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










Base de dados
Intervalo de ano de publicação
1.
Eur J Neurosci ; 49(9): 1091-1101, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30565792

RESUMO

Synapse degeneration and dendritic spine dysgenesis are believed to be crucial early steps in Alzheimer's disease (AD), and correlate with cognitive deficits in AD patients. Soluble amyloid beta (Aß)-derived oligomers, also termed Aß-derived diffusible ligands (ADDLs), accumulate in the brain of AD patients and play a crucial role in AD pathogenesis. ADDLs bind to mature hippocampal neurons, induce structural changes in dendritic spines and contribute to neuronal death. However, mechanisms underlying structural and toxic effects are not fully understood. Here, we report that ADDLs bind to cultured mature cortical pyramidal neurons and induce spine dysgenesis. ADDL treatment induced the rapid depletion of kalirin-7, a brain-specific guanine-nucleotide exchange factor for the small GTPase Rac1, from spines. Kalirin-7 is a key regulator of dendritic spine morphogenesis and maintenance in forebrain pyramidal neurons and here we show that overexpression of kalirin-7 prevents ADDL-induced spine degeneration. Taken together, our results suggest that kalirin-7 may play a role in the early events leading to synapse degeneration, and its pharmacological activation may prevent or delay synapse pathology in AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Animais , Células Cultivadas , Degeneração Neural , Células Piramidais/metabolismo , Células Piramidais/patologia , Ratos , Ratos Sprague-Dawley
2.
J Neurochem ; 142(6): 934-947, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28670737

RESUMO

Brain accumulation of soluble oligomers of the amyloid-ß peptide (AßOs) is increasingly considered a key early event in the pathogenesis of Alzheimer's disease (AD). A variety of AßO species have been identified, both in vitro and in vivo, ranging from dimers to 24mers and higher order oligomers. However, there is no consensus in the literature regarding which AßO species are most germane to AD pathogenesis. Antibodies capable of specifically recognizing defined subpopulations of AßOs would be a valuable asset in the identification, isolation, and characterization of AD-relevant AßO species. Here, we report the characterization of a human single chain antibody fragment (scFv) denoted NUsc1, one of a number of scFvs we have identified that stringently distinguish AßOs from both monomeric and fibrillar Aß. NUsc1 readily detected AßOs previously bound to dendrites in cultured hippocampal neurons. In addition, NUsc1 blocked AßO binding and reduced AßO-induced neuronal oxidative stress and tau hyperphosphorylation in cultured neurons. NUsc1 further distinguished brain extracts from AD-transgenic mice from wild type (WT) mice, and detected endogenous AßOs in fixed AD brain tissue and AD brain extracts. Biochemical analyses indicated that NUsc1 targets a subpopulation of AßOs with apparent molecular mass greater than 50 kDa. Results indicate that NUsc1 targets a particular AßO species relevant to AD pathogenesis, and suggest that NUsc1 may constitute an effective tool for AD diagnostics and therapeutics.

3.
Neurobiol Aging ; 36(6): 2043-52, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25862419

RESUMO

Amyloid-ß (Aß) oligomers are the suspected culprit as initiators of Alzheimer's disease (AD). However, their diffusion in the brain remains unknown. Here, we studied Aß oligomers' dissemination and evaluated their in vivo toxicity. Wild-type mice were injected with 50 pmol of synthetic Aß oligomers (of different size) in the hippocampus. Oligomers diffused largely in the brain as soon as 1 hour and up to 7 days after injection. A transient encephalopathy with memory impairment was induced by this unique injection. The immunoreactivity of the postsynaptic marker PSD95 was diffusely decreased. Similar results (both on memory and PSD95 immunoreactivity) were obtained with delipidated and high molecular weight oligomers (>50 kDa) but not with smaller assemblies. Tau hyperphosphorylation was observed in the oligomer-injected brains. Finally, fos immunostaining was increased in Aß-derived diffusible ligands-injected mice, suggesting neuronal hyperactivity. Rapid and widespread diffusion of Aß oligomers was demonstrated in vivo and associated with decreased synaptic markers and memory deficits which gives new insight to the pathogenicity of Aß.


Assuntos
Amnésia/induzido quimicamente , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Encefalopatias/induzido quimicamente , Doença Aguda , Doença de Alzheimer/etiologia , Amnésia/metabolismo , Peptídeos beta-Amiloides/administração & dosagem , Animais , Transporte Biológico , Biopolímeros , Encéfalo/metabolismo , Encefalopatias/metabolismo , Difusão , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases/metabolismo , Hipocampo , Injeções , Masculino , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Peso Molecular , Fosforilação , Sinapses/efeitos dos fármacos , Fatores de Tempo , Proteínas tau/metabolismo
4.
J Comp Neurol ; 521(18): 4318-38, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23881733

RESUMO

The two major histopathologic hallmarks of Alzheimer's disease (AD) are amyloid beta protein (Aß) plaques and neurofibrillary tangles (NFT). Aß pathology is a common feature in the aged nonhuman primate brain, whereas NFT are found almost exclusively in humans. Few studies have examined AD-related pathology in great apes, which are the closest phylogenetic relatives of humans. In the present study, we examined Aß and tau-like lesions in the neocortex and hippocampus of aged male and female western lowland gorillas using immunohistochemistry and histochemistry. Analysis revealed an age-related increase in Aß-immunoreactive plaques and vasculature in the gorilla brain. Aß plaques were more abundant in the neocortex and hippocampus of females, whereas Aß-positive blood vessels were more widespread in male gorillas. Plaques were also Aß40-, Aß42-, and Aß oligomer-immunoreactive, but only weakly thioflavine S- or 6-CN-PiB-positive in both sexes, indicative of the less fibrillar (diffuse) nature of Aß plaques in gorillas. Although phosphorylated neurofilament immunostaining revealed a few dystrophic neurites and neurons, choline acetyltransferase-immunoreactive fibers were not dystrophic. Neurons stained for the tau marker Alz50 were found in the neocortex and hippocampus of gorillas at all ages. Occasional Alz50-, MC1-, and AT8-immunoreactive astrocyte and oligodendrocyte coiled bodies and neuritic clusters were seen in the neocortex and hippocampus of the oldest gorillas. This study demonstrates the spontaneous presence of both Aß plaques and tau-like lesions in the neocortex and hippocampus in old male and female western lowland gorillas, placing this species at relevance in the context of AD research.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/veterinária , Hipocampo/patologia , Neocórtex/patologia , Doença de Alzheimer/metabolismo , Animais , Feminino , Gorilla gorilla , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Neocórtex/metabolismo , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Proteínas tau/metabolismo
5.
Neurobiol Aging ; 34(9): 2081-90, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23582659

RESUMO

Amyloid-ß peptide (Aß) fragment misfolding may play a crucial role in the progression of Alzheimer's disease (AD) pathophysiology as well as epigenetic mechanisms at the DNA and histone level. We hypothesized that histone H3 homeostasis is disrupted in association with the appearance of soluble Aß at an early stage in AD progression. We identified, localized, and compared histone H3 modifications in multiple model systems (neural-like SH-SY5Y, primary neurons, Tg2576 mice, and AD neocortex), and narrowed our focus to investigate 3 key motifs associated with regulating transcriptional activation and inhibition: acetylated lysine 14, phosphorylated serine 10 and dimethylated lysine 9. Our results in vitro and in vivo indicate that multimeric soluble Aß may be a potent signaling molecule indirectly modulating the transcriptional activity of DNA by modulating histone H3 homeostasis. These findings reveal potential loci of transcriptional disruption relevant to AD. Identifying genes that undergo significant epigenetic alterations in response to Aß could aid in the understanding of the pathogenesis of AD, as well as suggesting possible new treatment strategies.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/fisiologia , Histonas/metabolismo , Homeostase/genética , Neocórtex/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , DNA/genética , Epigênese Genética/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Terapia de Alvo Molecular , Neuroblastoma/metabolismo , Neurônios/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Solubilidade , Ativação Transcricional
6.
J Alzheimers Dis ; 35(4): 777-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23542865

RESUMO

Alzheimer's disease (AD) is a global health crisis with limited treatment options. Despite major advances in neurotherapeutics, poor brain penetration due to the blood-brain barrier continues to pose a big challenge in overcoming the access of therapeutics to the central nervous system. In that regard, the non-invasive intranasal route of brain targeting is gaining considerable attention. The nasal mucosa offers a large surface area, rapid absorption, and avoidance of first-pass metabolism increasing drug bioavailability with less systemic side effects. Intranasal delivery is known to utilize olfactory, rostral migratory stream, and trigeminal routes to reach the brain. This investigation confirmed that intranasal delivery of oligomeric amyloid-ß antibody (NU4) utilized all three routes to enter the brain with a resident time of 96 hours post single bolus intranasal administration, and showed evidence of perikaryal and parenchymal uptake of NU4 in 5XFAD mouse brain, confirming the intranasal route as a non-invasive and efficient way of delivering therapeutics to the brain. In addition, this study demonstrated that intranasal delivery of NU4 antibody lowered cerebral amyloid-ß and improved spatial learning in 5XFAD mice.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/imunologia , Anticorpos/metabolismo , Anticorpos/uso terapêutico , Encéfalo/metabolismo , Administração Intranasal , Animais , Anticorpos/administração & dosagem , Cognição/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Nervo Trigêmeo/efeitos dos fármacos
7.
ACS Chem Neurosci ; 3(11): 972-81, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23173076

RESUMO

Amyloid ß42 self-assembly is complex, with multiple pathways leading to large insoluble fibrils or soluble oligomers. Oligomers are now regarded as most germane to Alzheimer's pathogenesis. We have investigated the hypothesis that oligomer formation itself occurs through alternative pathways, with some leading to synapse-binding toxins. Immediately after adding synthetic peptide to buffer, solutions of Aß42 were separated by a 50 kDa filter and fractions assessed by SDS-PAGE silver stain, Western blot, immunoprecipitation, and capacity for synaptic binding. Aß42 rapidly assembled into aqueous-stable oligomers, with similar protein abundance in small (<50 kDa) and large (>50 kDa) oligomer fractions. Initially, both fractions were SDS-labile and resolved into tetramers, trimers, and monomers by SDS-PAGE. Upon continued incubation, the larger oligomers developed a small population of SDS-stable 10-16mers, and the smaller oligomers generated gel-impermeant complexes. The two fractions associated differently with neurons, with prominent synaptic binding limited to larger oligomers. Even within the family of larger oligomers, synaptic binding was associated with only a subset of these species, as a new scFv antibody (NUsc1) immunoprecipitated only a small portion of the oligomers while eliminating synaptic binding. Interestingly, low doses of the peptide KLVFFA blocked assembly of the 10-16mers, and this result was associated with loss of the smaller clusters of oligomers observed at synaptic sites. What distinguishes these smaller clusters from the unaffected larger clusters is not yet known. Results indicate that distinct species of Aß oligomers are generated by alternative assembly pathways and that synapse-binding subpopulations of Aß oligomers could be specifically targeted for Alzheimer's therapeutics.


Assuntos
Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/farmacologia , Anticorpos de Cadeia Única/farmacologia , Sinapses/química , Western Blotting , Eletroforese em Gel de Poliacrilamida , Humanos , Imunoprecipitação
8.
Aging Cell ; 11(5): 823-33, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22708890

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting both the hippocampus and the cerebral cortex. Reduced synaptic density that occurs early in the disease process seems to be partially due to the overactivation of N-methyl-d-aspartate receptors (NMDARs) leading to excitotoxicity. Recently, we demonstrated that amyloid-beta oligomers (AßO), the species implicated in synaptic loss during the initial disease stages, induce endoplasmic reticulum (ER) stress in cultured neurons. Here, we investigated whether AßO trigger ER stress by an NMDAR-dependent mechanism leading to neuronal dysfunction and analyzed the contribution of GluN2A and GluN2B subunits of this glutamate receptor. Our data revealed that AßO induce ER stress in mature hippocampal cultures, activating ER stress-associated sensors and increasing the levels of the ER chaperone GRP78. We also showed that AßO induce NADPH oxidase (NOX)-mediated superoxide production downstream of GluN2B and impairs ER and cytosolic Ca2+ homeostasis. These events precede changes in cell viability and activation of the ER stress-mediated apoptotic pathway, which was associated with translocation of the transcription factor GADD153 / CHOP to the nucleus and occurred by a caspase-12-independent mechanism. Significantly, ER stress took place after AßO interaction with GluN2B subunits. In addition, AßO-induced ER stress and hippocampal dysfunction were prevented by ifenprodil, an antagonist of GluN2B subunits, while the GluN2A antagonist NVP-AAM077 only slightly attenuated AßO-induced neurotoxicity. Taken together, our results highlight the role of GluN2B subunit of NMDARs on ER stress-mediated hippocampal dysfunction caused by AßO suggesting that it might be a potential therapeutic target during the early stages of AD.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Estresse do Retículo Endoplasmático/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Peptídeos beta-Amiloides/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos
9.
Neurobiol Aging ; 33(4): 825.e1-13, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21683475

RESUMO

In this study, we examined the relationship between various ß-amyloid (Aß) oligomer assemblies in autopsy brain with the levels of fibrillar Aß and cholinergic synaptic function. Brain tissues obtained from the frontal cortex of 14 Alzheimer's disease (AD) patients grouped into early-onset AD (EOAD) and late-onset AD (LOAD) and 12 age-matched control subjects were used to extract and quantify Aß oligomers in soluble (TBS), detergent soluble (TBST), and insoluble (GuHCl) fractions. The predominant oligomeric Aß assemblies detected were dodecamers, decamers, and pentamers, and different patterns of expression were observed between EOAD and LOAD patients. There was no association between any of the detected Aß oligomer assemblies and fibrillar Aß levels measured by N-methyl[(3)H] 2-(40-methylaminophenyl)-6-hydroxy-benzothiazole ([(3)H]PIB) binding. Levels of pentamers in the soluble fraction significantly correlated with a reduction in choline acetyltransferase activity in AD patients. The number of nicotinic acetylcholine receptors negatively correlated with the total amount of Aß oligomers in the insoluble fraction in EOAD patients, and with decamers in the soluble fraction in LOAD patients. These novel findings suggest that distinct Aß oligomers induce impairment of cholinergic neurotransmission in AD pathogenesis.


Assuntos
Idade de Início , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Colinérgicos/metabolismo , Fragmentos de Peptídeos/metabolismo , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Compostos de Anilina , Apolipoproteínas E/genética , Benzotiazóis/farmacocinética , Encéfalo/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Genótipo , Humanos , Imunoprecipitação , Masculino , Pessoa de Meia-Idade , Nicotina/farmacocinética , Agonistas Nicotínicos/farmacocinética , Mudanças Depois da Morte , Ligação Proteica/efeitos dos fármacos , Estatística como Assunto , Tiazóis , Trítio/farmacocinética
10.
Mol Neurodegener ; 6(1): 61, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21861874

RESUMO

BACKGROUND: The ß-amyloid peptide (Aß) contains a Gly-XXX-Gly-XXX-Gly motif in its C-terminal region that has been proposed to form a "glycine zipper" that drives the formation of toxic Aß oligomers. We have tested this hypothesis by examining the toxicity of Aß variants containing substitutions in this motif using a neuronal cell line, primary neurons, and a transgenic C. elegans model. RESULTS: We found that a Gly37Leu substitution dramatically reduced Aß toxicity in all models tested, as measured by cell dysfunction, cell death, synaptic alteration, or tau phosphorylation. We also demonstrated in multiple models that Aß Gly37Leu is actually anti-toxic, thereby supporting the hypothesis that interference with glycine zipper formation blocks assembly of toxic Aß oligomers. To test this model rigorously, we engineered second site substitutions in Aß predicted by the glycine zipper model to compensate for the Gly37Leu substitution and expressed these in C. elegans. We show that these second site substitutions restore in vivo Aßtoxicity, further supporting the glycine zipper model. CONCLUSIONS: Our structure/function studies support the view that the glycine zipper motif present in the C-terminal portion of Aß plays an important role in the formation of toxic Aß oligomers. Compounds designed to interfere specifically with formation of the glycine zipper could have therapeutic potential.

11.
Cell Mol Neurobiol ; 31(6): 939-48, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21538118

RESUMO

Aß oligomers cause a collection of molecular events associated with memory loss in Alzheimer's disease, centering on disrupting the maintenance of synapse structure and function. In this brief review of the synaptotoxic effects of Aß oligomers, we focus on the neuronal properties governing oligomer targeting and toxicity-especially with respect to binding sites and mechanisms of binding. We also discuss ways in which mechanistic insights from other diseases offer clues in the pursuit of the molecular basis of Alzheimer's disease.


Assuntos
Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Degeneração Neural/complicações , Sinapses/patologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Humanos , Memória/efeitos dos fármacos , Degeneração Neural/fisiopatologia , Estrutura Quaternária de Proteína , Sinapses/efeitos dos fármacos
12.
Neuron ; 66(5): 739-54, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20547131

RESUMO

Soluble oligomers of amyloid beta (Abeta) play a role in the memory impairment characteristic of Alzheimer's disease. Acting as pathogenic ligands, Abeta oligomers bind to particular synapses and perturb their function, morphology, and maintenance. Events that occur shortly after oligomer binding have been investigated here in live hippocampal neurons by single particle tracking of quantum dot-labeled oligomers and synaptic proteins. Membrane-attached oligomers initially move freely, but their diffusion is hindered markedly upon accumulation at synapses. Concomitantly, individual metabotropic glutamate receptors (mGluR5) manifest strikingly reduced lateral diffusion as they become aberrantly clustered. This clustering of mGluR5 elevates intracellular calcium and causes synapse deterioration, responses prevented by an mGluR5 antagonist. As expected, clustering by artificial crosslinking also promotes synaptotoxicity. These results reveal a mechanism whereby Abeta oligomers induce the abnormal accumulation and overstabilization of a glutamate receptor, thus providing a mechanistic and molecular basis for Abeta oligomer-induced early synaptic failure.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Matriz Extracelular/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Células Cultivadas , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Ligação Proteica/fisiologia , Estabilidade Proteica , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/fisiologia , Sinapses/metabolismo , Sinapses/patologia
13.
J Alzheimers Dis ; 18(4): 787-98, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19661619

RESUMO

Loss of synapses has been correlated with dementia in Alzheimer's disease (AD) as an early event during the disease progression. Hence, synaptogenesis and neurogenesis in adulthood could serve as a therapeutic target for the prevention and treatment of AD. Recently, we have demonstrated enhanced hippocampal neurogenesis by oral administration of Ginkgo biloba extract (EGb 761) to a mouse model of AD. This study aims to identify the constituents that contribute to EGb 761-induced neurogenesis. Among the constituents tested, bilobalide and quercetin significantly increased cell proliferation in the hippocampal neurons in a dose-dependent manner. Bilobalide and quercetin also enhanced phosphorylation of cyclic-AMP Response Element Binding Protein (CREB) in these cells, and elevated the levels of pCREB and, brain-derived neurotrophic factor in mice brain. Immunofluorescence staining of synaptic markers shows remarkable dendritic processes in hippocampal neurons treated with either quercetin or bilobalide. Furthermore, both constituents restored amyloid-beta oligomers (also known as ADDL)-induced synaptic loss and phosphorylation of CREB. The present findings suggest that enhanced neurogenesis and synaptogenesis by bilobalide and quercetin may share a common final signaling pathway mediated by phosphorylation of CREB. Despite a recent report showing that EGb 761 was insufficient in prevent dementia, its constituents still warrant future investigation.


Assuntos
Doença de Alzheimer/fisiopatologia , Ciclopentanos/farmacologia , Furanos/farmacologia , Ginkgolídeos/farmacologia , Hipocampo/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Extratos Vegetais/farmacologia , Quercetina/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Animais , Western Blotting , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ginkgo biloba , Hipocampo/metabolismo , Camundongos , Fosforilação , Ratos , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos
14.
J Biol Chem ; 284(28): 18742-53, 2009 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-19406747

RESUMO

Accumulation of amyloid beta (Abeta) oligomers in the brain is toxic to synapses and may play an important role in memory loss in Alzheimer disease. However, how these toxins are built up in the brain is not understood. In this study we investigate whether impairments of insulin and insulin-like growth factor-1 (IGF-1) receptors play a role in aggregation of Abeta. Using primary neuronal culture and immortal cell line models, we show that expression of normal insulin or IGF-1 receptors confers cells with abilities to reduce exogenously applied Abeta oligomers (also known as ADDLs) to monomers. In contrast, transfection of malfunctioning human insulin receptor mutants, identified originally from patient with insulin resistance syndrome, or inhibition of insulin and IGF-1 receptors via pharmacological reagents increases ADDL levels by exacerbating their aggregation. In healthy cells, activation of insulin and IGF-1 receptor reduces the extracellular ADDLs applied to cells via seemingly the insulin-degrading enzyme activity. Although insulin triggers ADDL internalization, IGF-1 appears to keep ADDLs on the cell surface. Nevertheless, both insulin and IGF-1 reduce ADDL binding, protect synapses from ADDL synaptotoxic effects, and prevent the ADDL-induced surface insulin receptor loss. Our results suggest that dysfunctions of brain insulin and IGF-1 receptors contribute to Abeta aggregation and subsequent synaptic loss.


Assuntos
Peptídeos beta-Amiloides/química , Antígenos CD/química , Insulina/química , Receptor de Insulina/química , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Humanos , Insulina/metabolismo , Resistência à Insulina , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Modelos Biológicos , Mutação , Células NIH 3T3 , Neurônios/metabolismo , Neurotoxinas/química , Ratos , Sinapses/metabolismo
15.
Neurobiol Aging ; 29(9): 1334-47, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17403556

RESUMO

Alzheimer's disease (AD) is characterized by presence of extracellular fibrillar A beta in amyloid plaques, intraneuronal neurofibrillary tangles consisting of aggregated hyperphosphorylated tau and elevated brain levels of soluble A beta oligomers (ADDLs). A major question is how these disparate facets of AD pathology are mechanistically related. Here we show that, independent of the presence of fibrils, ADDLs stimulate tau phosphorylation in mature cultures of hippocampal neurons and in neuroblastoma cells at epitopes characteristically hyperphosphorylated in AD. A monoclonal antibody that targets ADDLs blocked their attachment to synaptic binding sites and prevented tau hyperphosphorylation. Tau phosphorylation was blocked by the Src family tyrosine kinase inhibitor, 4-amino-5-(4-chlorophenyl)-7(t-butyl)pyrazol(3,4-D)pyramide (PP1), and by the phosphatidylinositol-3-kinase inhibitor LY294002. Significantly, tau hyperphosphorylation was also induced by a soluble aqueous extract containing A beta oligomers from AD brains, but not by an extract from non-AD brains. A beta oligomers have been increasingly implicated as the main neurotoxins in AD, and the current results provide a unifying mechanism in which oligomer activity is directly linked to tau hyperphosphorylation in AD pathology.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/administração & dosagem , Hipocampo/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Células Cultivadas , Hipocampo/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Fosforilação/efeitos dos fármacos
16.
J Neurosci ; 27(4): 796-807, 2007 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-17251419

RESUMO

The basis for memory loss in early Alzheimer's disease (AD) seems likely to involve synaptic damage caused by soluble Abeta-derived oligomers (ADDLs). ADDLs have been shown to build up in the brain and CSF of AD patients and are known to interfere with mechanisms of synaptic plasticity, acting as gain-of-function ligands that attach to synapses. Because of the correlation between AD dementia and synaptic degeneration, we investigated here the ability of ADDLs to affect synapse composition, structure, and abundance. Using highly differentiated cultures of hippocampal neurons, a preferred model for studies of synapse cell biology, we found that ADDLs bound to neurons with specificity, attaching to presumed excitatory pyramidal neurons but not GABAergic neurons. Fractionation of ADDLs bound to forebrain synaptosomes showed association with postsynaptic density complexes containing NMDA receptors, consistent with observed attachment of ADDLs to dendritic spines. During binding to hippocampal neurons, ADDLs promoted a rapid decrease in membrane expression of memory-related receptors (NMDA and EphB2). Continued exposure resulted in abnormal spine morphology, with induction of long thin spines reminiscent of the morphology found in mental retardation, deafferentation, and prionoses. Ultimately, ADDLs caused a significant decrease in spine density. Synaptic deterioration, which was accompanied by decreased levels of the spine cytoskeletal protein drebrin, was blocked by the Alzheimer's therapeutic drug Namenda. The observed disruption of dendritic spines links ADDLs to a major facet of AD pathology, providing strong evidence that ADDLs in AD brain cause neuropil damage believed to underlie dementia.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Forma Celular , Sinapses/patologia , Peptídeos beta-Amiloides/fisiologia , Animais , Contagem de Células , Forma Celular/efeitos dos fármacos , Forma Celular/fisiologia , Células Cultivadas , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Sinapses/efeitos dos fármacos , Sinapses/fisiologia
17.
J Neurochem ; 100(1): 23-35, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17116235

RESUMO

Amyloid beta (Abeta) immunotherapy for Alzheimer's disease has shown initial success in mouse models of Alzheimer's disease and in human patients. However, because of meningoencephalitis in clinical trials of active vaccination, approaches using therapeutic antibodies may be preferred. As a novel antigen to generate monoclonal antibodies, the current study has used Abeta oligomers (amyloid beta-derived diffusible ligands, ADDLs), pathological assemblies known to accumulate in Alzheimer's disease brain. Clones were selected for the ability to discriminate Alzheimer's disease from control brains in extracts and tissue sections. These antibodies recognized Abeta oligomers and fibrils but not the physiologically prevalent Abeta monomer. Discrimination derived from an epitope found in assemblies of Abeta1-28 and ADDLs but not in other sequences, including Abeta1-40. Immunoneutralization experiments showed that toxicity and attachment of ADDLs to synapses in culture could be prevented. ADDL-induced reactive oxygen species (ROS) generation was also inhibited, establishing this response to be oligomer-dependent. Inhibition occurred whether ADDLs were prepared in vitro or obtained from Alzheimer's disease brain. As conformationally sensitive monoclonal antibodies that selectively immunoneutralize binding and function of pathological Abeta assemblies, these antibodies provide tools by which pathological Abeta assemblies from Alzheimer's disease brain might be isolated and evaluated, as well as offering a valuable prototype for new antibodies useful for Alzheimer's disease therapeutics.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/imunologia , Anticorpos Monoclonais/fisiologia , Especificidade de Anticorpos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação a Calmodulina/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Epitopos , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Immunoblotting/métodos , Imuno-Histoquímica/métodos , Camundongos , Neurônios/metabolismo , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Coelhos , Espécies Reativas de Oxigênio/metabolismo , Sais de Tetrazólio , Tiazóis
18.
J Neurosci ; 24(45): 10191-200, 2004 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-15537891

RESUMO

The cognitive hallmark of early Alzheimer's disease (AD) is an extraordinary inability to form new memories. For many years, this dementia was attributed to nerve-cell death induced by deposits of fibrillar amyloid beta (Abeta). A newer hypothesis has emerged, however, in which early memory loss is considered a synapse failure caused by soluble Abeta oligomers. Such oligomers rapidly block long-term potentiation, a classic experimental paradigm for synaptic plasticity, and they are strikingly elevated in AD brain tissue and transgenic-mouse AD models. The current work characterizes the manner in which Abeta oligomers attack neurons. Antibodies raised against synthetic oligomers applied to AD brain sections were found to give diffuse stain around neuronal cell bodies, suggestive of a dendritic pattern, whereas soluble brain extracts showed robust AD-dependent reactivity in dot immunoblots. Antigens in unfractionated AD extracts attached with specificity to cultured rat hippocampal neurons, binding within dendritic arbors at discrete puncta. Crude fractionation showed ligand size to be between 10 and 100 kDa. Synthetic Abeta oligomers of the same size gave identical punctate binding, which was highly selective for particular neurons. Image analysis by confocal double-label immunofluorescence established that >90% of the punctate oligomer binding sites colocalized with the synaptic marker PSD-95 (postsynaptic density protein 95). Synaptic binding was accompanied by ectopic induction of Arc, a synaptic immediate-early gene, the overexpression of which has been linked to dysfunctional learning. Results suggest the hypothesis that targeting and functional disruption of particular synapses by Abeta oligomers may provide a molecular basis for the specific loss of memory function in early AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/farmacologia , Fragmentos de Peptídeos/farmacologia , Sinapses/química , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/química , Animais , Sítios de Ligação , Córtex Cerebral/química , Proteínas do Líquido Cefalorraquidiano/metabolismo , Cromatografia Líquida de Alta Pressão , Proteínas do Citoesqueleto/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Masculino , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Fragmentos de Peptídeos/química , Ligação Proteica , Ratos , Sinapses/fisiologia , Extratos de Tecidos/metabolismo , Extratos de Tecidos/farmacologia
19.
Biochemistry ; 42(44): 12749-60, 2003 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-14596589

RESUMO

Amyloid beta 1-42 (Abeta(1-42)) is a self-associating peptide that becomes neurotoxic upon aggregation. Toxicity originally was attributed to the presence of large, readily formed Abeta fibrils, but a variety of other toxic species are now known. The current study shows that Abeta(1-42) can self-assemble into small, stable globular assemblies free of fibrils and protofibrils. Absence of large molecules was verified by atomic force microscopy (AFM) and nondenaturing gel electrophoresis. Denaturing electrophoresis revealed that the globular assemblies comprised oligomers ranging from trimers to 24mers. Oligomers prepared at 4 degrees C stayed fibril-free for days and remained so when shifted to 37 degrees C, although the spectrum of sizes shifted toward larger oligomers at the higher temperature. The soluble, globular Abeta(1-42) oligomers were toxic to PC12 cells, impairing reduction of MTT and interfering with ERK and Rac signal transduction. Occasionally, oligomers were neither toxic nor recognized by toxicity-neutralizing antibodies, suggesting that oligomers could assume alternative conformations. Tests for oligomerization-blocking activity were carried out by dot-blot immunoassays and showed that neuroprotective extracts of Ginkgo biloba could inhibit oligomer formation at very low doses. The observed neurotoxicity, structure, and stability of synthetic Abeta(1-42) globular assemblies support the hypothesis that Abeta(1-42) oligomers play a role in triggering nerve cell dysfunction and death in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Neurotoxinas/química , Neurotoxinas/toxicidade , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/toxicidade , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/isolamento & purificação , Animais , Temperatura Corporal , Sobrevivência Celular/efeitos dos fármacos , Difusão , Epitopos/química , Ginkgo biloba , Humanos , Ligantes , Fármacos Neuroprotetores/química , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/isolamento & purificação , Nitroazul de Tetrazólio/química , Células PC12 , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/isolamento & purificação , Extratos Vegetais/química , Conformação Proteica , Ratos
20.
Proc Natl Acad Sci U S A ; 100(18): 10417-22, 2003 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12925731

RESUMO

A molecular basis for memory failure in Alzheimer's disease (AD) has been recently hypothesized, in which a significant role is attributed to small, soluble oligomers of amyloid beta-peptide (A beta). A beta oligomeric ligands (also known as ADDLs) are known to be potent inhibitors of hippocampal long-term potentiation, which is a paradigm for synaptic plasticity, and have been linked to synapse loss and reversible memory failure in transgenic mouse AD models. If such oligomers were to build up in human brain, their neurological impact could provide the missing link that accounts for the poor correlation between AD dementia and amyloid plaques. This article, using antibodies raised against synthetic A beta oligomers, verifies the predicted accumulation of soluble oligomers in AD frontal cortex. Oligomers in AD reach levels up to 70-fold over control brains. Brain-derived and synthetic oligomers show structural equivalence with respect to mass, isoelectric point, and recognition by conformation-sensitive antibodies. Both oligomers, moreover, exhibit the same striking patterns of attachment to cultured hippocampal neurons, binding on dendrite surfaces in small clusters with ligand-like specificity. Binding assays using solubilized membranes show oligomers to be high-affinity ligands for a small number of nonabundant proteins. Current results confirm the prediction that soluble oligomeric A beta ligands are intrinsic to AD pathology, and validate their use in new approaches to therapeutic AD drugs and vaccines.


Assuntos
Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/metabolismo , Química Encefálica , Oligopeptídeos/análise , Fragmentos de Peptídeos/análise , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Córtex Cerebral/química , Córtex Cerebral/metabolismo , Immunoblotting , Ligantes , Potenciação de Longa Duração , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Extratos de Tecidos/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...