aggregates is not known, a beta-hairpin conformation is known to be accessible to monomeric Abeta. Here we show that this beta-hairpin is a building block of toxic Abeta oligomers by engineering a double-cysteine mutant (called Abetacc) in which the beta-hairpin is stabilized by an intramolecular disulfide bond. Abeta(40)cc and Abeta(42)cc both spontaneously form stable oligomeric species with distinct molecular weights and secondary-structure content, but both are unable to convert into amyloid fibrils. Biochemical and biophysical experiments and assays with conformation-specific antibodies used to detect Abeta aggregates in vivo indicate that the wild-type oligomer structure is preserved and stabilized in Abetacc oligomers. Stable oligomers are expected to become highly toxic and, accordingly, we find that beta-sheet-containing Abeta(42)cc oligomers or protofibrillar species formed by these oligomers are 50 times more potent inducers of neuronal apoptosis than amyloid fibrils or samples of monomeric wild-type Abeta(42), in which toxic aggregates are only transiently formed. The possibility of obtaining completely stable and physiologically relevant neurotoxic Abeta oligomer preparations will facilitate studies of their structure and role in the pathogenesis of AD. For example, here we show how kinetic partitioning into different aggregation pathways can explain why Abeta(42) is more toxic than the shorter Abeta(40), and why certain inherited mutations are linked to protofibril formation and early-onset AD.


Assuntos
Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Engenharia de Proteínas/métodos , Doença de Alzheimer/metabolismo , Amiloide/química , Amiloide/ultraestrutura , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Humanos , Cinética , Microscopia de Força Atômica , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Peso Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Espectroscopia de Infravermelho com Transformada de Fourier
8.
Front Biosci (Landmark Ed) ; 15(1): 373-96, 2010 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-20036826

RESUMO

Protein misfolding and aggregation are implicated in a wide range of increasingly prevalent human diseases ranging from dementia to diabetes. In this review we discuss the current experimental strategies that are being employed in the investigation of the pathogenesis of three important protein misfolding disorders. The first, Alzheimer's disease (AD), is the most prevalent neurodegenerative disease and is thought to be initiated by the aggregation of a natively unstructured peptide called amyloid beta (Abeta). We discuss methods for the characterization of the aggregation properties of Abeta in vitro and how the results of such experiments can be correlated with data from animal models of disease. We then consider another form of amyloidosis, where a systemic distribution of amyloid deposit is caused by aggregation and deposition of mutational variants of lysozyme. We describe how experiments in vitro, and more recently in vivo, have provided insights into the origins of this disease. Finally we outline the varied paradigms that have been employed in the study of the serpinopathies, and in particular, a dementia caused by neuroserpin polymerization.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Amiloidose/metabolismo , Muramidase/química , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Amiloidose/patologia , Animais , Dicroísmo Circular , Humanos , Microscopia Eletrônica de Transmissão , Muramidase/metabolismo , Conformação Proteica , Dobramento de Proteína
9.
Curr Opin Chem Biol ; 12(1): 25-31, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18295611

RESUMO

Protein misfolding is the underlying cause of many highly debilitating disorders ranging from Alzheimer's Disease to Cystic Fibrosis. Great strides have been made recently in understanding what causes proteins to misfold, primarily through the use of biophysical and computational techniques that enable systematic and quantitative analysis of the effects of a range of different perturbations in proteins. Correlation of the results of such analyses with observations made in animal models of disease has however been limited by their seemingly irreconcilable differences in methodology and scope. Several recent studies have however begun to overcome this limitation by combining the two approaches. This strategy has made it possible to investigate many of the consequences of protein misfolding in vivo, ranging from disease pathogenesis to epigenetic regulation, in the context of the fundamental physico-chemical principles derived from extensive and highly detailed studies undertaken in vitro.


Assuntos
Modelos Animais de Doenças , Doença , Dobramento de Proteína , Proteínas/química , Proteínas/metabolismo , Animais , Humanos , Proteínas/genética
10.
PLoS Comput Biol ; 3(9): 1727-38, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17941703

RESUMO

Increasing evidence indicates that oligomeric protein assemblies may represent the molecular species responsible for cytotoxicity in a range of neurological disorders including Alzheimer and Parkinson diseases. We use all-atom computer simulations to reveal that the process of oligomerization can be divided into two steps. The first is characterised by a hydrophobic coalescence resulting in the formation of molten oligomers in which hydrophobic residues are sequestered away from the solvent. In the second step, the oligomers undergo a process of reorganisation driven by interchain hydrogen bonding interactions that induce the formation of beta sheet rich assemblies in which hydrophobic groups can become exposed. Our results show that the process of aggregation into either ordered or amorphous species is largely determined by a competition between the hydrophobicity of the amino acid sequence and the tendency of polypeptide chains to form arrays of hydrogen bonds. We discuss how the increase in solvent-exposed hydrophobic surface resulting from such a competition offers an explanation for recent observations concerning the cytotoxicity of oligomeric species formed prior to mature amyloid fibrils.


Assuntos
Amiloide/química , Amiloide/ultraestrutura , Modelos Químicos , Modelos Moleculares , Análise de Sequência de Proteína/métodos , Sequência de Aminoácidos , Sítios de Ligação , Simulação por Computador , Dimerização , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica
11.
J Neurosci ; 26(31): 8160-7, 2006 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-16885229

RESUMO

More than 40 human diseases are associated with fibrillar deposits of specific peptides or proteins in tissue. Amyloid fibrils, or their precursors, can be highly toxic to cells, suggesting their key role in disease pathogenesis. Proteins not associated with any disease are able to form oligomers and amyloid assemblies in vitro displaying structures and cytotoxicity comparable with those of aggregates of disease-related polypeptides. In isolated cells, such toxicity has been shown to result from increased membrane permeability with disruption of ion homeostasis and oxidative stress. Here we microinjected into the nucleus basalis magnocellularis of rat brains aggregates of an Src homology 3 domain and the N-terminal domain of the prokaryotic HypF, neither of which is associated with amyloid disease. Prefibrillar aggregates of both proteins, but not their mature fibrils or soluble monomers, impaired cholinergic neuron viability in a dose-dependent manner similar to that seen in cell cultures. Contrary to the situation with cultured cells, however, under our experimental conditions, cell stress in tissue is not followed by a comparable level of cell death, a result that is very likely to reflect the presence of protective mechanisms reducing aggregate toxicity. These findings support the hypothesis that neurodegenerative disorders result primarily from a generic cell dysfunction caused by early misfolded species in the aggregation process.


Assuntos
Amiloide/toxicidade , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Proteínas/toxicidade , Animais , Carboxil e Carbamoil Transferases , Células Cultivadas , Relação Dose-Resposta a Droga , Proteínas de Escherichia coli , Masculino , Doenças Neurodegenerativas/induzido quimicamente , Ratos , Ratos Wistar , Toxinas Biológicas/toxicidade
12.
Neurosci Lett ; 381(3): 294-8, 2005 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-15896487

RESUMO

Parkinson's disease (PD) is a heterogeneous disease that can be difficult to diagnose, and for which we have no simple effective biomarker. In this study we have investigated whether peripheral alpha-synuclein might represent a useful biomarker given that it has a central role in the pathogenesis of PD. We found that full length and truncated alpha-synuclein is present in platelets, but the amount is very variable and does not correlate with disease presence or severity. Furthermore, we show that alpha-synuclein can be detected by immunoblotting in some, but not all, human skin biopsies, but again its level does not correlate with disease presence or severity. We conclude that skin or platelet alpha-synuclein would not be an appropriate diagnostic biomarker for PD.


Assuntos
Biomarcadores/análise , Plaquetas/química , Proteínas do Tecido Nervoso/metabolismo , Doença de Parkinson/diagnóstico , Pele/química , Biópsia , Plaquetas/metabolismo , Western Blotting , Humanos , Projetos Piloto , Pele/metabolismo , Sinucleínas , alfa-Sinucleína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA