Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 294(17): 6719-6732, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30814252

RESUMO

Amyloid-ß (Aß) pathology in Alzheimer's disease (AD) is characterized by the formation of polymorphic deposits comprising diffuse and cored plaques. Because diffuse plaques are predominantly observed in cognitively unaffected, amyloid-positive (CU-AP) individuals, pathogenic conversion into cored plaques appears to be critical to AD pathogenesis. Herein, we identified the distinct Aß species associated with amyloid polymorphism in brain tissue from individuals with sporadic AD (s-AD) and CU-AP. To this end, we interrogated Aß polymorphism with amyloid conformation-sensitive dyes and a novel in situ MS paradigm for chemical characterization of hyperspectrally delineated plaque morphotypes. We found that maturation of diffuse into cored plaques correlated with increased Aß1-40 deposition. Using spatial in situ delineation with imaging MS (IMS), we show that Aß1-40 aggregates at the core structure of mature plaques, whereas Aß1-42 localizes to diffuse amyloid aggregates. Moreover, we observed that diffuse plaques have increased pyroglutamated Aßx-42 levels in s-AD but not CU-AP, suggesting an AD pathology-related, hydrophobic functionalization of diffuse plaques facilitating Aß1-40 deposition. Experiments in tgAPPSwe mice verified that, similar to what has been observed in human brain pathology, diffuse deposits display higher levels of Aß1-42 and that Aß plaque maturation over time is associated with increases in Aß1-40. Finally, we found that Aß1-40 deposition is characteristic for cerebral amyloid angiopathy deposition and maturation in both humans and mice. These results indicate that N-terminal Aßx-42 pyroglutamation and Aß1-40 deposition are critical events in priming and maturation of pathogenic Aß from diffuse into cored plaques, underlying neurotoxic plaque development in AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Ácido Pirrolidonocarboxílico/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Animais , Progressão da Doença , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Conformação Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
2.
ACS Chem Neurosci ; 8(12): 2778-2790, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28925253

RESUMO

Multimodal chemical imaging using matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) can provide comprehensive molecular information in situ within the same tissue sections. This is of relevance for studying different brain pathologies such as Alzheimer's disease (AD), where recent data suggest a critical relevance of colocalizing Aß peptides and neuronal lipids. We here developed a novel trimodal, high-resolution (10 µm) MALDI imaging MS (IMS) paradigm for negative and positive ion mode lipid analysis and subsequent protein ion imaging on the same tissue section. Matrix sublimation of 1,5-diaminonaphthalene (1,5-DAN) enabled dual polarity lipid MALDI IMS on the same pixel points at high spatial resolutions (10 µm) and with high spectral quality. This was followed by 10 µm resolution protein imaging on the same measurement area, which allowed correlation of lipid signals with protein distribution patterns within distinct cerebellar regions in mouse brain. The demonstrated trimodal imaging strategy (IMS3) was further shown to be an efficient approach for simultaneously probing Aß plaque-associated lipids and Aß peptides within the hippocampus of 18 month-old transgenic AD mice (tgArcSwe). Here, IMS3 revealed a strong colocalization of distinct lipid species including ceramides, phosphatidylinositols, sulfatides (Cer 18:0, PI 38:4, ST 24:0) and lysophosphatidylcholines (LPC 16:0, LPC 18:0) with plaque-associated Aß isoforms (Aß 1-37, Aß 1-38, Aß 1-40). This highlights the potential of IMS3 as an alternative, superior approach to consecutively performed immuno-based Aß staining strategies. Furthermore, the IMS3 workflow allowed for multimodal in situ MS/MS analysis of both lipids and Aß peptides. Altogether, the here presented IMS3 approach shows great potential for comprehensive, high-resolution molecular analysis of histological features at cellular length scales with high chemical specificity. It therefore represents a powerful approach for probing the complex molecular pathology of, e.g., neurodegenerative diseases that are characterized by neurotoxic protein aggregation.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Metabolismo dos Lipídeos , Microscopia de Fluorescência/métodos , Placa Amiloide/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Doença de Alzheimer/patologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Imagem Molecular/métodos , Imagem Multimodal/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
3.
Anal Chem ; 89(8): 4685-4694, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28318232

RESUMO

Matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS) enables acquisition of spatial distribution maps for molecular species in situ. This can provide comprehensive insights on the pathophysiology of different diseases. However, current sample preparation and MALDI-IMS acquisition methods have limitations in preserving molecular and histological tissue morphology, resulting in interfered correspondence of MALDI-IMS data with subsequently acquired immunofluorescent staining results. We here investigated the histology compatibility of MALDI-IMS to image neuronal lipids in rodent brain tissue with subsequent immunohistochemistry and fluorescent staining of histological features. This was achieved by sublimation of a low ionization energy matrix compound, 1,5-diaminonapthalene (1,5-DAN), minimizing the number of low-energy laser shots. This yielded improved lipid spectral quality and speed of data acquisition and reduced matrix cluster formation along with preservation of specific histological information at cellular levels. This gentle, histology-compatible MALDI-IMS protocol also diminished thermal effects and mechanical stress created during nanosecond laser ablation processes that were prominent in subsequent immunofluorescent staining images but not with classical hematoxylin and eosin (H&E) staining on the same tissue section. Furthermore, this methodology proved to be a powerful strategy for investigating ß-amyloid (Aß) plaque-associated neuronal lipids as exemplified by performing high-resolution MALDI-IMS with subsequent fluorescent amyloid staining in a transgenic mouse model of Alzheimer's disease (tgSwe).


Assuntos
Lipídeos/análise , Neurônios/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
4.
Talanta ; 165: 84-91, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28153323

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder linked to protein aggregation, like more than twenty other human pathologies. One major protein incriminated in AD is the 42-residue amyloid-ß peptide (Aß1-42) which aggregates to form neurotoxic oligomers and fibrils. While, low molecular weight oligomers have been evidenced as neurotoxic species, only scarce methods raise the challenge to monitor the beginning of the aggregation process, called oligomerization. We propose here an innovative and fast monitoring of the time-dependent Aß1-42 oligomerization pattern by electrospray differential mobility analysis (ES-DMA). We developed a non-denaturing method based on ES-DMA to afford a real-time and direct characterization of the early, metastable and neurotoxic species. This technique provided their size distribution over time. At the beginning of the in vitro oligomerization process of Aß1-42, the size distribution is characterized by two populations with modal diameters around 3.5 and 4nm, corresponding to Monomer and Small oligomers. After few hours, larger species around 10nm are observed. The results were correlated to those obtained by capillary electrophoresis. We also demonstrated the ability of our method to evaluate Aß1-42 kinetics modulators. Thereby, ES-DMA provides new insights on Aß1-42 oligomerization in the presence of sugar-based peptidomimetic analogs which were recently described as modulators of Aß1-42 self-assembly and neurotoxicity inhibitors.


Assuntos
Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/química , Monitoramento de Medicamentos/métodos , Eletroforese Capilar/métodos , Espectrometria de Mobilidade Iônica/métodos , Doença de Alzheimer , Humanos , Técnicas In Vitro , Cinética
5.
ACS Chem Neurosci ; 8(2): 347-355, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984697

RESUMO

The major pathological hallmarks of Alzheimer's disease (AD) are the progressive aggregation and accumulation of beta-amyloid (Aß) and hyperphosphorylated tau protein into neurotoxic deposits. Aß aggregation has been suggested as the critical early inducer, driving the disease progression. However, the factors that promote neurotoxic Aß aggregation remain elusive. Imaging mass spectrometry (IMS) is a powerful technique to comprehensively elucidate the spatial distribution patterns of lipids, peptides, and proteins in biological tissue sections. In the present study, matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS)-based imaging was used on transgenic Alzheimer's disease mouse (tgArcSwe) brain tissue to investigate the sphingolipid microenvironment of individual Aß plaques and elucidate plaque-associated sphingolipid alterations. Multivariate data analysis was used to interrogate the IMS data for identifying pathologically relevant, anatomical features based on their lipid chemical profile. This approach revealed sphingolipid species that distinctly located to cortical and hippocampal deposits, whose Aß identity was further verified using fluorescent amyloid staining and immunohistochemistry. Subsequent multivariate statistical analysis of the spectral data revealed significant localization of gangliosides and ceramides species to Aß positive plaques, which was accompanied by distinct local reduction of sulfatides. These plaque-associated changes in sphingolipid levels implicate a functional role of sphingolipid metabolism in Aß plaque pathology and AD pathogenesis. Taken together, the presented data highlight the potential of imaging mass spectrometry as a powerful approach for probing Aß plaque-associated lipid changes underlying AD pathology.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , Esfingolipídeos/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Análise Multivariada , Mutação/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
6.
J Med Chem ; 59(5): 2025-40, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26789783

RESUMO

How anti-Alzheimer's drug candidates that reduce amyloid 1-42 peptide fibrillization interact with the most neurotoxic species is far from being understood. We report herein the capacity of sugar-based peptidomimetics to inhibit both Aß1-42 early oligomerization and fibrillization. A wide range of bio- and physicochemical techniques, such as a new capillary electrophoresis method, nuclear magnetic resonance, and surface plasmon resonance, were used to identify how these new molecules can delay the aggregation of Aß1-42. We demonstrate that these molecules interact with soluble oligomers in order to maintain the presence of nontoxic monomers and to prevent fibrillization. These compounds totally suppress the toxicity of Aß1-42 toward SH-SY5Y neuroblastoma cells, even at substoichiometric concentrations. Furthermore, demonstration that the best molecule combines hydrophobic moieties, hydrogen bond donors and acceptors, ammonium groups, and a hydrophilic ß-sheet breaker element provides valuable insight for the future structure-based design of inhibitors of Aß1-42 aggregation.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Desenho de Fármacos , Glicopeptídeos/farmacologia , Neuroblastoma/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptidomiméticos , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Glicopeptídeos/síntese química , Glicopeptídeos/química , Humanos , Estrutura Molecular , Neuroblastoma/patologia , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica/efeitos dos fármacos , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
7.
Electrophoresis ; 35(23): 3302-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25219962

RESUMO

We report an improved CE method to monitor in vitro the self-assembly of monomeric amyloid ß-peptide (42 amino acids amyloid ß-peptide, Aß1-42 ) and in particular the crucial early steps involved in the formation of the neurotoxic oligomers. In order to start the kinetics from the beginning, sample preparation was optimized to provide samples containing exclusively the monomeric form. The CE method was also improved using a dynamic coating and by reducing the separation distance. Using this method, the disappearance of the monomer as well as the progressive formation of four species during the self-assembly process can now be monitored and quantified over time. The hydrodynamic radius of the species present at the initial kinetics step was estimated around 1.8 nm by Taylor dispersion analysis while SDS-PAGE analyses showed the predominance of the monomer. These results confirmed that the Aß1-42 species present at this initial time was the monomer. Methylene blue, an anti-Alzheimer disease candidate, was then evaluated. In spite of an oligomerization inhibition, the enhanced disappearance of the Aß1-42 monomer provoked by methylene blue was demonstrated for the first time. This method, allowing the monomeric and smallest oligomeric species to be monitored, represents a new accurate and precise way to evaluate compounds for drug discovery.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Descoberta de Drogas/métodos , Eletroforese Capilar/métodos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer , Amiloide/química , Amiloide/metabolismo , Humanos , Cinética , Azul de Metileno
8.
Eur J Med Chem ; 86: 752-8, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25238173

RESUMO

Alzheimer's disease is a neurodegenerative disorder linked to oligomerization and fibrillization of amyloid ß peptides. Aß1-42 being the most aggregative and neurotoxic amyloid peptide, we report herein the capacity of sugar-based pentapeptide analogs to modulate the Aß1-42 aggregation process using thioflavin fluorescence and transmission electron microscopy assays. The importance of the free hydroxyl groups of the sugar moiety, used as a ß-breaker element, is confirmed since hydroxylated compounds inhibit the aggregation process while benzylated ones enhance it. Furthermore, the most effective molecules were also evaluated by a recently developed capillary electrophoresis method, providing in vitro monitoring of the crucial, very early stages of the self-assembly process. This technique allowed us to investigate the effect of these compounds on the small non-fibrillar Aß1-42 oligomers suspected by several groups worldwide as highly neurotoxic. We clearly demonstrated that molecules delaying the aggregation can stabilize the monomeric peptide or promote the formation of soluble oligomeric species. In contrast, molecules that accelerate the aggregation can prevent the presence of small toxic oligomers.


Assuntos
Peptídeos beta-Amiloides/química , Carboidratos/química , Oligopeptídeos/química , Fragmentos de Peptídeos/química , Peptidomiméticos/química , Peptidomiméticos/farmacologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Oligopeptídeos/efeitos dos fármacos , Fragmentos de Peptídeos/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Relação Estrutura-Atividade
9.
J Org Chem ; 77(3): 1316-27, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22141919

RESUMO

An efficient access to 2-substituted 3-arylbenzofurans through a palladium-catalyzed C3 direct arylation of 2-substituted benzofurans with aryl bromides is described. The scope and limitation of this reaction was studied. The method tolerates a variety of functional groups on the aryl halide and has been successfully extended to polysubstituted benzofurans to obtain the corresponding 3-arylbenzofurans with good to excellent yields.


Assuntos
Benzofuranos/química , Paládio/química , Catálise , Transporte de Elétrons
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...