Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 39(17): 3188-3203, 2019 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-30804097

RESUMO

Secreted amyloid precursor protein-alpha (sAPPα) has growth factor-like properties and can modulate long-term potentiation (LTP) and memory. Here, we demonstrate that exposure to sAPPα converts short-lasting LTP into protein-synthesis-dependent late LTP in hippocampal slices from male rats. sAPPß had no discernable effect. We hypothesized that sAPPα facilitated LTP via regulated glutamate receptor trafficking and de novo protein synthesis. We found using a linear mixed model that sAPPα stimulated trafficking of GluA2-lacking AMPARs, as well as NMDARs to the extrasynaptic cell surface, in a calcium/calmodulin-dependent kinase II and protein kinase G-dependent manner. Both cell surface receptor accumulation and LTP facilitation were present even after sAPPα washout and inhibition of receptor trafficking or protein synthesis prevented all these effects. Direct visualization of newly synthesized proteins (FUNCAT-PLA) confirmed the ability of sAPPα to stimulate de novo protein synthesis and revealed GluA1 as one of the upregulated proteins. Therefore, sAPPα generates a coordinated synthesis and trafficking of glutamate receptors to the cell surface that facilitate LTP.SIGNIFICANCE STATEMENT Secreted amyloid precursor protein-alpha (sAPPα) is a neurotrophic and neuroprotective protein that can promote synaptic plasticity and memory, yet the molecular mechanisms underlying these effects are still not well understood. Here, we show that sAPPα facilitates long-term potentiation (LTP) in a concentration-dependent fashion through cellular processes involving de novo protein synthesis and trafficking of both GluA2-lacking AMPARs and NMDARs to the extrasynaptic cell surface. sAPPα also enhances GluA1, but not GluA2, synthesis. The trafficking effects, along with the LTP facilitation, persist after sAPPα washout, revealing a metaplastic capability of exogenous sAPPα administration. sAPPα thus facilitates LTP through coordinated activation of protein synthesis and trafficking of glutamate receptors to the cell surface, where they are positioned for priming LTP.


Assuntos
Precursor de Proteína beta-Amiloide/farmacologia , Hipocampo/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Receptores de Glutamato/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Biossíntese de Proteínas/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley
2.
Neurobiol Learn Mem ; 138: 291-299, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27521248

RESUMO

Secreted amyloid precursor protein-α (sAPPα) is a neurotrophic and neuroprotective molecule which can enhance learning and synaptic plasticity. Aging is associated with memory decline and impaired long-term potentiation (LTP). SAPPα therefore has potential as a nootropic agent which could be used to offset age-related cognitive decline. In this study we investigated the effects of sAPPα on spatial memory tasks and LTP in aged and young Long-Evans rats. Two hippocampus-dependent tasks were employed to measure spatial memory that is susceptible to impairments during aging. Aged rats showed a mild deficit in the novel object location task, but memory was significantly enhanced by bilateral intrahippocampal injections of sAPPα. There was no effect on the performance of young animals. In the watermaze task, however, sAPPα did not alleviate age-related decline in spatial memory. In subsequent electrophysiological experiments, LTP was impaired in slices from aged animals, but plasticity was rescued in a concentration-dependent manner by exogenous sAPPα administration. In contrast, LTP was impaired in young animals by sAPPα. Overall, these data support the hypothesis that sAPPα has therapeutic potential as a treatment for age-related cognitive decline.


Assuntos
Envelhecimento/fisiologia , Precursor de Proteína beta-Amiloide/farmacologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Ratos , Ratos Long-Evans , Memória Espacial/fisiologia
3.
Front Mol Neurosci ; 14: 660208, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33867938

RESUMO

Regulation of AMPA receptor expression by neuronal activity and neuromodulators is critical to the expression of both long-term potentiation (LTP) and memory. In particular, Ca2+-permeable AMPARs (CP-AMPAR) play a unique role in these processes due to their transient, activity-regulated expression at synapses. Secreted amyloid precursor protein-alpha (sAPPα), a metabolite of the parent amyloid precursor protein (APP) has been previously shown to enhance hippocampal LTP as well as memory formation in both normal animals and in Alzheimer's disease models. In earlier work we showed that sAPPα promotes trafficking of GluA1-containing AMPARs to the cell surface and specifically enhances synthesis of GluA1. To date it is not known whether de novo synthesized GluA1 form CP-AMPARs or how they contribute to sAPPα-mediated plasticity. Here, using fluorescent non-canonical amino acid tagging-proximity ligation assay (FUNCAT-PLA), we show that brief treatment of primary rat hippocampal neurons with sAPPα (1 nM, 30 min) rapidly enhanced the cell-surface expression of de novo GluA1 homomers and reduced levels of de novo GluA2, as well as extant GluA2/3-AMPARs. The de novo GluA1-containing AMPARs were localized to extrasynaptic sites and later internalized by sAPPα-driven expression of the activity-regulated cytoskeletal-associated protein, Arc. Interestingly, longer exposure to sAPPα increased synaptic levels of GluA1/2 AMPARs. Moreover, the sAPPα-mediated enhancement of LTP in area CA1 of acute hippocampal slices was dependent on CP-AMPARs. Together, these findings show that sAPPα engages mechanisms which specifically enhance the synthesis and cell-surface expression of GluA1 homomers, underpinning the sAPPα-driven enhancement of synaptic plasticity in the hippocampus.

4.
Neuropharmacology ; 157: 107670, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31202608

RESUMO

Processing of the amyloid precursor protein by alternative secretases results in ectodomain shedding of either secreted amyloid precursor protein-α (sAPPα) or its counterpart secreted amyloid precursor protein-ß (sAPPß). Although sAPPα contains only 16 additional amino acids at its C-terminus compared to sAPPß, it displays significantly greater potency in neuroprotection, neurotrophism and enhancement of long-term potentiation (LTP). In the current study, this 16 amino acid peptide sequence (CTα16) was characterised for its ability to replicate the synaptic plasticity-enhancing properties of sAPPα. An N-acetylated version of CTα16 produced concentration-dependent increases in the induction and persistence of LTP at Schaffer collateral/commissural synapses in area CA1 of young adult rat hippocampal slices. A scrambled peptide had no effect. CTα16 significantly enhanced de novo protein synthesis, and correspondingly its enhancement of LTP was blocked by the protein synthesis inhibitor cycloheximide, as well as by the α7-nicotinic receptor blocker α-bungarotoxin. The impaired LTP of 14-16 month old APPswe/PS1dE9 transgenic mice, a mouse model of Alzheimer's disease, was completely restored to the wild-type level by CTα16. These results indicate that the CTα16 peptide fragment of sAPPα mimics the larger protein's functionality with respect to LTP, stimulation of protein synthesis and activation of α7-nAChRs, and thus like sAPPα may have potential as a therapeutic agent against the plasticity and cognitive deficits observed in AD and other neurological disorders.


Assuntos
Doença de Alzheimer/fisiopatologia , Potenciação de Longa Duração/efeitos dos fármacos , Doença de Alzheimer/genética , Animais , Bungarotoxinas/farmacologia , Região CA1 Hipocampal/fisiologia , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Ratos
5.
Exp Neurol ; 303: 29-37, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29410317

RESUMO

Secreted amyloid precursor protein alpha (sAPPα) is a potent neurotrophin in the CNS but a dedicated receptor has not been found. However, protein interactions involving amyloid beta (Aß), a peptide cleaved from the same parent peptide as sAPPα, indicate that insulin receptors (IRs) could be a target of amyloid peptides. In this study, in vitro analysis of cortical neuronal cultures revealed that exogenous sAPPα increased IR phosphorylation in the absence of insulin. Furthermore, in an APP overexpressing mouse model, sAPPα bound IRs in the cortex with significantly greater binding in hypoinsulinemic animals. To further examine the effects of sAPPα on the diabetic brain, we next rendered sAPPα overexpressing mice insulin depleted and found that sAPPα blocked aberrant tau phosphorylation (T231) in cortical tissue after 16 weeks diabetes. sAPPα overexpression also prevented hyperphosphorylation of AKT/GSK3 and activation of the unfolded protein response (UPR). In total, these data show sAPPα binds and activates neuronal IRs and that sAPPα has a protective effect on diabetic brain tissue.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Encefalopatias/etiologia , Encefalopatias/prevenção & controle , Diabetes Mellitus Experimental/complicações , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptor de Insulina/metabolismo , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/farmacologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Hemoglobinas Glicadas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Fosforilação/fisiologia , Ligação Proteica/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Resposta a Proteínas não Dobradas/genética , Proteínas tau/metabolismo
6.
J Mol Neurosci ; 66(1): 1-9, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30062438

RESUMO

Immunotherapy for Alzheimer's disease (AD) remains promising in the improvement of cognition and memory via the clearing of amyloid-ß protein (Aß) in the AD brain, despite some side effects. Our previous studies demonstrated that the 31-35 sequence of the Aß molecule was the shortest active center and that polyclonal anti-Aß31-35 antibody reduced neuronal apoptosis and cognitive impairments induced with acute Aß application. The present study designed a novel single-chain variable fragment (scFv) monoclonal anti-Aß31-35 antibody (scFv17) that specifically recognized extracellular Aß and observed protective effects of scFv17 on pathological impairments in APP/PS1 transgenic mice. We also investigated its cellular and molecular mechanisms and found that scFv17 and 6E10 (a positive control) exhibited similar Aß-clearing ability and that scFv17 produced a stronger effect in clearing Aß oligomers than 6E10. scFv17, but not 6E10, enhanced anti-inflammatory responses with significant increases in IL-10 and TGF-ß. 6E10 decreased BACE1 levels, and scFv17 significantly increased the level of secreted amyloid precursor protein-α (sAPPα), which is an important physiological neurotrophin from APP generated by α-secretase. 6E10 and scFv17, especially the latter, dramatically down-regulated the expression of neprilysin, which is an enzyme expressed in proportion to Aß concentration. Therefore, the present study demonstrated that the novel monoclonal anti-Aß31-35 antibody scFv17 effectively reduced pathological impairments in APP/PS1 transgenic mice via modulation of inflammatory cytokines and Aß-related enzymes, which supports scFv17 as a new alternative in the current immunotherapy of AD.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Imunoterapia/métodos , Fragmentos de Peptídeos/imunologia , Anticorpos de Cadeia Única/uso terapêutico , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neprilisina/genética , Neprilisina/metabolismo , Fragmentos de Peptídeos/genética , Presenilina-1/genética , Anticorpos de Cadeia Única/imunologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
7.
Neuromolecular Med ; 19(2-3): 300-308, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28612181

RESUMO

Previous studies have demonstrated that the low-density lipoprotein receptor-related protein-1 (LRP1) plays conflicting roles in Alzheimer's disease (AD) pathogenesis, clearing ß-amyloid (Aß) from the brain while also enhancing APP endocytosis and resultant amyloidogenic processing. We have recently discovered that co-expression of mutant LRP1 C-terminal domain (LRP1-CT C4408R) with Swedish mutant amyloid precursor protein (APPswe) in Chinese hamster ovary (CHO) cells decreases Aß production, while also increasing sAPPα and APP α-C-terminal fragment (α-CTF), compared with CHO cells expressing APPswe alone. Surprisingly, the location of this mutation on LRP1 corresponded with the α-secretase cleavage site of APP. Further experimentation confirmed that in CHO cells expressing APPswe or wild-type APP (APPwt), co-expression of LRP1-CT C4408R decreases Aß and increases sAPPα and α-CTF compared with co-expression of wild-type LRP1-CT. In addition, LRP1-CT C4408R enhanced the unglycosylated form of LRP1-CT and reduced APP endocytosis as determined by flow cytometry. This finding identifies a point mutation in LRP1 which slows LRP1-CT-mediated APP endocytosis and amyloidogenic processing, while enhancing APP α-secretase cleavage, thus demonstrating a potential novel target for slowing AD pathogenesis.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Mutação de Sentido Incorreto , Mutação Puntual , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , Endocitose , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Domínios Proteicos , Proteínas Recombinantes/metabolismo
8.
Int J Clin Exp Med ; 1(4): 338-44, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19079679

RESUMO

Autism is a pervasive developmental disorder characterized by impairments in socialization and communication. There is currently no single molecular marker or laboratory tool capable of diagnosing autism at an early age. The purpose of this study is to explore the plausible use of peripheral biomarkers in the early diagnosis of autism via a sensitive ELISA. Here, we measured plasma secreted amyloid precursor protein alpha (sAPP-alpha) levels in autistic and aged-matched control blood samples and found a significantly increased level of sAPP-alpha in 60% of the known autistic children. We then tested 150 human umbilical cord blood (HUCB) samples and found significantly elevated levels of plasma sAPP-alpha in 10 of 150 samples. As an additional confirmatory measure, we performed Western blot analysis on these samples which consistently showed increased sAPP-alpha levels in autistic children and 10 of 150 HUCB samples; suggesting a group of autistic patients which could be identified in early childhood by levels of sAPP-alpha. While there is need for further studies of this concept, the measurement of sAPP-alpha levels in serum and human umbilical cord blood by ELISA is a potential tool for early diagnosis of autism.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA