Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 37(12): e23311, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37962096

RESUMO

Aggregation of α-synuclein (α-syn) into amyloid is the pathological hallmark of several neurodegenerative disorders, including Parkinson disease, dementia with Lewy bodies, and multiple system atrophy. It is widely accepted that α-syn aggregation is associated with neurodegeneration, although the mechanisms are not yet fully understood. Therefore, the inhibition of α-syn aggregation is a potential therapeutic approach against these diseases. This study used the photocatalyst for α-syn photo-oxygenation, which selectively adds oxygen atoms to fibrils. Our findings demonstrate that photo-oxygenation using this photocatalyst successfully inhibits α-syn aggregation, particularly by reducing its seeding ability. Notably, we also discovered that photo-oxygenation of the histidine at the 50th residue in α-syn aggregates is responsible for the inhibitory effect. These findings indicate that photo-oxygenation of the histidine residue in α-syn is a potential therapeutic strategy for synucleinopathies.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Humanos , alfa-Sinucleína/química , Histidina/análise , Doença de Parkinson/terapia , Doença de Parkinson/patologia , Corpos de Lewy/patologia , Fenômenos Fisiológicos Respiratórios
2.
Nihon Yakurigaku Zasshi ; 158(1): 21-25, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-36596482

RESUMO

The production of amyloid ß peptide (Aß) is an important process relating to the pathogenesis of Alzheimer disease (AD). It is widely known that the sequential cleavage of amyloid precursor protein (APP) by ß- and γ-secretases lead to the production of Aß. However, the precise regulatory mechanism for Aß production remains unclear. We have established a CRISPR-Cas9 based screening system to identify the novel regulators of Aß production. Calcium and integrin-binding protein 1 (CIB1) was identified as a novel potential negative regulator of Aß production. The knockdown and knockout of Cib1 significantly increased Aß levels. In addition, immunoprecipitation showed that CIB1 interacts with the γ-secretase complex but did not alter its enzymatic activity. Moreover, Cib1 disruption specifically reduced the cell-surface localization of the γ-secretase complex. Finally, the single-cell RNA-seq analysis in the human brain demonstrated that early-stage AD patients have lower neuronal CIB1 mRNA levels compared to healthy controls. Taken together, we have shown that CIB1 controls the subcellular localization of γ-secretase, resulting in the regulation of Aß production, suggesting the involvement of CIB1 in the development of AD pathogenesis.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Sistemas CRISPR-Cas , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Neurônios/metabolismo
3.
J Neurosci ; 2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-34099509

RESUMO

Alzheimer disease (AD) is characterized by the extensive deposition of amyloid-ß peptide (Aß) in the brain. Brain Aß level is regulated by a balance between Aß production and clearance. The clearance rate of Aß is decreased in the brains of sporadic AD patients, indicating that the dysregulation of Aß clearance mechanisms affects the pathological process of AD. Astrocytes are among the most abundant cells in the brain and are implicated in the clearance of brain Aß via their regulation of the blood-brain barrier, glymphatic system, and proteolytic degradation. The cellular morphology and activity of astrocytes are modulated by several molecules, including ω3 polyunsaturated fatty acids, such as docosahexaenoic acid, which is one of the most abundant lipids in the brain, via the G protein-coupled receptor GPR120/FFAR4. In this study, we analyzed the role of GPR120 signaling in the Aß-degrading activity of astrocytes. Treatment with the selective antagonist upregulated the matrix metalloproteinase (MMP) inhibitor-sensitive Aß-degrading activity in primary astrocytes. Moreover, the inhibition of GPR120 signaling increased the levels of Mmp2 and Mmp14 mRNAs, and decreased the expression levels of tissue inhibitor of metalloproteinases 3 (Timp3) and Timp4, suggesting that GPR120 negatively regulates the astrocyte-derived MMP network. Finally, the intracerebral injection of GPR120 specific antagonist substantially decreased the levels of Tris-buffered saline-soluble Aß in male AD model mice, and this effect was canceled by the coinjection of an MMP inhibitor. These data indicate that astrocytic GPR120 signaling negatively regulates the Aß degrading activity of MMPs.SIGNIFICANT STATEMENTThe level of amyloid ß (Aß) in the brain is a crucial determinant of the development of Alzheimer disease. Here we found that astrocytes, which are the most abundant cell type in the central nervous system, harbors degrading activity against amyloid ß, which is regulated by GPR120 signaling. GPR120 is involved in the inflammatory response and obesity in peripheral organs. However, the pathophysiological role of GPR120 in Alzheimer disease remains unknown. We found that selective inhibition of GPR120 signaling in astrocytes increased the Aß-degrading activity of matrix metalloproteases. Our results suggest that GPR120 in astrocytes is a novel therapeutic target for the development of anti-Aß therapeutics.

4.
Brain ; 144(6): 1884-1897, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-33851209

RESUMO

Amyloid formation and the deposition of the amyloid-ß peptide are hallmarks of Alzheimer's disease pathogenesis. Immunotherapies using anti-amyloid-ß antibodies have been highlighted as a promising approach for the prevention and treatment of Alzheimer's disease by enhancing microglial clearance of amyloid-ß peptide. However, the efficiency of antibody delivery into the brain is limited, and therefore an alternative strategy to facilitate the clearance of brain amyloid is needed. We previously developed an artificial photo-oxygenation system using a low molecular weight catalytic compound. The photocatalyst specifically attached oxygen atoms to amyloids upon irradiation with light, and successfully reduced the neurotoxicity of aggregated amyloid-ß via inhibition of amyloid formation. However, the therapeutic effect and mode of actions of the photo-oxygenation system in vivo remained unclear. In this study, we demonstrate that photo-oxygenation facilitates the clearance of aggregated amyloid-ß from the brains of living Alzheimer's disease model mice, and enhances the microglial degradation of amyloid-ß peptide. These results suggest that photo-oxygenation may represent a novel anti-amyloid-ß strategy in Alzheimer's disease, which is compatible with immunotherapy.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Compostos de Boro/farmacologia , Encéfalo/efeitos dos fármacos , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Microglia/metabolismo , Fototerapia/métodos , Agregados Proteicos/efeitos dos fármacos
5.
FASEB J ; 34(12): 16383-16396, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33090569

RESUMO

Several lines of evidence suggest that the aggregation and deposition of amyloid-ß peptide (Aß) initiate the pathology of Alzheimer's disease (AD). Recently, a genome-wide association study demonstrated that a single-nucleotide polymorphism proximal to the EPHA4 gene, which encodes a receptor tyrosine kinase, is associated with AD risk. However, the molecular mechanism of EphA4 in the pathogenesis of AD, particularly in Aß production, remains unknown. Here, we performed several pharmacological and biological experiments both in vitro and in vivo and demonstrated that EphA4 is responsible for the regulation of Aß production. Pharmacological inhibition of EphA4 signaling and knockdown of Epha4 led to increased Aß levels accompanied by increased expression of ß-site APP cleaving enzyme 1 (BACE1), which is an enzyme responsible for Aß production. Moreover, EPHA4 overexpression and activation of EphA4 signaling via ephrin ligands decreased Aß levels. In particular, the sterile-alpha motif domain of EphA4 was necessary for the regulation of Aß production. Finally, EPHA4 mRNA levels were significantly reduced in the brains of AD patients, and negatively correlated with BACE1 mRNA levels. Our results indicate a novel mechanism of Aß regulation by EphA4, which is involved in AD pathogenesis.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Neurônios/metabolismo , Receptor EphA4/metabolismo , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia
6.
FASEB J ; 34(6): 7661-7674, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32307772

RESUMO

The aberrant metabolism of amyloid ß peptide (Aß) has been implicated in the etiology of Alzheimer disease (AD). Aß is produced via the sequential cleavage of amyloid precursor protein (APP) by ß- and γ-secretases. However, the precise regulatory mechanism of Aß generation still remains unclear. To gain a better understanding of the molecular mechanism of Aß production, we established a genetic screening method based on the CRISPR/Cas9 system to identify novel regulators of Aß production. We successfully identified calcium and integrin-binding protein 1 (CIB1) as a potential negative regulator of Aß production. The disruption of Cib1 significantly upregulated Aß levels. In addition, immunoprecipitation experiments demonstrated that CIB1 interacts with the γ-secretase complex. Moreover, the disruption of Cib1 specifically reduced the cell-surface localization of mature Nicastrin (Nct), which is a component of the γ-secretase complex, without changing the intrinsic activity of γ-secretase. Finally, we confirmed using the single-cell RNA-seq data in human that CIB1 mRNA level in neuron was decreased in the early stage of AD. Taken together, our results indicate that CIB1 regulates Aß production via controlling the subcellular localization of γ-secretase, suggesting CIB1 is involved in the development of AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Sistemas CRISPR-Cas/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Células HEK293 , Humanos , Camundongos , Neurônios/metabolismo , Ligação Proteica/fisiologia , Transporte Proteico/fisiologia , Sinapsinas/metabolismo , Regulação para Cima/fisiologia
7.
J Biol Chem ; 294(28): 10886-10899, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31138646

RESUMO

A hallmark of Alzheimer's disease (AD) pathology is the appearance of senile plaques, which are composed of ß-amyloid (Aß) peptides. Aß is produced by sequential cleavages of amyloid precursor protein (APP) by ß- and γ-secretases. These cleavages take place in endosomes during intracellular trafficking of APP through the endocytic and recycling pathways. Genome-wide association studies have identified several risk factors for late-onset AD, one of which is CD2-associated protein (CD2AP), an adaptor molecule that regulates membrane trafficking. Although CD2AP's involvement in APP trafficking has recently been reported, how APP trafficking is regulated remains unclear. We sought to address this question by investigating the effect of CD2AP overexpression or knockdown on the intracellular APP distribution and degradation of APP in cultured COS-7 and HEK293 cells. We found that overexpression of CD2AP increases the localization of APP to Rab7-positive late endosomes, and decreases its localization to Rab5-positive early endosomes. CD2AP overexpression accelerated the onset of APP degradation without affecting its degradation rate. Furthermore, nutrient starvation increased the localization of APP to Rab7-positive late endosomes, and CD2AP overexpression stimulated starvation-induced lysosomal APP degradation. Moreover, the effect of CD2AP on the degradation of APP was confirmed by CD2AP overexpression and knockdown in primary cortical neurons from mice. We conclude that CD2AP accelerates the transfer of APP from early to late endosomes. This transfer in localization stimulates APP degradation by reducing the amount of time before degradation initiation. Taken together, these results may explain why impaired CD2AP function is a risk factor for AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Células COS , Chlorocebus aethiops , Proteínas do Citoesqueleto/genética , Endossomos/metabolismo , Estudo de Associação Genômica Ampla , Células HEK293 , Humanos , Lisossomos/metabolismo , Neurônios/metabolismo , Placa Amiloide/metabolismo , Transporte Proteico , Proteólise , Vesículas Transportadoras/metabolismo
8.
J Neurochem ; 147(1): 110-123, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29851073

RESUMO

The aberrant metabolism of amyloid-ß protein (Aß) in the human brain has been implicated in the etiology of Alzheimer disease (AD). γ-Secretase is the enzyme that generates various forms of Aß, such as Aß40 and Aß42, the latter being an aggregation-prone toxic peptide that is involved in the pathogenesis of AD. Recently, we found that clathrin-mediated endocytosis of γ-secretase affects the production and deposition of Aß42 in vivo, suggesting that the membrane trafficking of γ-secretase affects its enzymatic activity. However, the detailed intracellular trafficking pathway of γ-secretase and its contribution to Aß42 generation remain unclear. Here, we show that Retro-2, which inhibits the retrograde transport, elevated the Aß42-generating activity both in cultured cells and mice brain. However, the result of in vitro γ-secretase assay using a recombinant substrate suggested that Retro-2 did not elevate the intrinsic Aß42-production activity of γ-secretase. Immunocytochemistry and cell-surface biotinylation experiments revealed that γ-secretase is recycled via the endosome-to-trans-Golgi network transport. In addition, γ-secretase is retrogradely transported by syntaxin 5/6, known as targets of Retro-2, independent pathway. Conversely, TPT-260, which enhances the trafficking function of retromers, lowered Aß42 levels and the Aß42/(Aß40 + Aß42) ratio in secreted Aß from cultured cells. Our results strongly suggest that the endosome-to-trans-Golgi network trafficking of γ-secretase regulates its Aß42 production activity. Modulation of this trafficking pathway might be a potential target for the development of Aß42-lowering AD therapeutics. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/biossíntese , Endossomos/metabolismo , Fragmentos de Peptídeos/biossíntese , Rede trans-Golgi/metabolismo , Animais , Benzamidas/farmacologia , Biotinilação , Química Encefálica/efeitos dos fármacos , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Proteínas Qa-SNARE/metabolismo , Tiofenos/farmacologia
9.
Lab Invest ; 83(6): 797-812, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12808115

RESUMO

A membrane invasion culture system was used to study the ability of EBV to enhance invasion and migration of nasopharyngeal carcinoma (NPC) cells. Semi-reverse transcriptase-PCR analysis of matrix proteinases and angiogenic factors from EBV-infected, or EBV-positive (EBV+), cells demonstrated different degrees of elevated gene expression. In our animal model, EBV+ tumors grew faster and larger than EBV-free, or EBV-negative (EBV-), tumors and also had clonal EBV terminal repeat sequences. Double-localization of EBV and certain host proteins in EBV+ tumors and biopsy specimens demonstrated that EBV up-regulates host genes only in cells that express those genes but not in cells that do not express them. Double-localization of EBV and host genes in NPC biopsy specimens all showed EBV- tumor cells expressing those host genes. Our data strongly suggest that EBV infection enhances progression of NPC tumor growth. They do not rule out a role for EBV infection in the induction and early promotion of NPC development. Unidentified factors may also enhance NPC tumor growth independent of the effects of EBV.


Assuntos
Herpesvirus Humano 4/isolamento & purificação , Neoplasias Nasofaríngeas/virologia , Animais , Biópsia , Carcinoma/patologia , Carcinoma/virologia , Receptores ErbB/análise , Substâncias de Crescimento/análise , Herpesvirus Humano 4/genética , Humanos , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Neoplasias Nasofaríngeas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...