Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
FASEB J ; 37(12): e23311, 2023 12.
Article in English | MEDLINE | ID: mdl-37962096

ABSTRACT

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.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/chemistry , Histidine/analysis , Parkinson Disease/therapy , Parkinson Disease/pathology , Lewy Bodies/pathology , Respiratory Physiological Phenomena
2.
Nihon Yakurigaku Zasshi ; 158(1): 21-25, 2023.
Article in Japanese | MEDLINE | ID: mdl-36596482

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , CRISPR-Cas Systems , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Neurons/metabolism
3.
J Neurosci ; 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34099509

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-33851209

ABSTRACT

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.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Boron Compounds/pharmacology , Brain/drug effects , Animals , Brain/pathology , Disease Models, Animal , Humans , Mice , Microglia/metabolism , Phototherapy/methods , Protein Aggregates/drug effects
5.
FASEB J ; 34(12): 16383-16396, 2020 12.
Article in English | MEDLINE | ID: mdl-33090569

ABSTRACT

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.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Neurons/metabolism , Receptor, EphA4/metabolism , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Cell Line , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Signal Transduction/physiology
6.
FASEB J ; 34(6): 7661-7674, 2020 06.
Article in English | MEDLINE | ID: mdl-32307772

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , CRISPR-Cas Systems/physiology , Calcium-Binding Proteins/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , HEK293 Cells , Humans , Mice , Neurons/metabolism , Protein Binding/physiology , Protein Transport/physiology , Synapsins/metabolism , Up-Regulation/physiology
7.
J Biol Chem ; 294(28): 10886-10899, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31138646

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Protein Precursor/metabolism , Cytoskeletal Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , COS Cells , Chlorocebus aethiops , Cytoskeletal Proteins/genetics , Endosomes/metabolism , Genome-Wide Association Study , HEK293 Cells , Humans , Lysosomes/metabolism , Neurons/metabolism , Plaque, Amyloid/metabolism , Protein Transport , Proteolysis , Transport Vesicles/metabolism
8.
J Neurochem ; 147(1): 110-123, 2018 10.
Article in English | MEDLINE | ID: mdl-29851073

ABSTRACT

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/.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Endosomes/metabolism , Peptide Fragments/biosynthesis , trans-Golgi Network/metabolism , Animals , Benzamides/pharmacology , Biotinylation , Brain Chemistry/drug effects , Cells, Cultured , Female , Gene Expression Regulation , Humans , Metabolic Networks and Pathways/drug effects , Mice , Qa-SNARE Proteins/metabolism , Thiophenes/pharmacology
9.
Lab Invest ; 83(6): 797-812, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12808115

ABSTRACT

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.


Subject(s)
Herpesvirus 4, Human/isolation & purification , Nasopharyngeal Neoplasms/virology , Animals , Biopsy , Carcinoma/pathology , Carcinoma/virology , ErbB Receptors/analysis , Growth Substances/analysis , Herpesvirus 4, Human/genetics , Humans , Male , Mice , Mice, SCID , Middle Aged , Nasopharyngeal Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...