Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neurochem Res ; 44(6): 1346-1355, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29572646

RESUMO

The function of the ß-A4 amyloid protein precursor (APP) of Alzheimer's disease (AD) remains unclear. APP has a number of putative roles in neuronal differentiation, survival, synaptogenesis and cell adhesion. In this study, we examined the development of axons, dendrites and synapses in cultures of hippocampus neutrons derived from APP knockout (KO) mice. We report that loss of APP function reduces the branching of cultured hippocampal neurons, resulting in reduced synapse formation. Using a compartmentalised culture approach, we found reduced axonal outgrowth in cultured hippocampal neurons and we also identified abnormal growth characteristics of isolated hippocampal neuron axons. Although APP has previously been suggested to play an important role in promoting cell adhesion, we surprisingly found that APPKO hippocampal neurons adhered more strongly to a poly-L-lysine substrate and their neurites displayed an increased density of focal adhesion puncta. The findings suggest that the function of APP has an important role in both dendritic and axonal growth and that endogenous APP may regulate substrate adhesion of hippocampal neurons. The results may explain neuronal and synaptic morphological abnormalities in APPKO mice and the presence of abnormal APP expression in dystrophic neurites around amyloid deposits in AD.


Assuntos
Precursor de Proteína beta-Amiloide/deficiência , Axônios/metabolismo , Dendritos/metabolismo , Hipocampo/metabolismo , Sinapses/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Adesão Celular/fisiologia , Feminino , Técnicas de Inativação de Genes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Crescimento Neuronal/fisiologia , Gravidez
2.
J Neurosci Res ; 95(4): 992-999, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27546887

RESUMO

Alzheimer's disease (AD) is a complex, progressive neurological disorder characterized by the formation of extracellular amyloid plaques composed of ß-amyloid protein (Aß), the key component in pathogenesis of AD. Peripheral administration of enoxaparin (ENO) reportedly reduces the level of Aß and the amyloid plaques in the cortex of amyloid precursor protein (APP) transgenic mice. However, the exact mechanism of these effects is unclear. Our previous studies indicated that ENO can inhibit APP processing to Aß in primary cortical cells from Tg2576 mice by downregulating BACE1 levels. This study examines whether ENO-induced reduction of amyloid load is due to the decreased APP processing to Aß in Tg2576 mice. Surprisingly, our results indicated that ENO significantly increases the Aß42/Aß40 ratio in cortex and enhances the amyloid plaque load in both cortex and hippocampus, although overall APP processing was not influenced by ENO. Moreover, ENO stimulated the aggregation of both Aß40 and Aß42 in vitro. Although ENO has been reported to improve cognition in vivo and has potential as a therapeutic agent for AD, the results from our study suggest that ENO can exacerbate the amyloid pathology, and the strategy of using ENO for the treatment of AD may require further assessment. © 2016 Wiley Periodicals, Inc.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Enoxaparina/toxicidade , Fibrinolíticos/toxicidade , Placa Amiloide/induzido quimicamente , Proteína ADAM10/metabolismo , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Placa Amiloide/genética , Agregados Proteicos/efeitos dos fármacos , Agregados Proteicos/genética
3.
J Biol Chem ; 289(45): 31253-61, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25217641

RESUMO

Amyloid-ß precursor protein (APP) is well studied for its role in Alzheimer disease, although its normal function remains uncertain. It has been reported that APP stimulates the proliferation and neuronal differentiation of neural stem/progenitor cells (NSPCs). In this study we examined the role of APP in NSPC differentiation. To identify proteins that may mediate the effect of APP on NSPC differentiation, we used a gene array approach to find genes whose expression correlated with APP-induced neurogenesis. We found that the expression of neurogenin 2 (Ngn2), a basic helix-loop-helix transcription factor, was significantly down-regulated in NSPCs from APP knock-out mice (APPKO) and increased in APP transgenic (Tg2576) mice. Ngn2 overexpression in APPKO NSPCs promoted neuronal differentiation, whereas siRNA knockdown of Ngn2 expression in wild-type NSPCs decreased neuronal differentiation. The results demonstrate that APP-stimulated neuronal differentiation of NSPCs is mediated by Ngn2.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Células-Tronco Neurais/citologia , Neurogênese , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Regulação para Baixo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo
4.
J Biol Chem ; 288(26): 18853-62, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23671283

RESUMO

The amyloid precursor protein (APP) is well studied for its role in Alzheimer disease. However, little is known about its normal function. In this study, we examined the role of APP in neural stem/progenitor cell (NSPC) proliferation. NSPCs derived from APP-overexpressing Tg2576 transgenic mice proliferated more rapidly than NSPCs from the corresponding background strain (C57Bl/6xSJL) wild-type mice. In contrast, NSPCs from APP knock-out (APP-KO) mice had reduced proliferation rates when compared with NSPCs from the corresponding background strain (C57Bl/6). A secreted factor, identified as cystatin C, was found to be responsible for this effect. Levels of cystatin C were higher in the Tg2576 conditioned medium and lower in the APP-KO conditioned medium. Furthermore, immunodepletion of cystatin C from the conditioned medium completely removed the ability of the conditioned medium to increase NSPC proliferation. The results demonstrate that APP expression stimulates NSPC proliferation and that this effect is mediated via an increase in cystatin C secretion.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Cistatina C/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco/citologia , Precursor de Proteína beta-Amiloide/genética , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/fisiologia , Neurônios/metabolismo
5.
J Neurochem ; 129(5): 756-69, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24517464

RESUMO

The ß-amyloid precursor protein (APP) has been extensively studied for its role as the precursor of the ß-amyloid protein (Aß) of Alzheimer's disease. However, the normal function of APP remains largely unknown. This article reviews studies on the structure, expression and post-translational processing of APP, as well as studies on the effects of APP in vitro and in vivo. We conclude that the published data provide strong evidence that APP has a trophic function. APP is likely to be involved in neural stem cell development, neuronal survival, neurite outgrowth and neurorepair. However, the mechanisms by which APP exerts its actions remain to be elucidated. The available evidence suggests that APP interacts both intracellularly and extracellularly to regulate various signal transduction mechanisms. This article reviews studies on the structure, expression and post-translational processing of ß-amyloid precursor protein (APP), as well as studies on the effects of APP in vitro and in vivo. We conclude that the published data provide strong evidence that APP has a trophic function. APP is likely to be involved in neural stem cell development, neuronal survival, neurite outgrowth and neurorepair. However, the mechanisms by which APP exerts its actions remain to be elucidated. The available evidence suggests that APP interacts both intracellularly and extracellularly to regulate various signal transduction mechanisms.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/farmacologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Humanos , Camundongos , Células-Tronco Neurais/fisiologia , Regiões Promotoras Genéticas/genética
6.
J Neurosci Res ; 92(11): 1478-89, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24916405

RESUMO

The function of the ß-amyloid precursor protein (APP) of Alzheimer's disease is poorly understood. The secreted ectodomain fragment of APP (sAPPα) can be readily cleaved to produce a small N-terminal fragment (N-APP) that contains heparin-binding and metal-binding domains and that has been found to have biological activity. In the present study, we examined whether N-APP can bind to lipids. We found that N-APP binds selectively to phosphoinositides (PIPs) but poorly to most other lipids. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2 )-rich microdomains were identified on the extracellular surface of neurons and glia in primary hippocampal cultures. N-APP bound to neurons and colocalized with PIPs on the cell surface. Furthermore, the binding of N-APP to neurons increased the level of cell-surface PI(4,5)P2 and phosphatidylinositol 3,4,5-trisphosphate. However, PIPs were not the principal cell-surface binding site for N-APP, because N-APP binding to neurons was not inhibited by a short-acyl-chain PIP analogue, and N-APP did not bind to glial cells which also possessed PI(4,5)P2 on the cell surface. The data are explained by a model in which N-APP binds to two distinct components on neurons, one of which is an unidentified receptor and the second of which is a PIP lipid, which binds more weakly to a distinct site within N-APP. Our data provide further support for the idea that N-APP may be an important mediator of APP's biological activity.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/metabolismo , Hipocampo/citologia , Fosfatidilinositóis/metabolismo , Ligação Proteica/fisiologia , Precursor de Proteína beta-Amiloide/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Sítios de Ligação/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/efeitos dos fármacos , Fosfatos de Fosfatidilinositol/metabolismo , Ligação Proteica/efeitos dos fármacos
7.
Neurodegener Dis ; 13(2-3): 96-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23942027

RESUMO

Stem cell therapy may be a suitable approach for the treatment of many neurodegenerative diseases. However, one major impediment to the development of successful cell-based therapies is our limited understanding of the mechanisms that instruct neural stem cell behaviour, such as proliferation and cell fate specification. The ß-amyloid precursor protein (APP) of Alzheimer's disease (AD) may play an important role in neural stem cell proliferation and differentiation. Our recent work shows that in vitro, APP stimulates neural stem or progenitor cell proliferation and neuronal differentiation. The effect on proliferation is mediated by an autocrine factor that we have identified as cystatin C. As cystatin C expression is also reported to inhibit the development of amyloid pathology in APP transgenic mice, our finding has implications for the possible use of cystatin C for the therapy of AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Encéfalo/citologia , Diferenciação Celular , Proliferação de Células , Humanos , Células-Tronco Neurais/citologia
8.
IUBMB Life ; 65(2): 108-20, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23297096

RESUMO

Proteoglycans (PGs) are major components of the cell surface and extracellular matrix and play critical roles in development and maintenance of the central nervous system (CNS). PGs are a family of proteins, all of which contain a core protein to which glycosaminoglycan side chains are covalently attached. PGs possess diverse physiological roles, particularly in neural development, and are also implicated in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD). The main functions of PGs in the CNS are reviewed as are the roles of PGs in brain injury and in the development or treatment of AD.


Assuntos
Doença de Alzheimer/metabolismo , Sistema Nervoso Central/fisiopatologia , Proteoglicanas de Sulfatos de Condroitina/fisiologia , Proteoglicanas de Heparan Sulfato/fisiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Humanos , Plasticidade Neuronal , Sinapses/fisiologia
9.
Subcell Biochem ; 65: 211-24, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23225005

RESUMO

Amyloidoses are characterised by the deposition of insoluble protein that occurs in the extracellular compartment of various tissues. One form of amyloidosis is caused by transthyretin (TTR) misfolding and deposition in target tissues. It is clear that many amyloidoses share common features of fibrillogenesis and toxicity. This chapter examines the mechanisms of TTR aggregation with a view to understanding the possible therapeutic interventions in amyloid disease.


Assuntos
Neuropatias Amiloides Familiares/metabolismo , Amiloide/metabolismo , Pré-Albumina/metabolismo , Dobramento de Proteína , Amiloide/química , Neuropatias Amiloides Familiares/patologia , Animais , Humanos , Pré-Albumina/química
10.
Biochemistry ; 51(6): 1070-8, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22283417

RESUMO

Alzheimer's disease (AD) is a common form of dementia, which is characterized by the presence of extracellular amyloid plaques comprising the amyloid ß peptide (Aß). Although the mechanism underlying AD pathogenesis remains elusive, accumulating evidence suggests that the process of amyloid fibril formation is a surface-mediated event, which plays an important role in AD onset and progression. In this study, the mechanism of Aß aggregation on hydrophobic surfaces was investigated with dual polarization interferometry (DPI), which provides real-time information on early stages of the aggregation process. Aggregation was monitored on a hydrophobic C18 surface and a polar silicon oxynitride surface. The DPI results showed a characteristic Aß aggregation pattern involving a decrease in the density of Aß at the surface followed by an increase in the thickness on the hydrophobic C18 chip. Most importantly, the DPI measurements provided unique information on the early stages of Aß aggregation, which is characterized by the presence of initially slow nucleus formation process followed by exponential fibril elongation. The dimensions of the putative nucleus corresponded to a thickness of ∼5 nm for both Aß40 and Aß42, which may represent about 10-15 molecules. The results thus support the nucleation-dependent polymerization model as indicated by the presence of a nucleation phase followed by an exponential growth phase. These results are the first reported measurements of the real-time changes in Aß molecular structure during the early stages of amyloid formation at the nanometer level.


Assuntos
Peptídeos beta-Amiloides/química , Fragmentos de Peptídeos/química , Doença de Alzheimer/metabolismo , Amiloide/biossíntese , Amiloide/química , Peptídeos beta-Amiloides/metabolismo , Técnicas Biossensoriais , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Fragmentos de Peptídeos/metabolismo , Análise Serial de Proteínas , Ligação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Propriedades de Superfície
11.
J Neurochem ; 122(6): 1155-66, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22712562

RESUMO

Coordinated calcium signalling is vital for neuronal growth cone function and axon pathfinding. Although store-operated calcium entry (SOCE) has been suggested to be an important source of calcium in growth cone navigation, the mechanisms that regulate calcium signalling, particularly the regulation of internal calcium stores within growth cones, are yet to be fully determined. Stromal Interaction Molecule 1 (STIM1) is a calcium-sensing protein localized in the endoplasmic reticulum membrane that interacts with Orai proteins in the plasma membrane to initiate SOCE and refilling of intracellular calcium stores. We hypothesize that STIM1- and Orai1/2-mediated SOCE are necessary for growth cone turning responses to extracellular guidance cues. We show that STIM1 and Orai reorganize into puncta upon store depletion and during growth cone turning with STIM1 localization biased towards the turning side (high calcium side) of the growth cone. Importantly, STIM1 knock-down perturbed growth cone turning responses to the guidance cues brain-derived neurotrophic factor and semaphorin-3a (Sema-3a), as well as abolishing Sema-3a-induced growth cone collapse. Furthermore, STIM1 knock-down abolished SOCE induced by brain-derived neurotrophic factor, but not Sema-3a. Our data suggest that STIM1 is essential for correct growth cone navigation, playing multiple roles in growth cone motility, including the activation of SOCE.


Assuntos
Cálcio/fisiologia , Cones de Crescimento/fisiologia , Glicoproteínas de Membrana/fisiologia , Células Receptoras Sensoriais/fisiologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Células Cultivadas , Feminino , Cones de Crescimento/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Ratos , Ratos Sprague-Dawley , Molécula 1 de Interação Estromal
12.
J Neurochem ; 123(3): 447-57, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22909067

RESUMO

Alzheimer's disease is associated with abnormal accumulation of Aß, which is produced from the ß-amyloid precursor protein (APP) by the ß-site APP-cleaving enzyme (BACE1) and γ-secretase. Our previous studies showed that heparin can decrease APP processing by decreasing the levels of BACE1 and ADAM10. In this study, we examined the effects of glycosaminoglycans (GAGs) on APP processing and Aß production with the aim of understanding the specificity of the effects. Various GAG analogs were incubated with primary cortical cells derived from APP (SW)Tg2576 mice and the level of APP, proteolytic products of APP and APP-cleavage enzymes were measured. The effect of GAGs on APP processing was both size- and sulfation-dependent. 6-O-Sulfation was important for the effect on APP processing as heparin lacking 6-O sulfate were less potent than native heparin. However, deletion of carboxyl groups on heparin had no significant effect on APP processing. Our studies suggest that there is structural specificity to the effect of GAGs on APP processing and that certain GAGs have a greater effect on Aß production than others. This suggests that it might be possible to alter the structure of GAGs to achieve more specific inhibitors of APP processing that can cross the blood-brain barrier.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/metabolismo , Heparina/análogos & derivados , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteoglicanas/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiologia , Desenho de Fármacos , Heparina/metabolismo , Heparina/farmacologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Cultura Primária de Células , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/fisiologia , Proteoglicanas/metabolismo
13.
Clin Exp Pharmacol Physiol ; 39(8): 680-3, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21916933

RESUMO

Familial amyloid polyneuropathies (FAP) constitute a group of inherited amyloidoses that affect peripheral nerves. One common form of FAP is caused by transthyretin (TTR) misfolding and deposition in the peripheral nervous system, leading to neuronal toxicity and death. The molecular mechanisms responsible for this toxicity are unclear; however, there is good biochemical and histopathological evidence that the toxicity of TTR mutations is correlated to their aggregation state. In addition, neuronal calcium dysregulation is a mechanism that has been suggested to drive the pathogenesis of FAP. Amyloidogenic TTR mutations cause significant calcium influx via L-type calcium channels in neuronal cell lines, while in primary sensory neurons, TTR mediates a calcium influx via a novel mechanism of transient receptor potential melanostatin (TRPM8) and voltage-gated sodium and calcium channel activation. Significantly, calcium dysregulation is a pathological hallmark of other neurodegenerative diseases involving amyloidosis, for example Alzheimer's disease, and this mechanism could explain the molecular events that drive amyloid toxicity in other neurodegenerative diseases.


Assuntos
Neuropatias Amiloides Familiares/patologia , Doenças Neurodegenerativas/patologia , Neuropatias Amiloides Familiares/complicações , Neuropatias Amiloides Familiares/genética , Animais , Cálcio/metabolismo , Humanos , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/genética , Pré-Albumina/genética , Pré-Albumina/fisiologia , Deficiências na Proteostase/genética , Deficiências na Proteostase/patologia
14.
Neurodegener Dis ; 10(1-4): 76-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22301681

RESUMO

Amyloid-ß (Aß)-induced Ca(2+) influx into neurons has been well described since it was first reported almost 20 years ago. Ca(2+) influx can disrupt mechanisms of long-term potentiation and long-term depression and increase neuronal susceptibility to excitotoxicity. Our studies show that Aß also causes an increase in acetylcholinesterase (AChE) levels and induces AMPA receptor internalization through Ca(2+)-dependent mechanisms. As Aß-induced Ca(2+) entry may increase neuronal excitability, the increase in AChE and the downregulation of cell surface AMPA receptors may be part of a homeostatic mechanism which maintains normal levels of cholinergic and glutamatergic signaling.


Assuntos
Acetilcolinesterase/metabolismo , Doença de Alzheimer/metabolismo , Cálcio/metabolismo , Homeostase/fisiologia , Neurônios/metabolismo , Receptores de AMPA/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Homeostase/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Sinapses/efeitos dos fármacos
15.
J Neurochem ; 112(6): 1552-61, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20067575

RESUMO

The beta-site APP cleaving enzyme (BACE1) is responsible for the first step in the production of the beta-amyloid protein of Alzheimer's disease. BACE1 is synthesized as a partially active zymogen (proBACE1). We previously showed that the glycosaminoglycan (GAG) heparin can increase the enzyme activity of proBACE1. In this study, the structural requirements and the mechanism for the GAG-induced activation were examined. The effect of heparin on proBACE1 was influenced by the degree of sulfation and carboxylation of the GAG, as well as by the length of the sugar. Although low molecular weight heparin fragments did not strongly stimulate proBACE1, they inhibited heparin-induced activation of the enzyme. The structure of the zymogen was modeled using the known X-ray structures of the BACE1 catalytic domain and the homologous prodomain of porcine pepsinogen. The modeled structure suggested that a heparin-binding domain may reside close to the prodomain, and that movement of a loop region between residues 46-65, lying adjacent to the prodomain, may be needed to accommodate heparin binding. The presence of the loop domain adjacent to the active site may account for the lower activity of the zymogen relative to the mature enzyme. Movement of the loop region upon heparin binding could expose the active site region to allow for increased substrate binding. The results suggest a model in which conformational changes close to the prodomain may be involved in the mechanism of heparin-induced activation of proBACE1.


Assuntos
Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Fibrinolíticos/farmacologia , Heparina/farmacologia , Secretases da Proteína Precursora do Amiloide/química , Animais , Ácido Aspártico Endopeptidases/química , Simulação por Computador , Ativação Enzimática/efeitos da radiação , Precursores Enzimáticos/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Polieletrólitos , Polímeros/farmacologia , Carbonilação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato/efeitos dos fármacos
16.
J Neurochem ; 112(5): 1199-209, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20002523

RESUMO

Aggregation of beta-amyloid protein (Abeta) to form oligomers is considered to be a key step in generating neurotoxicity in the Alzheimer's disease brain. Agents that bind to Abeta and inhibit oligomerization have been proposed as Alzheimer's disease therapeutics. In this study, we investigated the binding of fluorescein-labeled Abeta(1-42) (FluoAbeta(1-42)) to SH-SY5Y neuroblastoma cells and examined the effect of the 39-kDa receptor-associated protein (RAP), on the Abeta cell interaction. FluoAbeta(1-42) bound to the cells in a punctate pattern. Surprisingly, when RAP was added to the incubations, FluoAbeta(1-42) and RAP were found to be co-localized on the cell surface, suggesting that RAP and Abeta may bind to each other. Experiments using the purified proteins confirmed that a RAP-Abeta complex was stable and resistant to sodium dodecyl sulfate. RAP also inhibited Abeta oligomerization. We next examined whether RAP could inhibit the neurotoxic effects of Abeta. Addition of Abeta(1-42) to SH-SY5Y cells caused an increase in intracellular Ca2+ that was inhibited by treatment of the Abeta peptide with RAP. RAP also blocked an Abeta-induced inhibition of long-term memory consolidation in 1-day-old chicks. This study demonstrates that RAP binds to Abeta and is an inhibitor of the neurotoxic effects of Abeta.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/uso terapêutico , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/metabolismo , Peptídeos beta-Amiloides/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Aprendizagem da Esquiva/efeitos dos fármacos , Comportamento Animal , Linhagem Celular Tumoral , Galinhas , Aprendizagem por Discriminação/efeitos dos fármacos , Modelos Animais de Doenças , Citometria de Fluxo/métodos , Fluoresceína/metabolismo , Humanos , Imunoprecipitação/métodos , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/farmacologia , Memória/efeitos dos fármacos , Microscopia de Força Atômica/métodos , Microscopia Confocal/métodos , Peso Molecular , Neuroblastoma , Síndromes Neurotóxicas/fisiopatologia , Proteínas Nucleares/metabolismo , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Ligação Proteica , Transporte Proteico/efeitos dos fármacos
17.
Biol Chem ; 391(8): 849-59, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20731541

RESUMO

Alzheimer's disease (AD) is characterized by the extracellular deposition of the beta-amyloid protein (Abeta). Abeta is a fragment of a much larger precursor protein, the amyloid precursor protein (APP). Sequential proteolytic cleavage of APP by beta-secretase and gamma-secretase liberates Abeta from APP. The aspartyl protease BACE1 (beta-site APP-cleaving enzyme 1) catalyses the rate-limiting step in the production of Abeta, and as such it is considered to be a major target for drug development in Alzheimer's disease. However, the development of a BACE1 inhibitor therapy is problematic for two reasons. First, BACE1 has been found to have important physiological roles. Therefore, inhibition of the enzyme could have toxic consequences. Second, the active site of BACE1 is relatively large, and many of the bulky compounds that are needed to inhibit BACE1 activity are unlikely to cross the blood-brain barrier. This review focuses on the structure BACE1, current therapeutic strategies based on developing active-site inhibitors, and new approaches to therapy involving targeting the expression or post-translational regulation of BACE1.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/fisiologia , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/fisiologia , Desenho de Fármacos , Inibidores de Proteases , Secretases da Proteína Precursora do Amiloide/química , Animais , Ácido Aspártico Endopeptidases/química , Domínio Catalítico , Regulação Enzimológica da Expressão Gênica , Humanos , Inibidores de Proteases/efeitos adversos , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Especificidade por Substrato
18.
Neurodegener Dis ; 7(1-3): 187-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20224283

RESUMO

BACKGROUND: The beta-site APP cleaving enzyme 1 (BACE1) is a major target for drug design in Alzheimer's disease. BACE1 binds strongly to heparin and other glycosaminoglycans, and there is evidence that the enzyme may interact with proteoglycans in vivo. Several studies suggest that heparin or heparan sulfate analogues may have value as therapeutic agents for the treatment of AD. OBJECTIVE: To determine whether heparin can inhibit Abeta production in cortical neurons by inhibiting BACE1. METHODS: Cortical neurons from APP (SW) Tg2576 mice were incubated with heparin and the amount of APP processing and Abeta production were measured by enzyme-linked immunosorbent assay and Western blotting. RESULTS: Treatment of cortical neurons with heparin inhibited Abeta secretion. However, this effect was not mediated via inhibition of BACE1. CONCLUSIONS: Heparin or other glycosaminoglycans may have value for the treatment of Alzheimer's disease. However, the data do not support the view that a heparin-induced decrease in Abeta secretion is due to inhibition of BACE1.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Córtex Cerebral/citologia , Fibrinolíticos/farmacologia , Heparina/farmacologia , Neurônios/efeitos dos fármacos , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Ácido Aspártico Endopeptidases/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo
19.
Brain Behav ; 10(7): e01672, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32484608

RESUMO

INTRODUCTION: One of the major neuropathological features of Alzheimer's disease (AD) is the accumulation of amyloid-ß (Aß) protein in the brain. Evidence suggests that the low-density lipoprotein receptor-associated protein (RAP) binds strongly to Aß and enhances its cellular uptake and that decreased RAP expression correlates with increased Aß production in animal models of AD. METHODS: The current study examined whether RAP levels change in AD human brain tissue and whether they are related to the amount of AD pathology. RAP and NeuN levels were determined by Western blot, while low-density lipoprotein receptor-related protein 1 (LRP1), tau and Aß levels were determined by ELISA in the temporal cortex of 17 AD and 16 control cases. RESULTS: An increase in total Aß and insoluble and soluble tau protein was observed in AD brain tissue. In contrast, RAP levels were significantly decreased in AD brain tissue compared to controls. Correlation analysis revealed that levels of RAP correlated with both total Aß and soluble and insoluble tau levels. Neither LRP1 nor NeuN levels were significantly altered in AD brain tissue homogenates and did not correlate with Aß or tau protein levels. CONCLUSION: Reduction in RAP may contribute to the accumulation and aggregation of Aß in the AD brain.


Assuntos
Doença de Alzheimer , Encéfalo/metabolismo , Proteínas tau , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Proteína Associada a Proteínas Relacionadas a Receptor de LDL , Proteínas tau/metabolismo
20.
Biochim Biophys Acta ; 1778(1): 198-205, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18061140

RESUMO

Deposition of transthyretin (TTR) amyloid is a pathological hallmark of familial amyloidotic polyneuropathy (FAP). Recently we showed that TTR binds to membrane lipids via electrostatic interactions and that membrane binding is correlated with the cytotoxicity induced by amyloidogenic TTR. In the present study, we examined the role of lipid composition in membrane binding of TTR by a surface plasmon resonance (SPR) approach. TTR bound to lipid bilayers through both high- and low-affinity interactions. Increasing the mole fraction of cholesterol in the bilayer led to an increase in the amount of high-affinity binding of an amyloidogenic mutant (L55P) TTR. In addition, a greater amount of L55P TTR bound with high affinity to membranes made from anionic phospholipids, phosphatidylglycerol (PG) and phosphatidylserine (PS), than to membranes made from zwitterionic phospholipid phosphatidylcholine (PC). The anionic phospholipids (PS and PG) promoted the aggregation of L55P TTR by accelerating the nucleation phase of aggregation, whereas the zwitterionic phospholipid PC had little effect. These results suggest that cholesterol and anionic phospholipids may be important for TTR aggregation and TTR-induced cytotoxicity.


Assuntos
Amiloide/metabolismo , Colesterol/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Pré-Albumina/metabolismo , Nefelometria e Turbidimetria , Pré-Albumina/química , Ligação Proteica , Estrutura Quaternária de Proteína , Soluções
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA