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










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 114(45): E9665-E9674, 2017 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-29078331

RESUMO

Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by pathological brain lesions and a decline in cognitive function. ß-Amyloid peptides (Aß), derived from proteolytic processing of amyloid precursor protein (APP), play a central role in AD pathogenesis. ß-Site APP cleaving enzyme 1 (BACE1), the transmembrane aspartyl protease which initiates Aß production, is axonally transported in neurons and accumulates in dystrophic neurites near cerebral amyloid deposits in AD. BACE1 is modified by S-palmitoylation at four juxtamembrane cysteine residues. S-palmitoylation is a dynamic posttranslational modification that is important for trafficking and function of several synaptic proteins. Here, we investigated the in vivo significance of BACE1 S-palmitoylation through the analysis of knock-in mice with cysteine-to-alanine substitution at the palmitoylated residues (4CA mice). BACE1 expression, as well as processing of APP and other neuronal substrates, was unaltered in 4CA mice despite the lack of BACE1 S-palmitoylation and reduced lipid raft association. Whereas steady-state Aß levels were similar, synaptic activity-induced endogenous Aß production was not observed in 4CA mice. Furthermore, we report a significant reduction of cerebral amyloid burden and BACE1 accumulation in dystrophic neurites in the absence of BACE1 S-palmitoylation in mouse models of AD amyloidosis. Studies in cultured neurons suggest that S-palmitoylation is required for dendritic spine localization and axonal targeting of BACE1. Finally, the lack of BACE1 S-palmitoylation mitigates cognitive deficits in 5XFAD mice. Using transgenic mouse models, these results demonstrate that intrinsic posttranslational S-palmitoylation of BACE1 has a significant impact on amyloid pathogenesis and the consequent cognitive decline.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Transtornos da Memória/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Amiloidogênicas/metabolismo , Amiloidose/metabolismo , Animais , Axônios/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Lipoilação/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia
2.
Mol Neurodegener ; 9: 1, 2014 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-24386896

RESUMO

BACKGROUND: BACE1 is one of the two enzymes that cleave amyloid precursor protein to generate Alzheimer's disease (AD) beta amyloid peptides. It is widely believed that BACE1 initiates APP processing in endosomes, and in the brain this cleavage is known to occur during axonal transport of APP. In addition, BACE1 accumulates in dystrophic neurites surrounding brain senile plaques in individuals with AD, suggesting that abnormal accumulation of BACE1 at presynaptic terminals contributes to pathogenesis in AD. However, only limited information is available on BACE1 axonal transport and targeting. RESULTS: By visualizing BACE1-YFP dynamics using live imaging, we demonstrate that BACE1 undergoes bi-directional transport in dynamic tubulo-vesicular carriers along axons in cultured hippocampal neurons and in acute hippocampal slices of transgenic mice. In addition, a subset of BACE1 is present in larger stationary structures, which are active presynaptic sites. In cultured neurons, BACE1-YFP is preferentially targeted to axons over time, consistent with predominant in vivo localization of BACE1 in presynaptic terminals. Confocal analysis and dual-color live imaging revealed a localization and dynamic transport of BACE1 along dendrites and axons in Rab11-positive recycling endosomes. Impairment of Rab11 function leads to a diminution of total and endocytosed BACE1 in axons, concomitant with an increase in the soma. Together, these results suggest that BACE1 is sorted to axons in endosomes in a Rab11-dependent manner. CONCLUSION: Our results reveal novel information on dynamic BACE1 transport in neurons, and demonstrate that Rab11-GTPase function is critical for axonal sorting of BACE1. Thus, we suggest that BACE1 transcytosis in endosomes contributes to presynaptic BACE1 localization.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Transporte Axonal/fisiologia , Axônios/metabolismo , Hipocampo/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Endossomos/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Neurônios/metabolismo , Técnicas de Cultura de Órgãos
3.
Cell Rep ; 5(6): 1552-63, 2013 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-24373286

RESUMO

Abnormal accumulation of ß-secretase (BACE1) in dystrophic neurites and presynaptic ß-amyloid (Aß) production contribute to Alzheimer's disease pathogenesis. Little, however, is known about BACE1 sorting and dynamic transport in neurons. We investigated BACE1 trafficking in hippocampal neurons using live-cell imaging and selective labeling. We report that transport vesicles containing internalized BACE1 in dendrites undergo exclusive retrograde transport toward the soma, whereas they undergo bidirectional transport in axons. Unidirectional dendritic transport requires Eps15-homology-domain-containing (EHD) 1 and 3 protein function. Furthermore, loss of EHD function compromises dynamic axonal transport and overall BACE1 levels in axons. EHD1/3 colocalize with BACE1 and APP ß-C-terminal fragments in hippocampal mossy fiber terminals, and their depletion in neurons significantly attenuates Aß levels. These results demonstrate unidirectional endocytic transport of a dendritic cargo and reveal a role for EHD proteins in neuronal BACE1 transcytosis and Aß production, processes that are highly relevant for Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Transporte Axonal , Proteínas de Transporte/metabolismo , Dendritos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Proteínas de Transporte/genética , Células Cultivadas , Células HEK293 , Células HeLa , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Camundongos , Transporte Proteico , Proteínas de Transporte Vesicular/genética
4.
J Biol Chem ; 288(37): 26955-66, 2013 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23902769

RESUMO

Alzheimer disease (AD), the leading cause of dementia, is characterized by the accumulation of ß-amyloid peptides (Aß) in senile plaques in the brains of affected patients. Many cellular mechanisms are thought to play important roles in the development and progression of AD. Several lines of evidence point to the dysregulation of Ca(2+) homeostasis as underlying aspects of AD pathogenesis. Moreover, direct roles in the regulation of Ca(2+) homeostasis have been demonstrated for proteins encoded by familial AD-linked genes such as PSEN1, PSEN2, and APP, as well as Aß peptides. Whereas these studies support the hypothesis that disruption of Ca(2+) homeostasis contributes to AD, it is difficult to disentangle the effects of familial AD-linked genes on Aß production from their effects on Ca(2+) homeostasis. Here, we developed a system in which cellular Ca(2+) homeostasis could be directly manipulated to study the effects on amyloid precursor protein metabolism and Aß production. We overexpressed stromal interaction molecule 1 (STIM1) and Orai1, the components of the store-operated Ca(2+) entry pathway, to generate cells with constitutive and store depletion-induced Ca(2+) entry. We found striking effects of Ca(2+) entry induced by overexpression of the constitutively active STIM1(D76A) mutant on amyloid precursor protein metabolism. Specifically, constitutive activation of Ca(2+) entry by expression of STIM1(D76A) significantly reduced Aß secretion. Our results suggest that disruptions in Ca(2+) homeostasis may influence AD pathogenesis directly through the modulation of Aß production.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Regulação da Expressão Gênica , Sinalização do Cálcio , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Homeostase , Humanos , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Molécula 1 de Interação Estromal
5.
J Neurosci ; 32(5): 1714-29, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22302812

RESUMO

Numerous physiological functions, including a role as a cell surface receptor, have been ascribed to Alzheimer's disease-associated amyloid precursor protein (APP). However, detailed analysis of intracellular signaling mediated by APP in neurons has been lacking. Here, we characterized intrinsic signaling associated with membrane-bound APP C-terminal fragments, which are generated following APP ectodomain release by α- or ß-secretase cleavage. We found that accumulation of APP C-terminal fragments or expression of membrane-tethered APP intracellular domain results in adenylate cyclase-dependent activation of PKA (protein kinase A) and inhibition of GSK3ß signaling cascades, and enhancement of axodendritic arborization in rat immortalized hippocampal neurons, mouse primary cortical neurons, and mouse neuroblastoma. We discovered an interaction between BBXXB motif of APP intracellular domain and the heterotrimeric G-protein subunit Gα(S), and demonstrate that Gα(S) coupling to adenylate cyclase mediates membrane-tethered APP intracellular domain-induced neurite outgrowth. Our study provides clear evidence that APP intracellular domain can have a nontranscriptional role in regulating neurite outgrowth through its membrane association. The novel functional coupling of membrane-bound APP C-terminal fragments with Gα(S) signaling identified in this study could impact several brain functions such as synaptic plasticity and memory formation.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Membranas Intracelulares/fisiologia , Transdução de Sinais/fisiologia , Adenilil Ciclases/metabolismo , Adenilil Ciclases/fisiologia , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/química , Animais , Células COS , Linhagem Celular Transformada , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/fisiologia , Proliferação de Células , Chlorocebus aethiops , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Membranas Intracelulares/química , Masculino , Camundongos , Dados de Sequência Molecular , Neuritos/fisiologia , Estrutura Terciária de Proteína , Ratos
6.
Mol Neurodegener ; 6: 87, 2011 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-22204304

RESUMO

BACKGROUND: p23 belongs to the highly conserved p24 family of type I transmembrane proteins, which participate in the bidirectional protein transport between the endoplasmic reticulum and Golgi apparatus. Mammalian p23 has been shown to interact with γ-secretase complex, and modulate secretory trafficking as well as intramembranous processing of amyloid precursor protein in cultured cells. Negative modulation of ß-amyloid production by p23 in cultured cell lines suggested that elevation of p23 expression in neurons might mitigate cerebral amyloid burden. RESULTS: We generated several lines of transgenic mice expressing human p23 in neurons under the control of Thy-1.2 promoter. We found that even a 50% increase in p23 levels in the central nervous system of mice causes post-natal growth retardation, severe neurological problems characterized by tremors, seizure, ataxia, and uncoordinated movements, and premature death. The severity of the phenotype closely correlated with the level of p23 overexpression in multiple transgenic lines. While the number and general morphology of neurons in Hup23 mice appeared to be normal throughout the brain, abnormal non-Golgi p23 localization was observed in a subset of neurons with high transgene expression in brainstem. Moreover, detailed immunofluorescence analysis revealed marked proliferation of astrocytes, activation of microglia, and thinning of myelinated bundles in brainstem of Hup23 mice. CONCLUSIONS: These results demonstrate that proper level of p23 expression is critical for neuronal function, and perturbing p23 function by overexpression initiates a cascade of cellular reactions in brainstem that leads to severe motor deficits and other neurological problems, which culminate in premature death. The neurological phenotype observed in Hup23 mice highlights significant adverse effects associated with manipulating neuronal expression of p23, a previously described negative modulator of γ-secretase activity and ß-amyloid production. Moreover, our report has broader relevance to molecular mechanisms in several neurodegenerative diseases as it highlights the inherent vulnerability of the early secretory pathway mechanisms that ensure proteostasis in neurons.


Assuntos
Proteínas de Membrana/metabolismo , Atividade Motora/fisiologia , Transtornos dos Movimentos/fisiopatologia , Neurônios/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Células Cultivadas , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos dos Movimentos/patologia , Proteína Básica da Mielina/metabolismo , Neurônios/citologia , Neurônios/patologia , Proteínas de Transporte Nucleocitoplasmático
7.
J Biol Chem ; 286(29): 26166-77, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21642424

RESUMO

Several lines of evidence implicate lipid raft microdomains in Alzheimer disease-associated ß-amyloid peptide (Aß) production. Notably, targeting ß-secretase (ß-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1)) exclusively to lipid rafts by the addition of a glycosylphosphatidylinositol (GPI) anchor to its ectodomain has been reported to elevate Aß secretion. Paradoxically, Aß secretion is not reduced by the expression of non-raft resident S-palmitoylation-deficient BACE1 (BACE1-4C/A (C474A/C478A/C482A/C485A)). We addressed this apparent discrepancy in raft microdomain-associated BACE1 processing of APP in this study. As previously reported, we found that expression of BACE1-GPI elevated Aß secretion as compared with wild-type BACE1 (WTBACE1) or BACE1-4C/A. However, this increase occurred without any difference in the levels of APP ectodomain released following BACE1 cleavage (soluble APPß), arguing against an overall increase in BACE1 processing of APP per se. Further analysis revealed that WTBACE1 cleaves APP at ß- and ß'-sites, generating +1 and +11 ß-C-terminal fragments and secreting intact as well as N-terminally truncated Aß. In contrast, three different BACE1-GPI chimeras preferentially cleaved APP at the ß-site, mainly generating +1 ß-C-terminal fragment and secreting intact Aß. As a consequence, cells expressing BACE1-GPI secreted relatively higher levels of intact Aß without an increase in BACE1 processing of APP. Markedly reduced cleavage at ß'-site exhibited by BACE1-GPI was cell type-independent and insensitive to subcellular localization of APP or the pathogenic KM/NL mutant. We conclude that the apparent elevation in Aß secretion by BACE1-GPI is mainly attributed to preferential cleavage at the ß-site and failure to detect +11 Aß species secreted by cells expressing WTBACE1.


Assuntos
Secretases da Proteína Precursora do Amiloide/química , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/química , Ácido Aspártico Endopeptidases/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/química , Animais , Ácido Aspártico Endopeptidases/genética , Sítios de Ligação , Membrana Celular/metabolismo , Regulação Enzimológica da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutagênese , Mutação , Fragmentos de Peptídeos/metabolismo , Estrutura Terciária de Proteína , Solubilidade , Especificidade por Substrato
8.
J Biol Chem ; 285(49): 38042-52, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20921220

RESUMO

The γ-secretase protein complex executes the intramembrane proteolysis of amyloid precursor protein (APP), which releases Alzheimer disease ß-amyloid peptide. In addition to APP, γ-secretase also cleaves several other type I membrane protein substrates including Notch1 and N-cadherin. γ-Secretase is made of four integral transmembrane protein subunits: presenilin (PS), nicastrin, APH1, and PEN2. Multiple lines of evidence indicate that a heteromer of PS-derived N- and C-terminal fragments functions as the catalytic subunit of γ-secretase. Only limited information is available on the domains within each subunit involved in the recognition and recruitment of diverse substrates and the transfer of substrates to the catalytic site. Here, we performed mutagenesis of two domains of PS1, namely the first luminal loop domain (LL1) and the second transmembrane domain (TM2), and analyzed PS1 endoproteolysis as well as the catalytic activities of PS1 toward APP, Notch, and N-cadherin. Our results show that distinct residues within LL1 and TM2 domains as well as the length of the LL1 domain are critical for PS1 endoproteolysis, but not for PS1 complex formation with nicastrin, APH1, and PEN2. Furthermore, our experimental PS1 mutants formed γ-secretase complexes with distinct catalytic properties toward the three substrates examined in this study; however, the mutations did not affect PS1 interaction with the substrates. We conclude that the N-terminal LL1 and TM2 domains are critical for PS1 endoproteolysis and the coordination between the putative substrate-docking site and the catalytic core of the γ-secretase.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Presenilina-1/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide , Animais , Caderinas/genética , Caderinas/metabolismo , Domínio Catalítico/fisiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutagênese , Mutação , Presenilina-1/genética , Estrutura Terciária de Proteína , Receptor Notch1/genética , Receptor Notch1/metabolismo , Especificidade por Substrato/fisiologia
9.
Biochim Biophys Acta ; 1801(8): 860-7, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20303415

RESUMO

Alzheimer's disease (AD), the most common age-associated dementing disorder, is pathologically manifested by progressive cognitive dysfunction concomitant with the accumulation of senile plaques consisting of amyloid-beta (Abeta) peptide aggregates in the brain of affected individuals. Abeta is derived from a type I transmembrane protein, amyloid precursor protein (APP), by the sequential proteolytic events mediated by beta-site APP cleaving enzyme 1 (BACE1) and gamma-secretase. Multiple lines of evidence have implicated cholesterol and cholesterol-rich membrane microdomains, termed lipid rafts in the amyloidogenic processing of APP. In this review, we summarize the cell biology of APP, beta- and gamma-secretases and the data on their association with lipid rafts. Then, we will discuss potential raft targeting signals identified in the secretases and their importance on amyloidogenic processing of APP.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Microdomínios da Membrana/fisiologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Biologia Celular , Humanos , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Processamento de Proteína Pós-Traducional/fisiologia
10.
J Biol Chem ; 284(6): 3793-803, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19074428

RESUMO

Alzheimer disease beta-amyloid (Abeta) peptides are generated via sequential proteolysis of amyloid precursor protein (APP) by BACE1 and gamma-secretase. A subset of BACE1 localizes to cholesterol-rich membrane microdomains, termed lipid rafts. BACE1 processing in raft microdomains of cultured cells and neurons was characterized in previous studies by disrupting the integrity of lipid rafts by cholesterol depletion. These studies found either inhibition or elevation of Abeta production depending on the extent of cholesterol depletion, generating controversy. The intricate interplay between cholesterol levels, APP trafficking, and BACE1 processing is not clearly understood because cholesterol depletion has pleiotropic effects on Golgi morphology, vesicular trafficking, and membrane bulk fluidity. In this study, we used an alternate strategy to explore the function of BACE1 in membrane microdomains without altering the cellular cholesterol level. We demonstrate that BACE1 undergoes S-palmitoylation at four Cys residues at the junction of transmembrane and cytosolic domains, and Ala substitution at these four residues is sufficient to displace BACE1 from lipid rafts. Analysis of wild type and mutant BACE1 expressed in BACE1 null fibroblasts and neuroblastoma cells revealed that S-palmitoylation neither contributes to protein stability nor subcellular localization of BACE1. Surprisingly, non-raft localization of palmitoylation-deficient BACE1 did not have discernible influence on BACE1 processing of APP or secretion of Abeta. These results indicate that post-translational S-palmitoylation of BACE1 is not required for APP processing, and that BACE1 can efficiently cleave APP in both raft and non-raft microdomains.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Microdomínios da Membrana/metabolismo , Ácidos Palmíticos/metabolismo , Processamento de Proteína Pós-Traducional , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/genética , Animais , Ácido Aspártico Endopeptidases/genética , Colesterol/genética , Colesterol/metabolismo , Estabilidade Enzimática/genética , Estabilidade Enzimática/fisiologia , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Humanos , Microdomínios da Membrana/genética , Camundongos , Camundongos Knockout , Transporte Proteico/genética
11.
J Biol Chem ; 284(3): 1373-84, 2009 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-19028695

RESUMO

Proteolytic processing of amyloid precursor protein (APP) by beta- and gamma-secretases generates beta-amyloid (Abeta) peptides, which accumulate in the brains of individuals affected by Alzheimer disease. Detergent-resistant membrane microdomains (DRM) rich in cholesterol and sphingolipid, termed lipid rafts, have been implicated in Abeta production. Previously, we and others reported that the four integral subunits of the gamma-secretase associate with DRM. In this study we investigated the mechanisms underlying DRM association of gamma-secretase subunits. We report that in cultured cells and in brain the gamma-secretase subunits nicastrin and APH-1 undergo S-palmitoylation, the post-translational covalent attachment of the long chain fatty acid palmitate common in lipid raft-associated proteins. By mutagenesis we show that nicastrin is S-palmitoylated at Cys(689), and APH-1 is S-palmitoylated at Cys(182) and Cys(245). S-Palmitoylation-defective nicastrin and APH-1 form stable gamma-secretase complexes when expressed in knock-out fibroblasts lacking wild type subunits, suggesting that S-palmitoylation is not essential for gamma-secretase assembly. Nevertheless, fractionation studies show that S-palmitoylation contributes to DRM association of nicastrin and APH-1. Moreover, pulse-chase analyses reveal that S-palmitoylation is important for nascent polypeptide stability of both proteins. Co-expression of S-palmitoylation-deficient nicastrin and APH-1 in cultured cells neither affects Abeta40, Abeta42, and AICD production, nor intramembrane processing of Notch and N-cadherin. Our findings suggest that S-palmitoylation plays a role in stability and raft localization of nicastrin and APH-1, but does not directly modulate gamma-secretase processing of APP and other substrates.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Lipoilação/fisiologia , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular , Endopeptidases , Estabilidade Enzimática/fisiologia , Humanos , Glicoproteínas de Membrana/genética , Lipídeos de Membrana/genética , Lipídeos de Membrana/metabolismo , Microdomínios da Membrana/genética , Proteínas de Membrana/genética , Camundongos , Peptídeo Hidrolases , Receptores Notch/genética , Receptores Notch/metabolismo
12.
Neurobiol Dis ; 32(1): 37-49, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18652896

RESUMO

Sequential processing of amyloid precursor protein by beta- and gamma-secretases generates Alzheimer's disease (AD)-associated beta-amyloid peptides. Recently it was reported that the transmembrane protein p23/TMP21 associates with gamma-secretase, and negatively regulates beta-amyloid production. Despite the link between p23 function and AD pathogenesis, the expression of p23 has not been examined in the brain. Here, we describe the detailed immunohistochemical characterization of p23 expression in rodent and human brain. We report that p23 is co-expressed with gamma-secretase subunits in select neuronal cell populations in rodent brain. Interestingly, the steady-state level of p23 in the brain is high during embryonic development and then declines after birth. Furthermore, the steady-state p23 levels are reduced in the brains of individuals with AD. We conclude that p23 is expressed in neurons throughout the brain and the decline in p23 expression during postnatal development may significantly contribute to enhanced beta-amyloid production in the adult brain.


Assuntos
Proteínas de Membrana/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Animais Recém-Nascidos , Química Encefálica/genética , Linhagem Celular Tumoral , Células Cultivadas , Cerebelo/química , Cerebelo/metabolismo , Lobo Frontal/química , Lobo Frontal/metabolismo , Células HeLa , Hipocampo/química , Hipocampo/metabolismo , Humanos , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Pessoa de Meia-Idade , Proteínas de Transporte Nucleocitoplasmático , Ratos , Ratos Sprague-Dawley
13.
J Biol Chem ; 283(28): 19489-98, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18456655

RESUMO

Cerebral deposition of beta-amyloid (Abeta) peptides is a pathological hallmark of Alzheimer disease. Intramembranous proteolysis of amyloid precursor protein by a multiprotein gamma-secretase complex generates Abeta. Previously, it was reported that CD147, a glycoprotein that stimulates production of matrix metalloproteinases (MMPs), is a subunit of gamma-secretase and that the levels of secreted Abeta inversely correlate with CD147 expression. Here, we show that the levels and localization of CD147 in fibroblasts, as well as postnatal expression and distribution in brain, are distinct from those of integral gamma-secretase subunits. Notably, we show that although depletion of CD147 increased extracellular Abeta levels in intact cells, membranes isolated from CD147-depleted cells failed to elevate Abeta production in an in vitro gamma-secretase assay. Consistent with an extracellular source that modulates Abeta metabolism, synthetic Abeta was degraded more rapidly in the conditioned medium of cells overexpressing CD147. Moreover, modulation of CD147 expression had no effect on epsilon-site cleavage of amyloid precursor protein and Notch1 receptor. Collectively, our results demonstrate that CD147 modulates Abeta levels not by regulating gamma-secretase activity, but by stimulating extracellular degradation of Abeta. In view of the known function of CD147 in MMP production, we postulate that CD147 expression influences Abeta levels by an indirect mechanism involving MMPs that can degrade extracellular Abeta.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Basigina/metabolismo , Metaloproteinases da Matriz/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Basigina/genética , Linhagem Celular , Cerebelo/metabolismo , Cerebelo/patologia , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Receptor Notch1/genética , Receptor Notch1/metabolismo
14.
Nat Clin Pract Neurol ; 3(7): 374-82, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17611486

RESUMO

Alzheimer's disease (AD) is the most common cause of age-related dementia. Pathologically, AD is characterized by the deposition in the brain of amyloid-beta peptides derived from proteolysis of amyloid precursor protein (APP) by beta-site APP cleaving enzyme 1 (BACE1) and gamma-secretase. A growing body of evidence implicates cholesterol and cholesterol-rich membrane microdomains in amyloidogenic processing of APP. Here, we review recent findings regarding the association of BACE1, gamma-secretase and APP in lipid rafts, and discuss potential therapeutic strategies for AD that are based on knowledge gleaned from the membrane environment that fosters APP processing.


Assuntos
Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/metabolismo , Microdomínios da Membrana/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Humanos , Transporte Proteico/fisiologia
15.
Mol Neurodegener ; 2: 4, 2007 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-17288597

RESUMO

BACKGROUND: Alzheimer's disease (AD) is characterized by cerebral deposition of beta-amyloid (Abeta) peptides. Abeta is released from ectodomain cleaved amyloid precursor protein (APP) via intramembranous proteolysis by gamma-secretase, a complex consisting of presenilin and a few other proteins. p23/TMP21, a member of the p24 family type I transmembrane proteins, was recently identified as a presenilin complex component capable of modulating gamma-secretase cleavage. The p24 family proteins form oligomeric complexes and regulate vesicular trafficking in the early secretory pathway, but their role in APP trafficking has not been investigated. RESULTS: Here, we report that siRNA-mediated depletion of p23 in N2a neuroblastoma and HeLa cells produces concomitant knockdown of additional p24 family proteins and increases secretion of sAPP. Furthermore, intact cell and cell-free Abeta production increases following p23 knockdown, similar to data reported earlier using HEK293 cells. However, we find that p23 is not present in mature gamma-secretase complexes isolated using an active-site gamma-secretase inhibitor. Depletion of p23 and expression of a familial AD-linked PS1 mutant have additive effects on Abeta42 production. Knockdown of p23 expression confers biosynthetic stability to nascent APP, allowing its efficient maturation and surface accumulation. Moreover, immunoisolation analyses show decrease in co-residence of APP and the APP adaptor Mint3. Thus, multiple lines of evidence indicate that p23 function influences APP trafficking and sAPP release independent of its reported role in gamma-secretase modulation. CONCLUSION: These data assign significance to p24 family proteins in regulating APP trafficking in the continuum of bidirectional transport between the ER and Golgi, and ascribe new relevance to the regulation of early trafficking in AD pathogenesis.

16.
Mol Neurodegener ; 1: 4, 2006 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-16930451

RESUMO

Mutations in PSEN1 and PSEN2 genes account for the majority of cases of early-onset familial Alzheimer disease. Since the first prediction of a genetic link between PSEN1 and PSEN2 with Alzheimer's disease, many research groups from both academia and pharmaceutical industry have sought to unravel how pathogenic mutations in PSEN cause presenile dementia. PSEN genes encode polytopic membrane proteins termed presenilins (PS1 and PS2), which function as the catalytic subunit of gamma-secretase, an intramembrane protease that has a wide spectrum of type I membrane protein substrates. Sequential cleavage of amyloid precursor protein by BACE and gamma-secretase releases highly fibrillogenic beta-amyloid peptides, which accumulate in the brains of aged individuals and patients with Alzheimer's disease. Familial Alzheimer's disease-associated presenilin variants are thought to exert their pathogenic function by selectively elevating the levels of highly amyloidogenic Abeta42 peptides. In addition to Alzheimer's disease, several recent studies have linked PSEN1 to familiar frontotemporal dementia. Here, we review the biology of PS1, its role in gamma-secretase activity, and discuss recent developments in the cell biology of PS1 with respect to Alzheimer's disease pathogenesis.

17.
Neurology ; 66(2 Suppl 1): S69-73, 2006 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-16432149

RESUMO

Trafficking and proteolytic processing of amyloid precursor protein (APP) have been the focus of numerous investigations in the past two decades, since the identification of Abeta as the principal component of brain senile plaques and the cloning of APP cDNA. Tremendous progress has been made in the recent past toward the characterization of beta- and gamma-secretases. Here, we review the salient features of Alzheimer disease amyloidogenesis, and discuss the current knowledge on APP trafficking and amyloidogenic processing of APP in intracellular membrane compartments and microdomains.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Compartimento Celular , Adulto , Idade de Início , Idoso , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide , Amiloidose Familiar/genética , Amiloidose Familiar/metabolismo , Ácido Aspártico Endopeptidases , Síndrome de Down/complicações , Síndrome de Down/genética , Endocitose , Endopeptidases/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Microdomínios da Membrana , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Placa Amiloide/metabolismo , Presenilina-1 , Presenilina-2 , Processamento de Proteína Pós-Traducional
18.
J Biol Chem ; 280(27): 25892-900, 2005 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-15886206

RESUMO

Gamma-secretase facilitates the regulated intramembrane proteolysis of select type I membrane proteins that play diverse physiological roles in multiple cell types and tissue. In this study, we used biochemical approaches to examine the distribution of amyloid precursor protein (APP) and several additional gamma-secretase substrates in membrane microdomains. We report that APP C-terminal fragments (CTFs) and gamma-secretase reside in Lubrol WX detergent-insoluble membranes (DIM) of cultured cells and adult mouse brain. APP CTFs that accumulate in cells lacking gamma-secretase activity preferentially associate with DIM. Cholesterol depletion and magnetic immunoisolation studies indicate recruitment of APP CTFs into cholesterol- and sphingolipid-rich lipid rafts, and co-residence of APP CTFs, PS1, and syntaxin 6 in DIM patches derived from the trans-Golgi network. Photoaffinity cross-linking studies provided evidence for the preponderance of active gamma-secretase in lipid rafts of cultured cells and adult brain. Remarkably, unlike the case of APP, CTFs derived from Notch1, Jagged2, deleted in colorectal cancer (DCC), and N-cadherin remain largely detergent-soluble, indicative of their spatial segregation in non-raft domains. In embryonic brain, the majority of PS1 and nicastrin is present in Lubrol WX-soluble membranes, wherein the CTFs derived from APP, Notch1, DCC, and N-cadherin also reside. We suggest that gamma-secretase residence in non-raft membranes facilitates proteolysis of diverse substrates during embryonic development but that the translocation of gamma-secretase to lipid rafts in adults ensures processing of certain substrates, including APP CTFs, while limiting processing of other potential substrates.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Membrana Celular/enzimologia , Lipídeos de Membrana/metabolismo , Fatores Etários , Secretases da Proteína Precursora do Amiloide , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/enzimologia , Linhagem Celular Tumoral , Colesterol/metabolismo , Endopeptidases , Deleção de Genes , Proteína Jagged-2 , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Neuroblastoma , Polietilenoglicóis , Presenilina-1 , Presenilina-2 , Proteínas Qa-SNARE , Receptor Notch1 , Receptores de Superfície Celular/metabolismo , Esfingolipídeos/metabolismo , Especificidade por Substrato , Fatores de Transcrição/metabolismo
19.
J Biol Chem ; 280(17): 17020-6, 2005 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-15711015

RESUMO

gamma-Secretase, which is responsible for the intramembranous cleavage of Alzheimer beta-amyloid precursor protein and the signaling receptor Notch, is a multiprotein complex consisting of at least four components: presenilin (PS); nicastrin (Nct); APH-1 (anterior pharynx-defective-1); and presenilin enhancer-2 (PEN-2). Presenilin 1 (PS1) is known to be essential for the stability, interaction, and trafficking of the other PS1/gamma-secretase components. However, the precise functions of the other components remain elusive. Here, we investigated the functions of Nct within the PS1/gamma-secretase complex. We demonstrated that the loss of Nct expression in the embryonic fibroblast cells (Nct KO cells) results in dramatically decreased levels of APH-1, PEN-2, and PS1 fragments accompanied by a significant accumulation of full-length PS1. In the absence of Nct, PEN-2 and full-length PS1 are subjected to proteasome-mediated degradation, whereas the degradation of APH-1 is mediated by both proteasomal and lysosomal pathways. Unlike the case of wild type cells in which the gamma-secretase complex mainly locates in the trans-Golgi network, the majority of residual PEN-2, APH-1, and the uncleaved full-length PS1 in Nct KO cells reside in the endoplasmic reticulum, which remain associated with each other in the absence of Nct. Interestingly, significant amounts of full-length PS1 and PEN-2, but not APH-1, are detected on the plasma membrane in Nct KO cells, suggesting the Nct-independent cell surface delivery of the PEN-2.PS1. Finally, the diminished PEN-2 protein level in Nct-deficient cells can be partially restored by overexpression of exogenous PS1, APH-1, or PEN-2 individually or collectively, indicating a dispensable role for Nct in controlling PEN-2 level. Taken together, our study demonstrates a critical role of Nct in the stability and proper intracellular trafficking of other components of the PS1/ gamma-secretase complex but not in maintaining the association of PEN-2, APH-1, and full-length PS1.


Assuntos
Endopeptidases/metabolismo , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/fisiologia , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Biotinilação , Membrana Celular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Imunoprecipitação , Cinética , Lisossomos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Modelos Biológicos , Presenilina-1 , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Transporte Proteico , Interferência de RNA , Receptores Notch , Rede trans-Golgi/metabolismo
20.
J Biol Chem ; 279(43): 44945-54, 2004 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-15322084

RESUMO

Alzheimer's disease-associated beta-amyloid peptides (Abeta) are generated by the sequential proteolytic processing of amyloid precursor protein (APP) by beta- and gamma-secretases. There is growing evidence that cholesterol- and sphingolipid-rich membrane microdomains are involved in regulating trafficking and processing of APP. BACE1, the major beta-secretase in neurons is a palmitoylated transmembrane protein that resides in lipid rafts. A subset of APP is subject to amyloidogenic processing by BACE1 in lipid rafts, and this process depends on the integrity of lipid rafts. Here we describe the association of all four components of the gamma-secretase complex, namely presenilin 1 (PS1)-derived fragments, mature nicastrin, APH-1, and PEN-2, with cholesterol-rich detergent insoluble membrane (DIM) domains of non-neuronal cells and neurons that fulfill the criteria of lipid rafts. In PS1(-/-)/PS2(-/-) and NCT(-/-) fibroblasts, gamma-secretase components that still remain fail to become detergent-resistant, suggesting that raft association requires gamma-secretase complex assembly. Biochemical evidence shows that subunits of the gamma-secretase complex and three TGN/endosome-resident SNAREs cofractionate in sucrose density gradients, and show similar solubility or insolubility characteristics in distinct non-ionic and zwitterionic detergents, indicative of their co-residence in membrane microdomains with similar protein-lipid composition. This notion is confirmed using magnetic immunoisolation of PS1- or syntaxin 6-positive membrane patches from a mixture of membranes with similar buoyant densities following Lubrol WX extraction or sonication, and gradient centrifugation. These findings are consistent with the localization of gamma-secretase in lipid raft microdomains of post-Golgi and endosomes, organelles previously implicated in amyloidogenic processing of APP.


Assuntos
Endopeptidases/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Membranas Intracelulares/enzimologia , Microdomínios da Membrana/enzimologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Transporte Biológico , Linhagem Celular Tumoral , Colesterol/química , Detergentes/farmacologia , Endocitose , Fibroblastos/metabolismo , Células HeLa , Humanos , Lipídeos/química , Magnetismo , Glicoproteínas de Membrana/química , Microdomínios da Membrana/química , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Mutação , Neurônios/metabolismo , Peptídeo Hidrolases , Proteínas Qa-SNARE , Proteínas R-SNARE , Frações Subcelulares , Sacarose/farmacologia , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...