Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 75(2): 301-322, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28799085

RESUMO

Proteolytic cleavage of the amyloid precursor protein (APP) by α-, ß- and γ-secretases is a determining factor in Alzheimer's disease (AD). Imbalances in the activity of all three enzymes can result in alterations towards pathogenic Aß production. Proteolysis of APP is strongly linked to its subcellular localization as the secretases involved are distributed in different cellular compartments. APP has been shown to dimerize in cis-orientation, affecting Aß production. This might be explained by different substrate properties defined by the APP oligomerization state or alternatively by altered APP monomer/dimer localization. We investigated the latter hypothesis using two different APP dimerization systems in HeLa cells. Dimerization caused a decreased localization of APP to the Golgi and at the plasma membrane, whereas the levels in the ER and in endosomes were increased. Furthermore, we observed via live cell imaging and biochemical analyses that APP dimerization affects its interaction with LRP1 and SorLA, suggesting that APP dimerization modulates its interplay with sorting molecules and in turn its localization and processing. Thus, pharmacological approaches targeting APP oligomerization properties might open novel strategies for treatment of AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Endossomos/metabolismo , Feminino , Complexo de Golgi/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Relacionadas a Receptor de LDL/genética , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Ligação Proteica , Multimerização Proteica , Transporte Proteico
2.
Cell Death Differ ; 19(9): 1413-23, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22361682

RESUMO

Neurodegeneration associated with amyloid ß (Aß) peptide accumulation, synaptic loss, neuroinflammation, tauopathy, and memory impairments encompass the pathophysiological features of Alzheimer's disease (AD). We previously reported that the scaffolding protein RanBP9, which is overall increased in brains of AD patients, simultaneously promotes Aß generation and focal adhesion disruption by accelerating the endocytosis of amyloid precursor protein (APP) and ß1-integrin, respectively. Here, we show that RanBP9 protein levels are increased by fourfold in FAD mutant APP transgenic mice. Accordingly, RanBP9 transgenic mice demonstrate significantly increased synapse loss, neurodegeneration, gliosis, and spatial memory deficits. RanBP9 overexpression promotes apoptosis and potentiates Aß-induced neurotoxicity independent of its capacity to promote Aß generation. Conversely, RanBP9 reduction by siRNA or gene dosage mitigates Aß-induced neurotoxicity. Importantly, RanBP9 activates/dephosphorylates cofilin, a key regulator of actin dynamics and mitochondria-mediated apoptosis, and siRNA knockdown of cofilin abolishes both Aß and RanBP9-induced apoptosis. These findings implicate the RanBP9-cofilin pathway as critical therapeutic targets not only for stemming Aß generation but also antagonizing Aß-induced neurotoxicity.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Apoptose , Encéfalo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Fatores de Despolimerização de Actina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Encéfalo/patologia , Proteínas do Citoesqueleto/genética , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Nucleares/genética , Fosforilação/genética
3.
J Int Med Res ; 34(2): 140-51, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16749409

RESUMO

Both ischaemic preconditioning (IPC) and amiodarone protect against myocardial ischaemia. We examined whether a combination of IPC and amiodarone demonstrated an additive protective effect in isolated rat hearts (n = 40). The controls (group I) were subjected to ischaemia/ reperfusion injury; group II was subjected to cycles of IPC prior to ischaemia/ reperfusion injury; group III was subjected to ischaemia in the presence of amiodarone (10(-10) mol/1); and group IV was subjected to IPC followed by ischaemia in the presence of amiodarone (10(-10) mol/l). Amiodarone produced the best preserved left ventricular end-systolic pressure and dP/dtmax, less developed ventricular stiffness, the shortest arrhythmia duration, and the smallest infarct size among the groups. All of the myocardial protective effects against ischaemia/reperfusion injury were diminished or abolished when IPC and amiodarone were applied sequentially.


Assuntos
Amiodarona/farmacologia , Antiarrítmicos/farmacologia , Precondicionamento Isquêmico Miocárdico , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Pressão Sanguínea/efeitos dos fármacos , Cardiotônicos/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Precondicionamento Isquêmico Miocárdico/métodos , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ratos , Ratos Sprague-Dawley
4.
FASEB J ; 20(8): 1254-6, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16636103

RESUMO

Amyloid beta-peptide (Abeta) is postulated to play a central role in the pathogenesis of Alzheimer's disease. We recently proposed a pathway of Abeta-induced toxicity that is APP dependent and involves the facilitation of APP complex formation by Abeta. The APP-dependent component requires cleavage of APP at position 664 in the cytoplasmic domain, presumably by caspases or caspase-like proteases, with release of a potentially cytotoxic C31 peptide. In this study we show that Abeta interacted directly and specifically with membrane-bound APP to facilitate APP homo-oligomerization. Using chimeric APP molecules, this interaction was shown to take place between Abeta and its homologous sequence on APP. Consistent with this finding, we demonstrated that Abeta also facilitated the oligomerization of beta-secretase cleaved APP C-terminal fragment (C99). We found that the YENPTY domain in the APP cytoplasmic tail and contained within C31 is critical for this cell death pathway. Deletion or alanine- scanning mutagenesis through this domain significantly attenuated cell death apparently without affecting either APP dimerization or cleavage at position 664. This indicated that sequences within C31 are required after its release from APP. As the YENPTY domain has been shown to interact with a number of cytosolic adaptor molecules, it is possible that the interaction of APP, especially dimeric forms of APP, with these molecules contribute to cell death.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Precursor de Proteína beta-Amiloide/química , Fragmentos de Peptídeos/toxicidade , Motivos de Aminoácidos , Sequência de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Morte Celular , Linhagem Celular Tumoral , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Ratos
5.
Nature ; 414(6860): 212-6, 2001 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-11700559

RESUMO

Epidemiological studies have documented a reduced prevalence of Alzheimer's disease among users of nonsteroidal anti-inflammatory drugs (NSAIDs). It has been proposed that NSAIDs exert their beneficial effects in part by reducing neurotoxic inflammatory responses in the brain, although this mechanism has not been proved. Here we report that the NSAIDs ibuprofen, indomethacin and sulindac sulphide preferentially decrease the highly amyloidogenic Abeta42 peptide (the 42-residue isoform of the amyloid-beta peptide) produced from a variety of cultured cells by as much as 80%. This effect was not seen in all NSAIDs and seems not to be mediated by inhibition of cyclooxygenase (COX) activity, the principal pharmacological target of NSAIDs. Furthermore, short-term administration of ibuprofen to mice that produce mutant beta-amyloid precursor protein (APP) lowered their brain levels of Abeta42. In cultured cells, the decrease in Abeta42 secretion was accompanied by an increase in the Abeta(1-38) isoform, indicating that NSAIDs subtly alter gamma-secretase activity without significantly perturbing other APP processing pathways or Notch cleavage. Our findings suggest that NSAIDs directly affect amyloid pathology in the brain by reducing Abeta42 peptide levels independently of COX activity and that this Abeta42-lowering activity could be optimized to selectively target the pathogenic Abeta42 species.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Fragmentos de Peptídeos/metabolismo , Sulindaco/análogos & derivados , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Doença de Alzheimer/etiologia , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Ácido Aspártico Endopeptidases , Encéfalo/metabolismo , Células CHO , Cricetinae , Modelos Animais de Doenças , Endopeptidases/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Ibuprofeno/farmacologia , Indometacina/farmacologia , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Prostaglandina-Endoperóxido Sintases/metabolismo , Sulindaco/farmacologia , Células Tumorais Cultivadas
6.
Arch Ophthalmol ; 119(11): 1629-34, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11709013

RESUMO

OBJECTIVES: To use microarray analysis to identify genes expressed in human donor corneas and to create a preliminary, comprehensive database of human corneal gene expression. METHODS: A complementary DNA (cDNA) library was constructed from transplant-quality, human donor corneas. Biotin-labeled RNA was transcribed from the cDNA library and hybridized in duplicate to microarrays containing approximately 5600 human genes. Results were analyzed using a gene database of the National Institutes of Health, Bethesda, Md. Reverse transcriptase polymerase chain reaction analysis was performed to confirm corneal expression of genes identified by microarray analysis. RESULTS: Duplicate microarrays identified the expression of 1200 genes in human donor corneas. Chromosomal loci had been assigned to 1025 (85%) of these genes. A preliminary database of human corneal gene expression was compiled. A Web site containing these genes was created. Six collagen genes were identified that had not previously been localized within the cornea. Five apoptosis-related genes were identified, 4 of which had not previously been localized within the cornea. Three genes previously shown to cause corneal diseases were identified. Reverse transcriptase polymerase chain reaction analysis of genes identified by microarray analysis confirmed the corneal expression of 2 apoptosis-related genes and 1 collagen gene. CONCLUSIONS: Microarray analysis of healthy human donor corneas has produced a preliminary, comprehensive database of corneal gene expression. Large-scale analysis of gene expression has the potential to generate large amounts of data, which should be made readily accessible to the scientific community. The Internet offers many potential advantages as a medium for the maintenance of these large data sets. CLINICAL RELEVANCE: Identification of structural, apoptosis-related, and disease-causing genes within the cornea by microarrays may increase the understanding of normal and abnormal corneal function with likely relevance to corneal diseases and transplants.


Assuntos
Córnea/metabolismo , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Doadores de Tecidos , Cromossomos Humanos/genética , DNA Complementar/metabolismo , Proteínas do Olho/metabolismo , Humanos , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Proc Natl Acad Sci U S A ; 98(19): 10863-8, 2001 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-11517342

RESUMO

Presenilin 1 (PS1) is required for the proteolytic processing of Notch and the beta-amyloid precursor protein (APP), molecules that play pivotal roles in cell-fate determination during development and Alzheimer's disease pathogenesis, respectively. In addition, PS1 interacts with beta-catenin and promotes its turnover through independent mechanisms. Consistent with this activity, we report here that PS1 is important in controlling epidermal cell proliferation in vivo. PS1 knockout mice that are rescued through neuronal expression of human PS1 transgene develop spontaneous skin cancers. PS1-null keratinocytes exhibit higher cytosolic beta-catenin and beta-catenin/lymphoid enhancer factor-1/T cell factor (beta-catenin/LEF)-mediated signaling. This effect can be reversed by reintroducing wild-type PS1, but not a PS1 mutant active in Notch processing but defective in beta-catenin binding. Nuclear beta-catenin protein can be detected in tumors. Elevated beta-catenin/LEF signaling is correlated with activation of its downstream target cyclin D1 and accelerated entry from G(1) into S phase of the cell cycle. This report demonstrates a function of PS1 in adult tissues, and our analysis suggests that deregulation of beta-catenin pathway contributes to the skin tumor phenotype.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/fisiologia , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Transativadores , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Ciclina D1/metabolismo , Citosol/metabolismo , Fase G1 , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Presenilina-1 , Fase S , Neoplasias Cutâneas/patologia , beta Catenina
8.
Biochem Biophys Res Commun ; 285(4): 997-1006, 2001 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-11467851

RESUMO

Deposition of plaques containing Abeta is considered important in the pathogenesis of Alzheimer's disease. Phorbol esters that activate protein kinase C (PKC) promote alpha-secretase-mediated processing of the beta amyloid precursor protein (APP), which generally reduces formation of Abeta. To determine which PKC isozymes mediate this process, we studied CHO cells that express human APP751. Phorbol 12-myristate, 13-acetate (PMA)-stimulated APP secretion, which was reduced by a general PKC inhibitor bisindoylmaleimide I, but not by Gö 6976, which inhibits PKCalpha, beta, gamma, and mu. Since PKCdelta and epsilon were the only other PMA-sensitive isozymes present, we studied cells that express selective peptide inhibitors of these isozymes. Expression of the PKCepsilon inhibitor inhibited PMA-induced APPs secretion and suppression of Abeta production. In contrast, the PKCdelta inhibitor had no effect. These results provide evidence that PKCepsilon decreases Abeta production by promoting alpha-secretase mediated cleavage of APP.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Endopeptidases/metabolismo , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Doença de Alzheimer/etiologia , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases , Células CHO , Carbazóis/farmacologia , Cricetinae , Regulação para Baixo , Ativação Enzimática , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-épsilon , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
9.
J Biol Chem ; 276(31): 29045-50, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11397796

RESUMO

Amyloid beta-protein (A beta) is the main constituent of senile plaques in Alzheimer's disease and is derived by proteolysis from the amyloid precursor protein (APP). Generation and secretion of both A beta 40 and A beta 42 isoforms depend largely on internalization of APP and occurs mainly in the endocytic pathway. Evidence has also been presented (Gervais, F. G., Xu, D., Robertson, G. S., Vaillancourt, J. P., Zhu, Y., Huang, J., LeBlanc, A., Smith, D., Rigby, M., Shearman, M. S., Clarke, E. E., Zheng, H., Van der Ploeg, L. H. T., Ruffolo, S. C., Thornberry, N. A., Xanthoudakis, S., Zamboni, R. J., Roy, S., and Nicholson, D. W. (1999) Cell, 97, 395--406) that caspase cleavage of APP at its cytosolic tail affects its processing such that it is redirected to a more amyloidogenic pathway, resulting in enhanced A beta generation. However, caspase cleavage of APP also results in loss of its endocytosis signal (YENP), an event that would predict a decline in internalization and a concomitant decrease, not an increase, in A beta generation. In the present work, we examined whether caspase cleavage of APP is relevant to amyloidogenesis. We found that 1) caspase cleavage of APP results in reduced internalization and, accordingly, a decline in A beta secretion; 2) masking of the caspase site in APP did not affect A beta levels and, 3) caspase activation in cells by serum withdrawal did not increase A beta secretion. Thus, caspase cleavage of APP is unlikely to play a direct role in amyloidogenesis.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Caspases/metabolismo , Doença de Alzheimer/patologia , Animais , Células CHO , Linhagem Celular , Cricetinae , Meios de Cultura Livres de Soro , Endocitose , Ativação Enzimática , Humanos , Neurônios , Isoformas de Proteínas/metabolismo , Ratos , Especificidade por Substrato
10.
J Cell Biol ; 152(4): 785-94, 2001 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11266469

RESUMO

In addition to its documented role in the proteolytic processing of Notch-1 and the beta-amyloid precursor protein, presenilin 1 (PS1) associates with beta-catenin. In this study, we show that this interaction plays a critical role in regulating beta-catenin/T Cell Factor/Lymphoid Enhancer Factor-1 (LEF) signaling. PS1 deficiency results in accumulation of cytosolic beta-catenin, leading to a beta-catenin/LEF-dependent increase in cyclin D1 transcription and accelerated entry into the S phase of the cell cycle. Conversely, PS1 specifically represses LEF-dependent transcription in a dose-dependent manner. The hyperproliferative response can be reversed by reintroducing PS1 expression or overexpressing axin, but not a PS1 mutant that does not bind beta-catenin (PS1Deltacat) or by two different familial Alzheimer's disease mutants. In contrast, PS1Deltacat restores Notch-1 proteolytic cleavage and Abeta generation in PS1-deficient cells, indicating that PS1 function in modulating beta-catenin levels can be separated from its roles in facilitating gamma-secretase cleavage of beta-amyloid precursor protein and in Notch-1 signaling. Finally, we show an altered response to Wnt signaling and impaired ubiquitination of beta-catenin in the absence of PS1, a phenotype that may account for the increased stability in PS1-deficient cells. Thus, PS1 adds to the molecules that are known to regulate the rapid turnover of beta-catenin.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Superfície Celular , Proteínas Repressoras , Transativadores , Fatores de Transcrição/metabolismo , Doença de Alzheimer , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Proteína Axina , Ciclina D1 , Citosol , Endopeptidases/metabolismo , Fibroblastos/citologia , Regulação da Expressão Gênica , Fator 1 de Ligação ao Facilitador Linfoide , Proteínas de Membrana/genética , Camundongos , Mutação , Presenilina-1 , Processamento de Proteína Pós-Traducional , Proteínas/metabolismo , Receptor Notch1 , Fase S , Transdução de Sinais , Ubiquitinas/metabolismo , Proteínas Wnt , Proteína Wnt3 , beta Catenina
11.
J Clin Invest ; 106(9): 1159-66, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11067868

RESUMO

Susceptibility to Alzheimer's disease (AD) is governed by multiple genetic factors. Remarkably, the LDL receptor-related protein (LRP) and its ligands, apoE and alpha2M, are all genetically associated with AD. In this study, we provide evidence for the involvement of the LRP pathway in amyloid deposition through sequestration and removal of soluble amyloid beta-protein (Abeta). We demonstrate in vitro that LRP mediates the clearance of both Abeta40 and Abeta42 through a bona fide receptor-mediated uptake mechanism. In vivo, reduced LRP expression is associated with LRP genotypes and is correlated with enhanced soluble Abeta levels and amyloid deposition. Although LRP has been proposed to be a clearance pathway for Abeta, this work provides the first in vivo evidence that the LRP pathway may modulate Abeta deposition and AD susceptibility by regulating the removal of soluble Abeta.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Receptores Imunológicos/metabolismo , Receptores de LDL/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Animais , Transporte Biológico Ativo , Estudos de Casos e Controles , Linhagem Celular , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Fragmentos de Peptídeos/metabolismo , Receptores Imunológicos/genética , Receptores de LDL/genética , Solubilidade
12.
Microsc Res Tech ; 50(4): 268-72, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10936878

RESUMO

To date, mutations in three genes, beta-amyloid precursor protein (APP), presenilin 1 (PS1), and presenilin 2 (PS2), have been found to be causally related to familial Alzheimer's disease (AD). In addition, polymorphisms in three other genes (among others), apolipoprotein E (apoE), alpha2-macroglobulin (alpham), and the low density lipoprotein receptor-related protein (LRP), are implicated to contribute to AD pathogenesis. Interestingly, the encoded gene products are all functionally related in various ways to LRP. Specifically apoE, alpha2m, secreted APP, and amyloid beta-protein (Abeta) complexed to either apoE or alpha2m are ligands of LRP. Furthermore, over-expression of presenilin 1 results in decreased expression of LRP. Since levels of many LRP ligands are increased in Alzheimer's disease and LRP and its ligands are present in senile plaques, decreased LRP function may be a central component in AD pathogenesis. This review explores the current knowledge of LRP in AD and its relationship to the other known AD susceptibility markers.


Assuntos
Doença de Alzheimer/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Complexo Antigênico da Nefrite de Heymann , Humanos , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Presenilina-1 , Presenilina-2 , Receptores de LDL/genética
13.
J Biol Chem ; 275(22): 17136-42, 2000 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-10748144

RESUMO

Presenilin 1 (PS1) and presenilin 2 (PS2) are polytopic membrane proteins that are mutated in the majority of early onset familial Alzheimer's disease (FAD) cases. Two lines of evidence establish a critical role for PS in the production of beta-amyloid peptides (Abeta). FAD-linked PS mutations elevate the levels of highly amyloidogenic Abeta ending at residue 42 (Abeta42), and cells with ablated PS1 alleles secrete low levels of Abeta. Several recent reports have shown that the hydrophilic loop (HL) domain, located between transmembrane domains 6 and 7, contains sites for phosphorylation, caspase cleavage, and sequences that bind several PS-interacting proteins. In the present report, we examined the metabolism of PS polypeptides lacking the HL domain and the influence of these molecules on Abeta production. We report that the deletion of the HL domain does not have a deleterious effect on the regulated endoproteolysis of PS, saturable accumulation of PS fragments, or the self-association of PS fragments. Abeta production was not significantly altered in cells expressing HL-deleted PS polypeptides compared with cells expressing full-length PS. Importantly, deletion of the HL domain did not affect FAD mutation-mediated elevation in the production of Abeta42. Furthermore, the deletion of the HL domain did not impair the role of PS1 or PS2 in facilitating Notch processing. Thus, our results argue against a biologically or pathologically relevant role for the HL domain phosphorylation and caspase cleavage and the association of PS HL domain-interacting proteins, in amyloid precursor protein metabolism and Abeta production or Notch cleavage.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/biossíntese , Idade de Início , Animais , Sequência de Bases , Células COS , Primers do DNA , Humanos , Hidrólise , Proteínas de Membrana/química , Camundongos , Presenilina-1 , Presenilina-2 , Conformação Proteica , Processamento de Proteína Pós-Traducional , Receptores Notch
14.
Nat Med ; 6(4): 397-404, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10742146

RESUMO

The amyloid beta-protein precursor gives rise to the amyloid beta-protein, the principal constituent of senile plaques and a cytotoxic fragment involved in the pathogenesis of Alzheimer disease. Here we show that amyloid beta-protein precursor was proteolytically cleaved by caspases in the C terminus to generate a second unrelated peptide, called C31. The resultant C31 peptide was a potent inducer of apoptosis. Both caspase-cleaved amyloid beta-protein precursor and activated caspase-9 were present in brains of Alzheimer disease patients but not in control brains. These findings indicate the possibility that caspase cleavage of amyloid beta-protein precursor with the generation of C31 may be involved in the neuronal death associated with Alzheimer disease.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Caspases/metabolismo , Peptídeos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose , Encéfalo/enzimologia , Encéfalo/patologia , Caspase 9 , Caspases/genética , Linhagem Celular Transformada , Ativação Enzimática , Humanos , Camundongos , Fragmentos de Peptídeos/metabolismo , Peptídeos/fisiologia , Especificidade por Substrato
15.
Biochem Biophys Res Commun ; 268(3): 750-6, 2000 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-10679277

RESUMO

Amyloid beta peptides are bound rapidly in the plasma complicating an accurate assessment of their in vivo abundance by immunoassay procedures. The extent of Abeta immunoassay interference was used to estimate the Abeta binding capacity of purified plasma proteins, erythrocytes and whole plasma. Human serum albumin bound Abeta peptides rapidly with a 1:1 stoichiometry and at physiological concentrations was capable of binding over 95% of an input of 5 ng/ml Abeta. Purified alpha2-macroglobulin was able to bind Abeta peptides and at physiological concentration bound 73% of 5 ng/ml of Abeta. Erythrocytes also sequestered the Abeta peptides, showing a preference for binding Abeta 1-42. Incubation of 5 ng/ml of Abeta in plasma revealed that about 30% of the peptides were still detectable by immunoassay, presumably reflecting the binding of Abeta peptides with albumin and other plasma molecules. Thus, our studies reveal that both the soluble and formed elements of the blood are capable of sequestering Abeta peptides. To avoid underestimating plasma Abeta values, we employed an improved column chromatography method under denaturing conditions to liberate Abeta from its associations with plasma proteins. Quantification of Abeta 40 and 42 levels in plasma from both normal and AD individuals after chromatography showed a large overlap between AD and control groups, despite the very large pool of Abeta present in the AD brains. The potential origins of the plasma Abeta pool are discussed.


Assuntos
Doença de Alzheimer/sangue , Peptídeos beta-Amiloides/sangue , Proteínas Sanguíneas/metabolismo , Eritrócitos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Análise Química do Sangue , Estudos de Casos e Controles , Feminino , Humanos , Imunoensaio , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/sangue , Ligação Proteica , Albumina Sérica/metabolismo , alfa-Macroglobulinas/metabolismo
16.
J Biol Chem ; 274(45): 32295-300, 1999 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-10542269

RESUMO

Amyloid beta-protein (Abeta) is the main constituent of amyloid fibrils found in senile plaques and cerebral vessels in Alzheimer's disease (AD) and is derived by proteolysis from the beta-amyloid precursor protein (APP). We have analyzed the amyloidogenic processing of APP using chimeric proteins stably transfected in Chinese hamster ovary cells. The extracellular and transmembrane domains of APP were fused to the cytoplasmic region derived from the CD3 gamma chain of the T cell antigen receptor (CD3gamma). CD3gamma contains an endoplasmic reticulum (ER) retention motif (RKK), in the absence of which the protein is targeted to lysosomes without going through the cell surface (Letourneur, F., and Klausner, R.D. (1992) Cell 69, 1143-1157). We used the wild-type sequence of CD3gamma to create an APP chimera predicted to remain in the ER (gammaAPP(ER)). Deletion of the RKK motif at the C terminus directed the protein directly to the lysosomes (gammaAPP(LYS)). A third chimera was created by removing both lysosomal targeting signals in addition to RKK (gammaAPP(DeltaDelta)). This last construct does not contain known targeting signals and consequently accumulates at the cell surface. We show by immunofluorescence and by biochemical methods that all three APP chimeras localize to the predicted compartments within the cell, thus providing a useful model to study the processing of APP. We found that Abeta(1-40) is generated in the early secretory and endocytic pathways, whereas Abeta(1-42) is made mainly in the secretory pathway. More importantly, we provide evidence that, unlike in neuronal models, both ER/intermediate compartment- and endocytic-derived Abeta forms can enter the secretable pool. Finally, we directly demonstrate that lysosomal processing is not involved in the generation or secretion of either Abeta(1-40) or Abeta(1-42).


Assuntos
Peptídeos beta-Amiloides/biossíntese , Complexo CD3/biossíntese , Endocitose , Fragmentos de Peptídeos/biossíntese , Sequência de Aminoácidos , Peptídeos beta-Amiloides/genética , Animais , Complexo CD3/genética , Células CHO , Cricetinae , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/biossíntese , Transfecção
18.
J Biol Chem ; 274(27): 18851-6, 1999 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-10383380

RESUMO

It has long been assumed that the C-terminal motif, NPXY, is the internalization signal for beta-amyloid precursor protein (APP) and that the NPXY tyrosine (Tyr743 by APP751 numbering, Tyr682 in APP695) is required for APP endocytosis. To evaluate this tenet and to identify the specific amino acids subserving APP endocytosis, we mutated all tyrosines in the APP cytoplasmic domain and amino acids within the sequence GYENPTY (amino acids 737-743). Stable cell lines expressing these mutations were assessed for APP endocytosis, secretion, and turnover. Normal APP endocytosis was observed for cells expressing Y709A, G737A, and Y743A mutations. However, Y738A, N740A, and P741A or the double mutation of Y738A/P741A significantly impaired APP internalization to a level similar to that observed for cells lacking nearly the entire APP cytoplasmic domain (DeltaC), arguing that the dominant signal for APP endocytosis is the tetrapeptide YENP. Although not an APP internalization signal, Tyr743 regulates rapid APP turnover because half-life increased by 50% with the Y743A mutation alone. Secretion of the APP-derived proteolytic fragment, Abeta, was tightly correlated with APP internalization, such that Abeta secretion was unchanged for cells having normal APP endocytosis but significantly decreased for endocytosis-deficient cell lines. Remarkably, secretion of the Abeta42 isoform was also reduced in parallel with endocytosis from internalization-deficient cell lines, suggesting an important role for APP endocytosis in the secretion of this highly pathogenic Abeta species.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Endocitose , Fragmentos de Peptídeos/metabolismo , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células CHO , Cricetinae , Ensaio de Imunoadsorção Enzimática , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Relação Estrutura-Atividade
19.
J Neurosci Methods ; 88(1): 45-54, 1999 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10379578

RESUMO

Endocytosis of full-length beta-amyloid precursor protein (APP) from the plasma membrane contributes to beta-amyloid peptide (Abeta) secretion, and, hence, potentially contributes to the molecular pathogenesis of Alzheimer's disease. We recently have demonstrated that central neuronal APP is endocytosed in a common vesicular compartment with recycling synaptic vesicle integral membrane proteins, but is then sorted away from synaptic vesicles for retrograde transport to the neuronal soma. For this report, we explore whether recombinant adenovirus can be used to modulate APP expression in cultured central neurons to study APP processing by the endocytotic pathway in these cells. Using a replication-deficient recombinant adenovirus that expresses a lacZ reporter (Ad5/CMV-lacZ), we demonstrate high efficiency of transfection (30-35%) at low viral titer (10-20 MOI), with no significant neuronal toxicity or cytoarchitectural change. In addition, we demonstrate that infection with the control virus does not result in re-direction of endogenous neuronal APP from usual endocytotic pathways. We have prepared, using the same genomic background as the control virus, an adenoviral vector that expresses the neuronal isoform of human APP (Ad5/CMV-APP). Infection with Ad5/CMV-APP at 10-20 MOI results in significantly increased immunoreactivity for endocytosed APP with preservation of usual endocytotic trafficking. These results demonstrate that recombinant adenovirus at low titer is an appropriate and effective vector for protein trafficking/processing studies in cultured central neurons.


Assuntos
Adenoviridae , Precursor de Proteína beta-Amiloide/genética , Endocitose/genética , Vetores Genéticos , Neurônios/citologia , Infecções por Adenoviridae , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/imunologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Anticorpos Monoclonais , Células Cultivadas , Cerebelo/citologia , Genes Reporter , Humanos , Óperon Lac , Microscopia Eletrônica , Neurônios/metabolismo , Neurônios/ultraestrutura , Ratos , Proteínas Recombinantes de Fusão/genética , Transfecção
20.
J Neurosci ; 19(11): 4229-37, 1999 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10341227

RESUMO

Although an association between the product of the familial Alzheimer's disease (FAD) gene, presenilin 1 (PS1), and beta-catenin has been reported recently, the cellular consequences of this interaction are unknown. Here, we show that both the full length and the C-terminal fragment of wild-type or FAD mutant PS1 interact with beta-catenin from transfected cells and brains of transgenic mice, whereas E-cadherin and adenomatous polyposis coli (APC) are not detected in this complex. Inducible overexpression of PS1 led to increased association of beta-catenin with glycogen synthase kinase-3beta (GSK-3beta), a negative regulator of beta-catenin, and accelerated the turnover of endogenous beta-catenin. In support of this finding, the beta-catenin half-life was dramatically longer in fibroblasts deficient in PS1, and this phenotype was completely rescued by replacement of PS1, demonstrating that PS1 normally stimulates the degradation of beta-catenin. In contrast, overexpression of FAD-linked PS1 mutants (M146L and DeltaX9) failed to enhance the association between GSK-3beta and beta-catenin and interfered with the constitutive turnover of beta-catenin. In vivo confirmation was demonstrated in the brains of transgenic mice in which the expression of the M146L mutant PS1 was correlated with increased steady-state levels of endogenous beta-catenin. Thus, our results indicate that PS1 normally promotes the turnover of beta-catenin, whereas PS1 mutants partially interfere with this process, possibly by failing to recruit GSK-3beta into the PS1-beta-catenin complex. These findings raise the intriguing possibility that PS1-beta-catenin interactions and subsequent activities may be consequential for the pathogenesis of AD.


Assuntos
Doença de Alzheimer/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/genética , Transdução de Sinais/fisiologia , Transativadores , Polipose Adenomatosa do Colo/metabolismo , Animais , Encéfalo/metabolismo , Caderinas/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Quinase 3 da Glicogênio Sintase , Quinases da Glicogênio Sintase , Camundongos , Camundongos Transgênicos , Mutação , Presenilina-1 , beta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...