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.
Eur J Neurosci ; 53(2): 637-648, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33169893

RESUMO

One of the key knowledge gaps in the field of Alzheimer's disease research is the lack of understanding of how amyloid beta and tau cooperate to cause neurodegeneration. We recently generated a mouse model (APP/PS1 + Tau) that develops amyloid plaque pathology and expresses human tau in the absence of endogenous murine tau. These mice exhibit an age-related behavioural hyperactivity phenotype and transcriptional deficits which are ameliorated by tau transgene suppression. We hypothesized that these mice would also display memory and hippocampal synaptic plasticity deficits as has been reported for many plaque bearing mouse models which express endogenous mouse tau. We observed that our APP/PS1 + Tau model does not exhibit novel object memory or robust long-term potentiation deficits with age, whereas the parent APP/PS1 line with mouse tau did develop the expected deficits. These data are important as they highlight potential functional differences between mouse and human tau and the need to use multiple models to fully understand Alzheimer's disease pathogenesis and develop effective therapeutic strategies.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Humanos , Potenciação de Longa Duração , Camundongos , Camundongos Transgênicos , Placa Amiloide , Presenilina-1 , Proteínas tau/genética
2.
Cell Rep ; 29(11): 3592-3604.e5, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31825838

RESUMO

A key knowledge gap blocking development of effective therapeutics for Alzheimer's disease (AD) is the lack of understanding of how amyloid beta (Aß) peptide and pathological forms of the tau protein cooperate in causing disease phenotypes. Within a mouse tau-deficient background, we probed the molecular, cellular, and behavioral disruption triggered by the influence of wild-type human tau on human Aß-induced pathology. We find that Aß and tau work cooperatively to cause a hyperactivity behavioral phenotype and to cause downregulation of transcription of genes involved in synaptic function. In both our mouse model and human postmortem tissue, we observe accumulation of pathological tau in synapses, supporting the potential importance of synaptic tau. Importantly, tau reduction in the mice initiated after behavioral deficits emerge corrects behavioral deficits, reduces synaptic tau levels, and substantially reverses transcriptional perturbations, suggesting that lowering synaptic tau levels may be beneficial in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Feminino , Humanos , Masculino , Camundongos , Microglia/metabolismo , Comportamento Espacial , Sinapses/metabolismo , Transcriptoma
3.
Acta Neuropathol Commun ; 7(1): 214, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31862015

RESUMO

Degeneration of synapses in Alzheimer's disease (AD) strongly correlates with cognitive decline, and synaptic pathology contributes to disease pathophysiology. We recently observed that the strongest genetic risk factor for sporadic AD, apolipoprotein E epsilon 4 (APOE4), is associated with exacerbated synapse loss and synaptic accumulation of oligomeric amyloid beta in human AD brain. To begin to understand the molecular cascades involved in synapse loss in AD and how this is mediated by APOE, and to generate a resource of knowledge of changes in the synaptic proteome in AD, we conducted a proteomic screen and systematic in silico analysis of synaptoneurosome preparations from temporal and occipital cortices of human AD and control subjects with known APOE gene status. We examined brain tissue from 33 subjects (7-10 per group). We pooled tissue from all subjects in each group for unbiased proteomic analyses followed by validation with individual case samples. Our analysis identified over 5500 proteins in human synaptoneurosomes and highlighted disease, brain region, and APOE-associated changes in multiple molecular pathways including a decreased abundance in AD of proteins important for synaptic and mitochondrial function and an increased abundance of proteins involved in neuroimmune interactions and intracellular signaling.


Assuntos
Doença de Alzheimer/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo , Proteoma , Sinapses/metabolismo , Adulto , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Apolipoproteína E4/metabolismo , Encéfalo/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Neurônios/patologia , Proteômica , Sinapses/patologia
4.
Brain ; 140(12): 3204-3214, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29177427

RESUMO

Dementia with Lewy bodies is characterized by the accumulation of Lewy bodies and Lewy neurites in the CNS, both of which are composed mainly of aggregated α-synuclein phosphorylated at Ser129. Although phosphorylated α-synuclein is believed to exert toxic effects at the synapse in dementia with Lewy bodies and other α-synucleinopathies, direct evidence for the precise synaptic localization has been difficult to achieve due to the lack of adequate optical microscopic resolution to study human synapses. In the present study we applied array tomography, a microscopy technique that combines ultrathin sectioning of tissue with immunofluorescence allowing precise identification of small structures, to quantitatively investigate the synaptic phosphorylated α-synuclein pathology in dementia with Lewy bodies. We performed array tomography on human brain samples from five patients with dementia with Lewy bodies, five patients with Alzheimer's disease and five healthy control subjects to analyse the presence of phosphorylated α-synuclein immunoreactivity at the synapse and their relationship with synapse size. Main analyses were performed in blocks from cingulate cortex and confirmed in blocks from the striatum of cases with dementia with Lewy bodies. A total of 1 318 700 single pre- or postsynaptic terminals were analysed. We found that phosphorylated α-synuclein is present exclusively in dementia with Lewy bodies cases, where it can be identified in the form of Lewy bodies, Lewy neurites and small aggregates (<0.16 µm3). Between 19% and 25% of phosphorylated α-synuclein deposits were found in presynaptic terminals mainly in the form of small aggregates. Synaptic terminals that co-localized with small aggregates of phosphorylated α-synuclein were significantly larger than those that did not. Finally, a gradient of phosphorylated α-synuclein aggregation in synapses (pre > pre + post > postsynaptic) was observed. These results indicate that phosphorylated α-synuclein is found at the presynaptic terminals of dementia with Lewy bodies cases mainly in the form of small phosphorylated α-synuclein aggregates that are associated with changes in synaptic morphology. Overall, our data support the notion that pathological phosphorylated α-synuclein may disrupt the structure and function of the synapse in dementia with Lewy bodies.


Assuntos
Giro do Cíngulo/metabolismo , Doença por Corpos de Lewy/metabolismo , Neostriado/metabolismo , Fosfoproteínas/metabolismo , Sinapses/metabolismo , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Estudos de Casos e Controles , Feminino , Imunofluorescência , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade
5.
Nat Commun ; 8: 15295, 2017 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-28492240

RESUMO

Tau is implicated in more than 20 neurodegenerative diseases, including Alzheimer's disease. Under pathological conditions, Tau dissociates from axonal microtubules and missorts to pre- and postsynaptic terminals. Patients suffer from early synaptic dysfunction prior to Tau aggregate formation, but the underlying mechanism is unclear. Here we show that pathogenic Tau binds to synaptic vesicles via its N-terminal domain and interferes with presynaptic functions, including synaptic vesicle mobility and release rate, lowering neurotransmission in fly and rat neurons. Pathological Tau mutants lacking the vesicle binding domain still localize to the presynaptic compartment but do not impair synaptic function in fly neurons. Moreover, an exogenously applied membrane-permeable peptide that competes for Tau-vesicle binding suppresses Tau-induced synaptic toxicity in rat neurons. Our work uncovers a presynaptic role of Tau that may be part of the early pathology in various Tauopathies and could be exploited therapeutically.


Assuntos
Terminações Pré-Sinápticas/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas tau/metabolismo , Actinas/metabolismo , Animais , Drosophila melanogaster/metabolismo , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Humanos , Mutação/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Domínios Proteicos , Transporte Proteico , Ratos , Transmissão Sináptica , Proteínas tau/química
6.
Eur J Neurosci ; 44(12): 3056-3066, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27748574

RESUMO

Alzheimer's disease is characterized by the presence of aggregates of amyloid beta (Aß) in senile plaques and tau in neurofibrillary tangles, as well as marked neuron and synapse loss. Of these pathological changes, synapse loss correlates most strongly with cognitive decline. Synapse loss occurs prominently around plaques due to accumulations of oligomeric Aß. Recent evidence suggests that tau may also play a role in synapse loss but the interactions of Aß and tau in synapse loss remain to be determined. In this study, we generated a novel transgenic mouse line, the APP/PS1/rTg21221 line, by crossing APP/PS1 mice, which develop Aß-plaques and synapse loss, with rTg21221 mice, which overexpress wild-type human tau. When compared to the APP/PS1 mice without human tau, the cross-sectional area of ThioS+ dense core plaques was increased by ~50%. Along with increased plaque size, we observed an increase in plaque-associated dystrophic neurites containing misfolded tau, but there was no exacerbation of neurite curvature or local neuron loss around plaques. Array tomography analysis similarly revealed no worsening of synapse loss around plaques, and no change in the accumulation of Aß at synapses. Together, these results indicate that adding human wild-type tau exacerbates plaque pathology and neurite deformation but does not exacerbate plaque-associated synapse loss.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Modelos Animais de Doenças , Placa Amiloide/metabolismo , Sinapses/metabolismo , Sinapses/patologia , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Astrócitos/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fosforilação , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas tau/genética
7.
J Alzheimers Dis ; 53(3): 787-800, 2016 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-27258414

RESUMO

Alzheimer's disease (AD) is characterized by memory loss, insidious cognitive decline, profound neurodegeneration, and the extracellular accumulation of amyloid-ß (Aß) peptide in senile plaques and intracellular accumulation of tau in neurofibrillary tangles. Loss and dysfunction of synapses are believed to underlie the devastating cognitive decline in AD. A large amount of evidence suggests that oligomeric forms of Aß associated with senile plaques are toxic to synapses, but the precise sub-synaptic localization of Aß and which forms are synaptotoxic remain unknown. Here, we characterize the sub-synaptic localization of Aß oligomers using three high-resolution imaging techniques, stochastic optical reconstruction microscopy, immunogold electron microscopy, and Förster resonance energy transfer in a plaque-bearing mouse model of AD. With all three techniques, we observe oligomeric Aß inside synaptic terminals. Further, we tested a panel of Aß antibodies using the relatively high-throughput array tomography technique to determine which forms are present in synapses. Our results show that different oligomeric Aß species are present in synapses and highlight the potential of array tomography for rapid testing of aggregation state specific Aß antibodies in brain tissue.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Emaranhados Neurofibrilares/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/ultraestrutura , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large/metabolismo , Transferência Ressonante de Energia de Fluorescência , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Microscopia , Microscopia Imunoeletrônica , Emaranhados Neurofibrilares/ultraestrutura , Neurônios/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Placa Amiloide/ultraestrutura , Presenilina-1/genética , Presenilina-1/metabolismo , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Sinapses/ultraestrutura
8.
Alzheimers Dement ; 12(9): 996-1013, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27130892

RESUMO

Amyloid-beta (Aß) peptide oligomerization plays a central role in the pathogenesis of Alzheimer's disease (AD), and Aß oligomers are collectively considered an appealing therapeutic target for the treatment of AD. However, the molecular mechanisms leading to the pathologic accumulation of oligomers are unclear, and the exact structural composition of oligomers is being debated. Using targeted and quantitative mass spectrometry, we reveal site-specific Aß autocleavage during the early phase of aggregation, producing a typical Aß fragment signature and that truncated Aß peptides can form stable oligomeric complexes with full-length Aß peptide. We show that the use of novel anti-Aß antibodies raised against these truncated Aß isoforms allows for monitoring and targeting the accumulation of truncated Aß fragments. Antibody-enabled screening of transgenic models of AD as well as human postmortem brain tissue and cerebrospinal fluid revealed that aggregation-associated Aß cleavage is a highly relevant clinical feature of AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Animais , Autoanticorpos , Encéfalo/metabolismo , Cromatografia em Gel , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Humanos , Immunoblotting , Espectrometria de Massas , Camundongos Transgênicos , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Estrutura Secundária de Proteína
9.
Acta Neuropathol Commun ; 3(1): 83, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26651483

RESUMO

The original version of this article [1] unfortunately contained several mistakes. The presentation of Table 2 and 3 was incorrect, in the HTML and PDF versions of this article. The corrected Tables 2 and 3 are given below.

10.
Acta Neuropathol Commun ; 3: 53, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26335101

RESUMO

INTRODUCTION: Non-pathological, age-related cognitive decline varies markedly between individuals andplaces significant financial and emotional strain on people, their families and society as a whole.Understanding the differential age-related decline in brain function is critical not only for the development oftherapeutics to prolong cognitive health into old age, but also to gain insight into pathological ageing suchas Alzheimer's disease. The Lothian Birth Cohort of 1936 (LBC1936) comprises a rare group of people forwhom there are childhood cognitive test scores and longitudinal cognitive data during older age, detailedstructural brain MRI, genome-wide genotyping, and a multitude of other biological, psycho-social, andepidemiological data. Synaptic integrity is a strong indicator of cognitive health in the human brain;however, until recently, it was prohibitively difficult to perform detailed analyses of synaptic and axonalstructure in human tissue sections. We have adapted a novel method of tissue preparation at autopsy toallow the study of human synapses from the LBC1936 cohort in unprecedented morphological andmolecular detail, using the high-resolution imaging techniques of array tomography and electronmicroscopy. This allows us to analyze the brain at sub-micron resolution to assess density, proteincomposition and health of synapses. Here we present data from the first donated LBC1936 brain andcompare our findings to Alzheimer's diseased tissue to highlight the differences between healthy andpathological brain ageing. RESULTS: Our data indicates that compared to an Alzheimer's disease patient, the cognitively normalLBC1936 participant had a remarkable degree of preservation of synaptic structures. However,morphological and molecular markers of degeneration in areas of the brain associated with cognition(prefrontal cortex, anterior cingulate cortex, and superior temporal gyrus) were observed. CONCLUSIONS: Our novel post-mortem protocol facilitates high-resolution neuropathological analysis of the well-characterized LBC1936 cohort, extending phenotyping beyond cognition and in vivo imaging to nowinclude neuropathological changes, at the level of single synapses. This approach offers an unprecedentedopportunity to study synaptic and axonal integrity during ageing and how it contributes to differences in agerelatedcognitive change.


Assuntos
Envelhecimento/patologia , Encéfalo/patologia , Cognição , Sinapses/patologia , Idoso , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Encéfalo/metabolismo , Estudos de Coortes , Feminino , Histonas/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Proteínas de Membrana/metabolismo , Microglia/patologia , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Mudanças Depois da Morte , Sinapses/metabolismo , Sinapses/ultraestrutura
11.
Proteomics ; 13(22): 3251-5, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24123801

RESUMO

Quantitative proteomics is entering its "third generation," where intricate experimental designs aim to increase the spatial and temporal resolution of protein changes. This paper re-analyses multiple internally consistent proteomic datasets generated from whole cell homogenates and fractionated brain tissue samples providing a unique opportunity to explore the different factors influencing experimental outcomes. The results clearly indicate that improvements in data handling are required to compensate for the increased mean CV associated with complex study design and intricate upstream tissue processing. Furthermore, applying arbitrary inclusion thresholds such as fold change in protein abundance between groups can lead to unnecessary exclusion of important and biologically relevant data.


Assuntos
Biomarcadores/análise , Bases de Dados de Proteínas , Proteômica/métodos , Animais , Biomarcadores/química , Química Encefálica , Linhagem Celular , Cromatografia Líquida/métodos , Espaço Intracelular/química , Camundongos , Proteínas/análise , Proteínas/química , Espectrometria de Massas em Tandem/métodos
12.
J Cereb Blood Flow Metab ; 33(5): 673-83, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23321784

RESUMO

Impaired energy metabolism in neurons is integral to a range of neurodegenerative diseases, from Alzheimer's disease to stroke. To investigate the complex molecular changes underpinning cellular adaptation to metabolic stress, we have defined the proteomic response of the SH-SY5Y human neuroblastoma cell line after exposure to a metabolic challenge of oxygen glucose deprivation (OGD) in vitro. A total of 958 proteins across multiple subcellular compartments were detected and quantified by label-free liquid chromatography mass spectrometry. The levels of 130 proteins were significantly increased (P<0.01) after OGD and the levels of 63 proteins were significantly decreased (P<0.01) while expression of the majority of proteins (765) was not altered. Network analysis identified novel protein-protein interactomes involved with mitochondrial energy production, protein folding, and protein degradation, indicative of coherent and integrated proteomic responses to the metabolic challenge. Approximately one third (61) of the differentially expressed proteins was associated with the endoplasmic reticulum and mitochondria. Electron microscopic analysis of these subcellular structures showed morphologic changes consistent with the identified proteomic alterations. Our investigation of the global cellular response to a metabolic challenge clearly shows the considerable adaptive capacity of the proteome to a slowly evolving metabolic challenge.


Assuntos
Doença de Alzheimer/metabolismo , Estresse do Retículo Endoplasmático , Mitocôndrias/metabolismo , Neurônios/metabolismo , Proteoma/metabolismo , Ribossomos/metabolismo , Estresse Fisiológico , Doença de Alzheimer/patologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Glucose/metabolismo , Humanos , Mitocôndrias/patologia , Proteínas Mitocondriais/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Proteínas Ribossômicas/metabolismo , Ribossomos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...