Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropathol Appl Neurobiol ; 46(7): 686-706, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32716575

RESUMO

BACKGROUND: Evidence suggests that amyloid ß (Aß) peptides play an important role in the degeneration of neurons during the development of Alzheimer's disease (AD), the prevalent cause of dementia affecting the elderly. The endosomal-lysosomal system, which acts as a major site for Aß metabolism, has been shown to exhibit abnormalities in vulnerable neurons of the AD brain, reflected by enhanced levels/expression of lysosomal enzymes including cathepsin D (CatD). At present, the implication of CatD in selective neuronal vulnerability in AD pathology remains unclear. METHODS: We evaluated the role of CatD in the degeneration of neurons in Aß-treated cultures, transgenic AD mouse model (that is 5xFAD) and post mortem AD brain samples. RESULTS: Our results showed that Aß1-42 -induced toxicity in cortical cultured neurons is associated with impaired lysosomal integrity, enhanced levels of carbonylated proteins and tau phosphorylation. The cellular and cytosolic levels/activity of CatD are also elevated in cultured neurons following exposure to Aß peptide. Additionally, we observed that CatD cellular and subcellular levels/activity are increased in the affected cortex, but not in the unaffected cerebellum, of 5xFAD mice and post mortem AD brains. Interestingly, treatment of cultured neurons with nanoparticles PLGA, which targets lysosomal system, attenuated Aß toxicity by reducing the levels of carbonylated proteins, tau phosphorylation and the level/distribution/activity of CatD. CONCLUSION: Our study reveals that increased cytosolic level/activity of CatD play an important role in determining neuronal vulnerability in AD. Additionally, native PLGA can protect neurons against Aß toxicity by restoring lysosomal membrane integrity, thus signifying its implication in attenuating AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Catepsina D/metabolismo , Lisossomos/metabolismo , Neurônios/patologia , Idoso , Idoso de 80 Anos ou mais , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fragmentos de Peptídeos/metabolismo , Proteínas tau/metabolismo
2.
Mol Cell Biol ; 38(11)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29530923

RESUMO

Amyloid ß (Aß) peptide, derived from amyloid precursor protein (APP), plays a critical role in the development of Alzheimer's disease. Current evidence indicates that altered levels or subcellular distribution of cholesterol can regulate Aß production and clearance, but it remains unclear how cholesterol sequestration within the endosomal-lysosomal (EL) system can influence APP metabolism. Thus, we evaluated the effects of U18666A, which triggers cholesterol redistribution within the EL system, on mouse N2a cells expressing different levels of APP in the presence or absence of extracellular cholesterol and lipids provided by fetal bovine serum (FBS). Our results reveal that U18666A and FBS differentially increase the levels of APP and its cleaved products, the α-, ß-, and η-C-terminal fragments, in N2a cells expressing normal levels of mouse APP (N2awt), higher levels of human wild-type APP (APPwt), or "Swedish" mutant APP (APPsw). The cellular levels of Aß1-40/Aß1-42 were markedly increased in U18666A-treated APPwt and APPsw cells. Our studies further demonstrate that APP and its cleaved products are partly accumulated in the lysosomes, possibly due to decreased clearance. Finally, we show that autophagy inhibition plays a role in mediating U18666A effects. Collectively, these results suggest that altered levels and distribution of cholesterol and lipids can differentially regulate APP metabolism depending on the nature of APP expression.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Androstenos/farmacologia , Colesterol/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Doença de Alzheimer/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Células Cultivadas , Endossomos/efeitos dos fármacos , Humanos , Lisossomos/efeitos dos fármacos
3.
Mol Neurobiol ; 55(7): 5757-5766, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29052144

RESUMO

Amyloid precursor protein (APP) is cleaved by a set of proteases including α-/ß-/γ- and recently identified η-secretases, generating C-terminal fragments (CTFs) of varying lengths and amyloid ß (Aß) peptides, which are considered to play a pivotal role in Alzheimer's disease (AD) pathogenesis. Cellular cholesterol content/distribution can regulate the production/clearance of APP metabolites and hence modify AD pathology. To determine the functional relation between endosomal-lysosomal (EL) cholesterol sequestration and APP metabolism, we used our recently developed mouse N2a-ANPC cells that overexpress Swedish mutant human APP in the absence of cholesterol-trafficking Niemann-Pick type C1 (Npc1) protein. Here, we report that neither increased levels nor EL cholesterol sequestration altered APP holoprotein levels but caused the intracellular accumulation of APP α-/ß-/η-CTFs and Aß1-40/42 peptides. The levels of APP-cleaved products increased as a function of extracellular serum concentration in N2a-ANPC cells, which are more vulnerable to death than the control cells. Additionally, we show that pH of the lysosomal vesicles in N2a-ANPC cells shifted to a less acidic range with increasing serum concentrations, thus making them less efficient functionally. Interestingly, the addition of cholesterol to the culture media not only increased the levels of cellular cholesterol and APP-cleaved products but also rendered the cells more vulnerable to toxicity. Collectively, our results suggest that extracellular cholesterol concentration in serum under conditions of Npc1 deficiency can influence intracellular cholesterol content/distribution and lysosomal efficacy, triggering the accumulation of toxic APP-cleaved products, eventually leading to cell death.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Espaço Extracelular/metabolismo , Proteína C1 de Niemann-Pick/deficiência , Soro/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Colesterol/metabolismo , Camundongos , Proteína C1 de Niemann-Pick/metabolismo
4.
Mol Neurobiol ; 54(4): 2636-2658, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-26993302

RESUMO

The insulin-like growth factor II/mannose 6-phosphate (IGF-II/M6P) receptor is a multifunctional single transmembrane glycoprotein. Recent studies have advanced our understanding of the structure, ligand-binding properties, and trafficking of the IGF-II/M6P receptor. This receptor has been implicated in a variety of important cellular processes including growth and development, clearance of IGF-II, proteolytic activation of enzymes, and growth factor precursors, in addition to its well-known role in the delivery of lysosomal enzymes. The IGF-II/M6P receptor, distributed widely in the central nervous system, has additional roles in mediating neurotransmitter release and memory enhancement/consolidation, possibly through activating IGF-II-related intracellular signaling pathways. Recent studies suggest that overexpression of the IGF-II/M6P receptor may have an important role in regulating the levels of transcripts and proteins involved in the development of Alzheimer's disease (AD)-the prevalent cause of dementia affecting the elderly population in our society. It is reported that IGF-II/M6P receptor overexpression can increase the levels/processing of amyloid precursor protein leading to the generation of ß-amyloid peptide, which is associated with degeneration of neurons and subsequent development of AD pathology. Given the significance of the receptor in mediating the transport and functioning of the lysosomal enzymes, it is being considered for therapeutic delivery of enzymes to the lysosomes to treat lysosomal storage disorders. Notwithstanding these results, additional studies are required to validate and fully characterize the function of the IGF-II/M6P receptor in the normal brain and its involvement in various neurodegenerative disorders including AD. It is also critical to understand the interaction between the IGF-II/M6P receptor and lysosomal enzymes in neurodegenerative processes, which may shed some light on developing approaches to detect and prevent neurodegeneration through the dysfunction of the receptor and the endosomal-lysosomal system.


Assuntos
Cátions/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Doenças Neurodegenerativas/metabolismo , Receptor IGF Tipo 2/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Humanos , Lisossomos/metabolismo , Receptor IGF Tipo 2/química
5.
Mol Cell Biol ; 35(14): 2368-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25939386

RESUMO

Amyloid ß (Aß) peptides originating from amyloid precursor protein (APP) in the endosomal-lysosomal compartments play a critical role in the development of Alzheimer's disease (AD), the most common type of senile dementia affecting the elderly. Since insulin-like growth factor II (IGF-II) receptors facilitate the delivery of nascent lysosomal enzymes from the trans-Golgi network to endosomes, we evaluated their role in APP metabolism and cell viability using mouse fibroblast MS cells deficient in the murine IGF-II receptor and corresponding MS9II cells overexpressing the human IGF-II receptors. Our results show that IGF-II receptor overexpression increases the protein levels of APP. This is accompanied by an increase of ß-site APP-cleaving enzyme 1 levels and an increase of ß- and γ-secretase enzyme activities, leading to enhanced Aß production. At the cellular level, IGF-II receptor overexpression causes localization of APP in perinuclear tubular structures, an increase of lipid raft components, and increased lipid raft partitioning of APP. Finally, MS9II cells are more susceptible to staurosporine-induced cytotoxicity, which can be attenuated by ß-secretase inhibitor. Together, these results highlight the potential contribution of IGF-II receptor to AD pathology not only by regulating expression/processing of APP but also by its role in cellular vulnerability.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Receptor IGF Tipo 2/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Endossomos/metabolismo , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Lisossomos/metabolismo , Camundongos , Microscopia de Fluorescência , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Interferência de RNA , Receptor IGF Tipo 2/genética , Estaurosporina/farmacologia , Transfecção
6.
Glia ; 58(11): 1267-81, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20607864

RESUMO

Niemann-Pick type C (NPC) disease is an autosomal recessive neurodegenerative disorder characterized by intracellular accumulation of cholesterol and glycosphingolipids in many tissues including the brain. The disease is caused by mutations of either NPC1 or NPC2 gene and is accompanied by a severe loss of neurons in the cerebellum, but not in the hippocampus. NPC pathology exhibits some similarities with Alzheimer's disease, including increased levels of amyloid beta (Abeta)-related peptides in vulnerable brain regions, but very little is known about the expression of amyloid precursor protein (APP) or APP secretases in NPC disease. In this article, we evaluated age-related alterations in the level/distribution of APP and its processing enzymes, beta- and gamma-secretases, in the hippocampus and cerebellum of Npc1(-/-) mice, a well-established model of NPC pathology. Our results show that levels and expression of APP and beta-secretase are elevated in the cerebellum prior to changes in the hippocampus, whereas gamma-secretase components are enhanced in both brain regions at the same time in Npc1(-/-) mice. Interestingly, a subset of reactive astrocytes in Npc1(-/-) mouse brains expresses high levels of APP as well as beta- and gamma-secretase components. Additionally, the activity of beta-secretase is enhanced in both the hippocampus and cerebellum of Npc1(-/-) mice at all ages, while the level of C-terminal APP fragments is increased in the cerebellum of 10-week-old Npc1(-/-) mice. These results, taken together, suggest that increased level and processing of APP may be associated with the development of pathology and/or degenerative events observed in Npc1(-/-) mouse brains.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Cerebelo/metabolismo , Hipocampo/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/patologia , Processamento de Proteína Pós-Traducional/genética , Proteínas/genética , Proteínas/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Cerebelo/enzimologia , Cerebelo/patologia , Modelos Animais de Doenças , Ativação Enzimática/genética , Hipocampo/enzimologia , Hipocampo/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/enzimologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo
7.
Neurobiol Aging ; 29(5): 724-38, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17222950

RESUMO

Nicastrin and presenilin 1 are integral components of the high molecular weight gamma-secretase complexes that regulate proteolytic processing of various type I membrane proteins including amyloid precursor protein and Notch. At present, there is little information regarding the cellular distribution of nicastrin in the developing or adult rat brain. We report here, using immunoblotting and immunohistochemical methods, that nicastrin in the adult rat brain is widely expressed and co-localized with presenilin 1 in select neuronal populations within all major areas, including the basal forebrain, striatum, cortex, hippocampus, amygdala, thalamus, hypothalamus, cerebellum and brainstem. We also observed dense neuropil labeling in many regions in the brain, suggesting that nicastrin gets transported to dendrites and/or axon terminals in the central nervous system. The levels of nicastrin are found to be relatively high at the early stages of postnatal development and then declined gradually to reach the adult profile. At the cellular level, nicastrin is localized predominantly in neuronal cell bodies at early postnatal stages, but is apparent both in cell bodies and dendrites/neuropil in all brain regions at the later stages. The regulation of nicastrin expression and localization during development and its distribution in a wide spectrum of neurons in the postnatal and adult rat brains provide an anatomical basis to suggest a multifunctional role for the gamma-secretase complex in the developing and adult rat brains.


Assuntos
Envelhecimento/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/metabolismo , Neurônios/enzimologia , Presenilina-1/metabolismo , Animais , Animais Recém-Nascidos , Masculino , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
8.
J Biol Chem ; 276(48): 44736-43, 2001 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11574534

RESUMO

Endoplasmic reticulum (ER) stress elicits protective responses of chaperone induction and translational suppression and, when unimpeded, leads to caspase-mediated apoptosis. Alzheimer's disease-linked mutations in presenilin-1 (PS-1) reportedly impair ER stress-mediated protective responses and enhance vulnerability to degeneration. We used cleavage site-specific antibodies to characterize the cysteine protease activation responses of primary mouse cortical neurons to ER stress and evaluate the influence of a PS-1 knock-in mutation on these and other stress responses. Two different ER stressors lead to processing of the ER-resident protease procaspase-12, activation of calpain, caspase-3, and caspase-6, and degradation of ER and non-ER protein substrates. Immunocytochemical localization of activated caspase-3 and a cleaved substrate of caspase-6 confirms that caspase activation extends into the cytosol and nucleus. ER stress-induced proteolysis is unchanged in cortical neurons derived from the PS-1 P264L knock-in mouse. Furthermore, the PS-1 genotype does not influence stress-induced increases in chaperones Grp78/BiP and Grp94 or apoptotic neurodegeneration. A similar lack of effect of the PS-1 P264L mutation on the activation of caspases and induction of chaperones is observed in fibroblasts. Finally, the PS-1 knock-in mutation does not alter activation of the protein kinase PKR-like ER kinase (PERK), a trigger for stress-induced translational suppression. These data demonstrate that ER stress in cortical neurons leads to activation of several cysteine proteases within diverse neuronal compartments and indicate that Alzheimer's disease-linked PS-1 mutations do not invariably alter the proteolytic, chaperone induction, translational suppression, and apoptotic responses to ER stress.


Assuntos
Cisteína Endopeptidases/metabolismo , Retículo Endoplasmático/enzimologia , Proteínas de Choque Térmico , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Doença de Alzheimer/genética , Animais , Apoptose , Calpaína/metabolismo , Proteínas de Transporte/metabolismo , Caspase 12 , Caspase 3 , Caspase 6 , Caspases/metabolismo , Células Cultivadas , Cisteína Endopeptidases/genética , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Ativação Enzimática , Glicosilação , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Camundongos , Chaperonas Moleculares/metabolismo , Neurônios/metabolismo , Presenilina-1 , Biossíntese de Proteínas , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Fisiológico , Frações Subcelulares , eIF-2 Quinase/metabolismo
9.
J Biol Chem ; 276(46): 43343-50, 2001 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-11564743

RESUMO

PS1 deficiency and expression of PS1 with substitutions of two conserved transmembrane aspartate residues ("PS1 aspartate variants") leads to the reduction of Abeta peptide secretion and the accumulation of amyloid precursor protein (APP) C-terminal fragments. To define the nature of the "dominant negative" effect of the PS1 aspartate variants, we stably expressed PS1 harboring aspartate to alanine substitutions at codons 257 (D257A) or 385 (D385A), singly or in combination (D257A/D385A), in mouse neuroblastoma, N2a cells. Expression of the PS1 aspartate variants resulted in marked accumulation of intracellular and cell surface APP C-terminal fragments. While expression of the D385A PS1 variant reduced the levels of secreted Abeta peptides, we now show that neither the PS1 D257A nor D257A/D385A variants impair Abeta production. Surprisingly, the stability of both immature and mature forms of APP is dramatically elevated in cells expressing PS1 aspartate variants, commensurate with an increase in the cell surface levels of APP. These findings lead us to conclude that the stability and trafficking of APP can be profoundly modulated by coexpression of PS1 with mutations at aspartate 257 and aspartate 385.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico/química , Proteínas de Membrana/química , Proteínas de Membrana/genética , Alanina/química , Secretases da Proteína Precursora do Amiloide , Animais , Ácido Aspártico Endopeptidases , Biotinilação , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Códon , DNA Complementar/metabolismo , Endopeptidases/química , Camundongos , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Mutação , Peptídeos/química , Testes de Precipitina , Presenilina-1 , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
10.
J Mol Neurosci ; 17(2): 183-92, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11816791

RESUMO

Understanding mechanisms involved in the production of Abeta has long been the central focus of cell biologists engaged in molecular AD research. The discovery of two genes that encode homologous polytopic membrane proteins termed Presenilins (PS), has lead to several exciting recent findings on the proteolytic processes responsible for generating the COOH-terminus of Abeta. What we now know is that PS proteins play an important role in Abeta production and are considered one of the therapeutic targets. Here I have reviewed the vast literature on the biology of PS, especially focusing on PS endoproteolysis and the accumulation of stable PS derivatives that are likely the functional units.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Membrana/metabolismo , Peptídeo Hidrolases/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Caspases/metabolismo , Domínio Catalítico/genética , Humanos , Proteínas de Membrana/genética , Peptídeo Hidrolases/genética , Presenilina-1 , Presenilina-2
11.
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
12.
Neurobiol Dis ; 7(2): 99-117, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10783294

RESUMO

We have investigated the subcellular distribution of presenilin-1 (PS1) and presenilin-2 (PS2) in a variety of mammalian cell lines. In Iodixanol-based density gradients, PS1 derivatives show a biphasic distribution, cofractionating with membranes containing ER-resident proteins and an additional population of membranes with low buoyant density that do not contain markers of the Golgi complex, ERGIC, COP II vesicles, ER exit compartment, COP II receptor, Golgi SNARE, trans-Golgi network, caveolar membranes, or endocytic vesicles. Confocal immunofluorescence and immunoelectron microscopy studies fully supported the fractionation studies. These data suggest that PS1 fragments accumulate in a unique subcompartment(s) of the ER or ER to Golgi trafficking intermediates. Interestingly, the FAD-linked PS1 variants show a marked redistribution toward the heavier region of the gradient. Finally, and in contrast to PS1, PS2 fragments are detected preponderantly in more densely sedimenting membranes, suggesting that the subcellular compartments in which these molecules accumulate are distinct.


Assuntos
Doença de Alzheimer/metabolismo , Membrana Celular/metabolismo , Proteínas de Membrana/metabolismo , Frações Subcelulares/metabolismo , Doença de Alzheimer/patologia , Animais , Técnicas de Cultura de Células , Membrana Celular/ultraestrutura , Imunofluorescência , Camundongos , Microscopia Confocal , Microscopia Eletrônica , Presenilina-1 , Presenilina-2 , Frações Subcelulares/ultraestrutura
13.
Nat Cell Biol ; 2(12): 863-70, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11146649

RESUMO

Presenilin 1 (PS1), a polytopic membrane protein, has a critical role in the trafficking and proteolysis of a selected set of transmembrane proteins. The vast majority of individuals affected with early onset familial Alzheimer's disease (FAD) carry missense mutations in PS1. Two studies have suggested that loss of PS1 function, or expression of FAD-linked PS1 variants, compromises the mammalian unfolded-protein response (UPR), and we sought to evaluate the potential role of PS1 in the mammalian UPR. Here we show that that neither the endoplasmic reticulum (ER) stress-induced accumulation of BiP and CHOP messenger RNA, nor the activation of ER stress kinases IRE1alpha and PERK, is compromised in cells lacking both PS1 and PS2 or in cells expressing FAD-linked PS1 variants. We also show that the levels of BiP are not significantly different in the brains of individuals with sporadic Alzheimer's disease or PS1-mediated FAD to levels in control brains. Our findings provide evidence that neither loss of PS1 and PS2 function, nor expression of PS1 variants, has a discernable impact on ER stress-mediated induction of the several established 'readouts' of the UPR pathway.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas de Transporte/genética , Proteínas de Choque Térmico , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/genética , Fatores de Transcrição/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Variação Genética , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Presenilina-1 , Presenilina-2 , Desnaturação Proteica , Dobramento de Proteína , Proteínas/química , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição CHOP , Regulação para Cima
14.
Ann N Y Acad Sci ; 920: 93-9, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11193182

RESUMO

Vidal et al. (1999. Nature 399: 776-778) discovered that the underlying genetic lesion in familial British dementia (FBD) is a T-A transversion at the termination codon of a membrane protein, termed BRI. The mutation creates an arginine codon; translational read-through generates a novel protein, termed BRI-L, that is extended by 11 amino acids at the carboxyl-terminus. BRI-L is the precursor of the ABri peptide, a component of amyloid deposits in FBD brain. We demonstrate that both BRI and its mutant counterpart are constitutively processed by furin, resulting in the secretion of carboxyl-terminal peptide derivatives that correspond to all, or part of, ABri. Notably, elevated levels of peptides are generated from the mutant BRI precursor, suggesting that subtle conformational alterations at the carboxyl-terminus may influence furin-mediated processing. We have examined BRI/BRI-L processing by other members of the prohormone convertase (PC) family (PACE4, LPC, PC 5/6) and found that these enzymes also process BRI, albeit inefficiently. Moreover, BRI-L processing by the other PC members is severely compromised. Finally, our electron microscopic studies reveal that synthetic ABri peptides assemble into insoluble beta-pleated fibrils. Collectively, our results support the view that enhanced furin-mediated processing of mutant BRI generates amyloidogenic peptides that initiate the pathogenesis of FBD.


Assuntos
Amiloide/genética , Amiloide/metabolismo , Encéfalo/metabolismo , Demência/genética , Fragmentos de Peptídeos/genética , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Amiloide/química , Animais , Células Cultivadas , Códon de Terminação , Furina , Humanos , Mamíferos , Glicoproteínas de Membrana , Proteínas de Membrana , Mutação de Sentido Incorreto , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Subtilisinas/metabolismo , Transfecção , Reino Unido
15.
Neuron ; 28(2): 461-73, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11144356

RESUMO

Amyloid precursor protein (APP) generates the beta-amyloid peptide, postulated to participate in the neurotoxicity of Alzheimer's disease. We report that APP and APLP bind to heme oxygenase (HO), an enzyme whose product, bilirubin, is antioxidant and neuroprotective. The binding of APP inhibits HO activity, and APP with mutations linked to the familial Alzheimer's disease (FAD) provides substantially greater inhibition of HO activity than wild-type APP. Cortical cultures from transgenic mice expressing Swedish mutant APP have greatly reduced bilirubin levels, establishing that mutant APP inhibits HO activity in vivo. Oxidative neurotoxicity is markedly greater in cerebral cortical cultures from APP Swedish mutant transgenic mice than wild-type cultures. These findings indicate that augmented neurotoxicity caused by APP-HO interactions may contribute to neuronal cell death in Alzheimer's disease.


Assuntos
Doença de Alzheimer/enzimologia , Precursor de Proteína beta-Amiloide/análogos & derivados , Precursor de Proteína beta-Amiloide/metabolismo , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Heme Oxigenase (Desciclizante)/metabolismo , Neurônios/enzimologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Bilirrubina/metabolismo , Ligação Competitiva/genética , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Relação Dose-Resposta a Droga , Heme Oxigenase-1 , Hemina/toxicidade , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/efeitos dos fármacos , Neurônios/patologia , Estresse Oxidativo/genética , Estrutura Terciária de Proteína/genética , Transfecção , Técnicas do Sistema de Duplo-Híbrido
16.
Methods Mol Med ; 32: 283-96, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-21318526

RESUMO

Mutations in two related genes, PS1 (1) and PS2 (2,3) located on chromosomes 14 and 1, respectively, account for the majority of early onset cases of familial Alzheimer's disease (FAD). PS1 and PS2 are predominantly localized in the endoplasmic reticulum and Golgi (4-7). PS1 is a 467 amino acid peptide predicted to contain between seven and nine transmembrane helices based on hydrophobicity profiles (1,8). The protein topology of PS1 and its C. elegans homologues, SEL-12 and HOP-1, have been examined by several investigators (7,9-13). This chapter describes two approaches we utilized to determine the topological orientation of the PS1 N-terminal, and C-terminal domains, and a hydrophilic "loop" region encompassing amino acids 263-407. The first approach is based on the proteolytic sensitivity of amyloid precursor protein (APP) protein chimeras to endoproteolytic cleavage by ß-secretase in the lumen of the Golgi. The second approach is based on selective permeabilization of the plasma membrane using a bacterial pore-forming toxin, streptolysin-O (SLO), and subsequent immunocytochemical probing for cytosolic epitopes using specific antibodies. Both of these methods can be easily adapted to determine the topology of other membrane proteins.

17.
Curr Biol ; 9(24): 1493-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10607593

RESUMO

Studies in invertebrates have indicated a functional requirement for presenilin (PS) genes in the Notch pathway [1-5]. One model of Notch signal transduction suggests that proteolysis releases an activated Notch fragment that migrates to the nucleus and regulates gene transcription in concert with CBF1/Su(H)/lag1 (CSL) proteins [6-9]. Recent studies suggest that PS genes control the proteolysis and nuclear access of the Notch intracellular domain [3,4,10,11], offering a basis for the functional interaction of PS and Notch genes [12]. Here, we report that Notch1 signaling elicited by the ligand Delta1 was quantitatively unchanged in PS1-deficient primary embryonic fibroblasts (PEFs). Notch1 signals were measured by both the activation of the hairy/enhancer of split (HES1) promoter and by the antagonism of MyoD-induced muscle creatine kinase (MCK) promoter activity. A membrane-tethered ligand-independent Notch1 construct also showed full efficacy in both assays, despite its presumed requirement for cleavage. Although signaling through Notch1 persisted in PS1-deficient cells, we found a marked reduction in the appearance of a complex of a cleaved, intracellular Notch fragment (NICD) and a CSL protein, as previously reported [6] [10]. These studies reveal that PS1 is not required for ligand-dependent Notch signaling, and that PS1 and PS2 may be redundant. Our data also suggest that the identified NICD fragment may not be necessary for Notch signal transduction [9].


Assuntos
Proteínas de Membrana/fisiologia , Receptores de Superfície Celular , Fatores de Transcrição , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Sítios de Ligação , Células Cultivadas , Regulação da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Proteínas de Membrana/química , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteína MyoD/fisiologia , Presenilina-1 , Presenilina-2 , Regiões Promotoras Genéticas , Receptor Notch1 , Transdução de Sinais , Fatores de Transcrição HES-1
19.
Nat Neurosci ; 2(11): 984-8, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10526337

RESUMO

The genetic lesion underlying familial British dementia (FBD), an autosomal dominant neurodegenerative disorder, is a T-A transversion at the termination codon of the BRI gene. The mutant gene encodes BRI-L, the precursor of ABri peptides that accumulate in amyloid deposits in FBD brain. We now report that both BRI-L and its wild-type counterpart, BRI, were constitutively processed by the proprotein convertase, furin, resulting in the secretion of carboxyl-terminal peptides that encompass all or part of ABri. Elevated levels of peptides were generated from the mutant BRI precursor. Electron microscopic studies revealed that synthetic ABri peptides assembled into irregular, short fibrils. Collectively, our results support the view that enhanced furin-mediated processing of mutant BRI generates fibrillogenic peptides that initiate the pathogenesis of FBD.


Assuntos
Amiloide/biossíntese , Demência/genética , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Neurofibrilas/metabolismo , Fragmentos de Peptídeos/biossíntese , Subtilisinas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Furina , Transtornos Heredodegenerativos do Sistema Nervoso/metabolismo , Glicoproteínas de Membrana , Proteínas de Membrana , Camundongos , Microscopia Eletrônica , Células Tumorais Cultivadas , Reino Unido
20.
J Biol Chem ; 274(38): 27249-56, 1999 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-10480944

RESUMO

Previous studies have established that in response to wounding, the expression of amyloid precursor-like protein 2 (APLP2) in the basal cells of migrating corneal epithelium is greatly up-regulated. To further our understanding of the functional significance of APLP2 in wound healing, we have measured the migratory response of transfected Chinese hamster ovary (CHO) cells expressing APLP2 isoforms to a variety of extracellular matrix components including laminin, collagen types I, IV, and VII, fibronectin, and heparan sulfate proteoglycans (HSPGs). CHO cells overexpressing either of two APLP2 variants, differing in chondroitin sulfate (CS) attachment, exhibit a marked increase in chemotaxis toward type IV collagen and fibronectin but not to laminin, collagen types I and VII, and HSPGs. Cells overexpressing APLP2-751 (CS-modified) exhibited a greater migratory response to fibronectin and type IV collagen than their non-CS-attached counterparts (APLP2-763), suggesting that CS modification enhanced APLP2 effects on cell migration. Moreover, in the presence of chondroitin sulfate, transfectants overexpressing APLP2-751 failed to exhibit this enhanced migration toward fibronectin. The APLP2-ECM interactions were also explored by solid phase adhesion assays. While overexpression of APLP2 isoforms moderately enhanced CHO adhesion to laminin, collagen types I and VII, and HSPGs lines, especially those overexpressing APLP2-751, exhibited greatly increased adhesion to type IV collagen and fibronectin. These observations suggest that APLP2 contributes to re-epithelialization during wound healing by supporting epithelial cell adhesion to fibronectin and collagen IV, thus influencing their capacity to migrate over the wound bed. Furthermore, APLP2 interactions with fibronectin and collagen IV appear to be potentiated by the addition of a CS chain to the core proteins.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Movimento Celular , Colágeno/metabolismo , Fibronectinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Células CHO , Adesão Celular , Sulfatos de Condroitina/metabolismo , Cricetinae
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...