17 results on '"Zhang, Zhentao"'
Search Results
2. Tau in the Pathophysiology of Parkinson’s Disease
- Author
-
Pan, Lina, Meng, Lanxia, He, Mingyang, and Zhang, Zhentao
- Published
- 2021
- Full Text
- View/download PDF
3. Exosomes from patients with Parkinson’s disease are pathological in mice
- Author
-
Han, Chao, Xiong, Nian, Guo, Xingfang, Huang, Jinsha, Ma, Kai, Liu, Ling, Xia, Yun, Shen, Yan, Li, Jie, Jiang, Haiyang, Wang, Luxi, Guo, Shiyi, Xu, Xiaoyun, Zhang, Guoxin, Liu, Jingyu, Cao, Xuebing, Zhang, Zhentao, Lin, Zhicheng, and Wang, Tao
- Published
- 2019
- Full Text
- View/download PDF
4. 27‐Hydroxycholesterol Drives the Spread of α‐Synuclein Pathology in Parkinson's Disease.
- Author
-
Dai, Lijun, Wang, Jiannan, Zhang, Xingyu, Yan, Mingmin, Zhou, Lingyan, Zhang, Guoxin, Meng, Lanxia, Chen, Liam, Cao, Xuebing, Zhang, Zhaohui, Wang, Gaohua, and Zhang, Zhentao
- Abstract
Background: The accumulation and aggregation of α‐synuclein (α‐Syn) are characteristic of Parkinson's disease (PD). Epidemiological evidence indicates that hyperlipidemia is associated with an increased risk of PD. The levels of 27‐hydroxycholesterol (27‐OHC), a cholesterol oxidation derivative, are increased in the brain and cerebrospinal fluid of patients with PD. However, whether 27‐OHC plays a role in α‐Syn aggregation and propagation remains elusive. Objective: The aim of this study was to determine whether 27‐OHC regulates α‐Syn aggregation and propagation. Methods: Purified recombinant α‐Syn, neuronal cultures, and α‐Syn fibril‐injected mouse model of PD were treated with 27‐OHC. In addition, CYP27A1 knockout mice were used to investigate the effect of lowering 27‐OHC on α‐Syn pathology in vivo. Results: 27‐OHC accelerates the aggregation of α‐Syn and enhances the seeding activity of α‐Syn fibrils. Furthermore, the 27‐OHC‐modified α‐Syn fibrils localize to the mitochondria and induce mitochondrial dysfunction and neurotoxicity. Injection of 27‐OHC‐modified α‐Syn fibrils induces enhanced spread of α‐Syn pathology and dopaminergic neurodegeneration compared with pure α‐Syn fibrils. Similarly, subcutaneous administration of 27‐OHC facilitates the seeding of α‐Syn pathology. Genetic deletion of cytochrome P450 27A1 (CYP27A1), the enzyme that converts cholesterol to 27‐OHC, ameliorates the spread of pathologic α‐Syn, degeneration of the nigrostriatal dopaminergic pathway, and motor impairments. These results indicate that the cholesterol metabolite 27‐OHC plays an important role in the pathogenesis of PD. Conclusions: 27‐OHC promotes the aggregation and spread of α‐Syn. Strategies aimed at inhibiting the CYP27A1‐27‐OHC axis may hold promise as a disease‐modifying therapy to halt the progression of α‐Syn pathology in PD. © 2023 International Parkinson and Movement Disorder Society. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. α‐Synuclein induces deficiency in clathrin‐mediated endocytosis through inhibiting synaptojanin1 expression.
- Author
-
Song, Dong‐Yan, Yuan, Lin, Cui, Na, Feng, Cong, Meng, Lanxia, Wang, Xin‐He, Xiang, Man, Liu, Di, Wang, Chun, Zhang, Zhentao, Li, Jia‐Yi, and Li, Wen
- Subjects
ENDOCYTOSIS ,COATED vesicles ,ALPHA-synuclein ,AXONAL transport ,SYNAPTIC vesicles ,PARKINSON'S disease ,NEUROLOGICAL disorders - Abstract
Parkinson's disease (PD) is an age‐related chronic neurological disorder, mainly characterized by the pathological feature of α‐synuclein (α‐syn) aggregation, with the exact disease pathogenesis unclear. During the onset and progression of PD, synaptic dysfunction, including dysregulation of axonal transport, impaired exocytosis, and endocytosis are identified as crucial events of PD pathogenesis. It has been reported that over‐expression of α‐syn impairs clathrin‐mediated endocytosis (CME) in the synapses. However, the underlying mechanisms still needs to be explored. In this study, we investigated the molecular events underlying the synaptic dysfunction caused by over‐expression of wild‐type human α‐syn and its mutant form, involving series of proteins participating in CME. We found that excessive human α‐syn causes impaired fission and uncoating of clathrin‐coated vesicles during synaptic vesicle recycling, leading to reduced clustering of synaptic vesicles near the active zone and increased size of plasma membrane and number of endocytic intermediates. Furthermore, over‐expressed human α‐syn induced changes of CME‐associated proteins, among which synaptojanin1 (SYNJ1) showed significant reduction in various brain regions. Over‐expression of SYNJ1 in primary hippocampal neurons from α‐syn transgenic mice recovered the synaptic vesicle density, clustering and endocytosis. Using fluorescence‐conjugated transferrin, we demonstrated that SYNJ1 re‐boosted the CME activity by restoring the phosphatidylinositol‐4,5‐bisphosphate homeostasis. Our data suggested that over‐expression of α‐syn disrupts synaptic function through interfering with vesicle recycling, which could be alleviated by re‐availing of SYNJ1. Our study unrevealed a molecular mechanism of the synaptic dysfunction in PD pathogenesis and provided a potential therapeutic target for treating PD. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Fine Particulate Matter Triggers α-Synuclein Fibrillization and Parkinson-like Neurodegeneration.
- Author
-
Yuan, Xin, Yang, Yingxu, Liu, Chaoyang, Tian, Ye, Xia, Danhao, Liu, Zehua, Pan, Lina, Xiong, Min, Xiong, Jing, Meng, Lanxia, Zhang, Zhaohui, Ye, Keqiang, Jiang, Haiqiang, and Zhang, Zhentao
- Subjects
PARTICULATE matter ,NERVE tissue proteins ,PARKINSON'S disease ,SYNUCLEINS ,RESEARCH funding ,ANIMALS ,MICE - Abstract
Background: The deposition of α-synuclein (α-Syn) in the brain is the pathological hallmark of Parkinson's disease (PD). Epidemiological data indicate that exposure to fine particulate matter (≤2.5 μm in aerodynamic diameter [PM2.5]) is associated with an increased risk for PD.Objective: The aim of this study is to investigate whether PM2.5 has a direct effect on α-Syn pathology and how it drives the risk for PD.Methods: PM2.5 was added into α-Syn monomers and different cell models to test whether PM2.5 can promote the fibrillization and aggregation of α-Syn. α-Syn A53T transgenic mice and α-Syn knockout mice were used to investigate the effects of PM2.5 on PD-like pathology.Results: PM2.5 triggers the fibrillization of α-Syn and promotes the formation of α-Syn fibrils with enhanced seeding activity and neurotoxicity. PM2.5 also induces mitochondrial dysfunction and oxidative stress. Intrastriatal injection or intranasal administration of PM2.5 exacerbates α-Syn pathology and dopaminergic neuronal degeneration in α-Syn A53T transgenic mice. The detrimental effect of PM2.5 was attenuated in α-Syn knockout mice.Conclusions: Our results identify that PM2.5 exposure could promote the α-Syn pathology, providing mechanistic insights into how PM2.5 increases the risk for PD. © 2022 International Parkinson and Movement Disorder Society. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
7. Silica Nanoparticles Promote α-Synuclein Aggregation and Parkinson's Disease Pathology.
- Author
-
Yuan, Xin, Yang, Yingxu, Xia, Danhao, Meng, Lanxia, He, Mingyang, Liu, Chaoyang, and Zhang, Zhentao
- Subjects
PARKINSON'S disease ,SILICA nanoparticles ,PATHOLOGY ,DRUG delivery systems ,CENTRAL nervous system - Abstract
Silica nanoparticles (SiO
2 NPs) are increasingly investigated for their potential in drug delivery systems. However, the neurotoxicity of SiO2 NPs remains to be fully clarified. Previously SiO2 NPs have been reported to be detected in the central nervous system, especially in the dopaminergic neurons which are deeply involved in Parkinson's disease (PD). In this article, we characterized the effects of SiO2 NPs on inducing PD-like pathology both in vitro and in vivo. Results showed that SiO2 NPs promote more severe hyperphosphorylation and aggregation of α-synuclein, mitochondria impairment, oxidative stress, autophagy dysfunction, and neuronal apoptosis in the α-Syn A53T transgenic mice intranasally administrated with SiO2 NPs compared with the control group. Our findings provide new evidence supporting that SiO2 NPs exposure might have a strong capability of promoting the initiation and development of PD. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
8. Transgenic Mice Expressing Human α-Synuclein 1-103 Fragment as a Novel Model of Parkinson's Disease.
- Author
-
Tian, Ye, He, Mingyang, Pan, Lina, Yuan, Xin, Xiong, Min, Meng, Lanxia, Yao, Zhaohui, Yu, Zhui, Ye, Keqiang, and Zhang, Zhentao
- Subjects
PARKINSON'S disease ,TRANSGENIC mice ,CENTRAL nervous system ,DOPAMINERGIC neurons ,SUBSTANTIA nigra ,DEFECATION - Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders. However, its cellular and molecular mechanisms still wrap in the mist. This is partially caused by the absence of appropriate animal models mimicking sporadic PD that constitutes the majority of cases. Previously, we reported that a cysteine protease, asparagine endopeptidase (AEP), is activated in an age-dependent manner, and cleaves α-synuclein in the brain of sporadic PD patients. The AEP-derived α-synuclein 1-103 fragment is required for the pathogenesis of PD. Thus, we designed and characterized a novel transgenic mouse line expressing α-synuclein 1-103 (designated N103 mice). This model shows an abundant accumulation of pathological α-synuclein in the central nervous system, loss of dopaminergic neurons in the substantia nigra, and progressive striatal synaptic degeneration. The N103 mice also manifest age-dependent PD-like behavioral impairments. Notably, the mice show weight loss and constipation, which are the common non-motor symptoms in PD. The RNA-sequencing analysis found that the transcriptomics pattern was extensively altered in N103 mice. In conclusion, the N103 mouse line, as a brand-new tool, might provide new insights into PD research. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
9. Lovastatin Alleviates α-Synuclein Aggregation and Phosphorylation in Cellular Models of Synucleinopathy.
- Author
-
Dai, Lijun, Wang, Jiannan, He, Mingyang, Xiong, Min, Tian, Ye, Liu, Chaoyang, and Zhang, Zhentao
- Subjects
PROTEIN kinase CK2 ,LOVASTATIN ,PARKINSON'S disease ,PHOSPHORYLATION ,HISTONE acetylation - Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Pathologically, it is characterized by the aberrant aggregation of α-synuclein (α-syn) in neurons. Clinical evidence shows that patients with hypercholesterolemia are more likely to get PD, while lovastatin users have a lower risk of suffering from it. In this study, we investigated the effects of lovastatin on the aggregation and phosphorylation of α-syn in vitro. Our results demonstrate that α-syn preformed fibrils induce the phosphorylation and aggregation of α-syn in HEK293 cells stably transfected with α-syn-GFP and SH-SY5Y cells as well, which could be attenuated by in a concentration-dependent manner. Besides, lovastatin inhibited oxidative stress, histone acetylation, and the activation of casein kinase 2 (CK2). Collectively, lovastatin alleviates α-syn aggregation and phosphorylation in cellular models of synucleinopathy, indicating its potential value of being adopted in the management of PD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Reactive microglia enhance the transmission of exosomal α-synuclein via toll-like receptor 2.
- Author
-
Xia, Yun, Zhang, Guoxin, Kou, Liang, Yin, Sijia, Han, Chao, Hu, Junjie, Wan, Fang, Sun, Yadi, Wu, Jiawei, Li, Yunna, Huang, Jinsha, Xiong, Nian, Zhang, Zhentao, and Wang, Tao
- Subjects
TOLL-like receptors ,MICROGLIA ,PARKINSON'S disease ,SUBSTANTIA nigra ,PHAGOCYTOSIS ,CELL metabolism ,BRAIN metabolism ,BRAIN ,RESEARCH ,NERVE tissue proteins ,EXOSOMES ,ANIMAL experimentation ,RESEARCH methodology ,CELL receptors ,MEDICAL cooperation ,EVALUATION research ,RATS ,COMPARATIVE studies ,CELLS ,MICE - Abstract
Increasing evidence suggests that microglial activation is strongly linked to the initiation and progression of Parkinson's disease. Cell-to-cell propagation of α-synuclein pathology is a highlighted feature of Parkinson's disease, and the focus of such research has been primarily on neurons. However, recent studies as well as the data contained herein suggest that microglia, the primary phagocytes in the brain, play a direct role in the spread of α-synuclein pathology. Recent data revealed that plasma exosomes derived from Parkinson's disease patients (PD-EXO) carry pathological α-synuclein and target microglia preferentially. Hence, PD-EXO are likely a key tool for investigating the role of microglia in α-synuclein transmission. We showed that intrastriatal injection of PD-EXO resulted in the propagation of exosomal α-synuclein from microglia to neurons following microglia activation. Toll-like receptor 2 (TLR2) in microglia was activated by exosomal α-synuclein and acted as a crucial mediator of PD-EXO-induced microglial activation. Additionally, partial microglia depletion resulted in a significant decrease of exogenous α-synuclein in the substantia nigra. Furthermore, exosomal α-synuclein internalization was initiated by binding to TLR2 of microglia. Excessive α-synuclein phagocytosis may induce the inflammatory responses of microglia and provide the seed for microglia-to-neuron transmission. Consistently, TLR2 silencing in microglia mitigated α-synuclein pathology in vivo. Overall, the present data support the idea that the interaction of exosomal α-synuclein and microglial TLR2 contribute to excessive α-synuclein phagocytosis and microglial activation, which lead to the further propagation and spread of α-synuclein pathology, thereby highlighting the pivotal roles of reactive microglia in α-synuclein transmission. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Dysfunction of Synaptic Vesicle Endocytosis in Parkinson's Disease.
- Author
-
Zou, Li, Tian, Ye, and Zhang, Zhentao
- Subjects
PARKINSON'S disease ,SYNAPTIC vesicles ,ENDOCYTOSIS ,ALZHEIMER'S disease ,DOPAMINERGIC neurons - Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease. It is a chronic and progressive disorder estimated to affect at least 4 million people worldwide. Although the etiology of PD remains unclear, it has been found that the dysfunction of synaptic vesicle endocytosis (SVE) in neural terminal happens before the loss of dopaminergic neurons. Recently, accumulating evidence reveals that the PD-linked synaptic genes, including DNAJC6 , SYNJ1 , and SH3GL2 , significantly contribute to the disruptions of SVE, which is vital for the pathogenesis of PD. In addition, the proteins encoded by other PD-associated genes such as SNCA , LRRK2 , PRKN , and DJ-1 also play key roles in the regulation of SVE. Here we present the facts about SVE-related genes and discussed their potential relevance to the pathogenesis of PD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. HMGB1 Mediates Autophagy Dysfunction via Perturbing Beclin1-Vps34 Complex in Dopaminergic Cell Model
- Author
-
Huang, Jinsha, Yang, Jiaolong, Shen, Yan, Jiang, Haiyang, Han, Chao, Zhang, Guoxin, Liu, Ling, Xu, Xiaoyun, Li, Jie, Lin, Zhicheng, Xiong, Nian, Zhang, Zhentao, Xiong, Jing, and Wang, Tao
- Subjects
HMGB1 ,autophagy ,α-synuclein ,Beclin1 ,Vps34 ,Parkinson’s disease - Abstract
Parkinson’s disease (PD), a progressive neurodegenerative disorder, is characterized by irreversible dopaminergic neuron loss and intra-neuronal α-synuclein aggregation. High mobility group box 1 (HMGB1) has been proven to be involved in autophagy dysfunction induced by α-synuclein accumulation, and the Beclin1-vacuolar protein sorting 34 (Vps34) complex is of great importance to the initiation of autophagy. Nevertheless, the concrete interaction mechanism between HMGB1, α-synuclein and autophagy remains elusive, especially in the context of PD. Here in this study, we investigated the interaction between HMGB1 and α-synuclein in rotenone-induced PD cell models and their roles in autophagy flux. Results revealed elevated expression and cytosolic translocation of endogenous HMGB1 upon rotenone exposure. Besides, HMGB1 was found to be able to co-localize and interact with α-synuclein. Moreover, it had also been proven that HMGB1 could aggravate α-synuclein aggregation induced autophagy dysfunction via perturbing Beclin1-Vps34 complex formation. Based on these findings, we propose that HMGB1 is involved in rotenone-induced dopaminergic cell death via interacting with α-synuclein, perturbing the autophagy process, aggravating protein aggregation and finally propelling dopaminergic neurons to move from morbidity to mortality.
- Published
- 2017
- Full Text
- View/download PDF
13. Environmental factors in Parkinson's disease: New insights into the molecular mechanisms.
- Author
-
Yuan, Xin, Tian, Ye, Liu, Chaoyang, and Zhang, Zhentao
- Subjects
- *
PARKINSON'S disease , *DYSAUTONOMIA , *SUBSTANTIA nigra , *ENVIRONMENTAL risk , *DOPAMINERGIC neurons , *NEURODEGENERATION - Abstract
• Environmental factors are important risk factors for PD. • We summarized the toxic mechanisms that different environmental factors played. • PM2.5 can increase the risk of PD, but the molecular mechanisms remain to be elucidated. Parkinson's disease is a chronic, progressive neurodegenerative disorder affecting 2–3% of the population ≥65 years. It has long been characterized by motor impairment, autonomic dysfunction, and psychological and cognitive changes. The pathological hallmarks are intracellular inclusions containing α-synuclein aggregates and the loss of dopaminergic neurons in the substantia nigra. Parkinson's disease is thought to be caused by a combination of various pathogenic factors, including genetic factors, environmental factors, and lifestyles. Although much research has focused on the genetic causes of PD, environmental risk factors also play a crucial role in the development of the disease. Here, we summarize the environmental risk factors that may increase the occurrence of PD, as well as the underlying molecular mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Cofilin 1 promotes the aggregation and cell-to-cell transmission of α-synuclein in Parkinson's disease.
- Author
-
Yan, Mingmin, Meng, Lanxia, Dai, Lijun, Zhang, Xingyu, Chen, Guiqin, Zheng, Yongfa, Zha, Yunhong, Zeng, Yan, and Zhang, Zhentao
- Subjects
- *
PARKINSON'S disease , *PATHOLOGY - Abstract
The histopathological hallmark of Parkinson's disease (PD) is the presence of fibrillar aggregates referred to as Lewy bodies (LBs), in which α-synuclein is the major component. Converging evidence supports the prion-like transmission of α-synuclein aggregates in the onset and progression of PD. Intracellular α-synuclein aggregates into pathological fibrils, which can be transferred from aggregate-producing cells to aggregate-free cells, triggering neuronal injury and the progression of pathology. However, the specific mechanisms mediating the aggregation and transmission of pathological α-synuclein remain unknown. Here we show that cofilin 1 binds to α-synuclein and promotes its aggregation. The mixed fibrils consist of cofilin 1 and α-synuclein are more compact and more potent than pure α-synuclein fibrils in seeding α-synuclein aggregation. Cofilin 1 also facilitates the uptake of α-synuclein fibrils and finally induces neuronal dysfunction. Together, these observations indicate that cofilin 1 acts as a crucial mediator in the aggregation and propagation of pathological α-synuclein, contributing to the pathogenesis of PD. • Cofilin 1 was highly expressed in human and mouse PD brains. • Cofilin 1 combined α-synuclein and promoted its aggregation. • Cofilin 1 facilitated the propagation of α-synuclein pathology. • Cofilin 1 enhanced the toxicity of α-synuclein fibrils. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
15. The pyrethroids metabolite 3-phenoxybenzoic acid induces dopaminergic degeneration.
- Author
-
Wan, Fang, Yu, Ting, Hu, Junjie, Yin, Sijia, Li, Yunna, Kou, Liang, Chi, Xiaosa, Wu, Jiawei, Sun, Yadi, Zhou, Qiulu, Zou, Wenkai, Zhang, Zhentao, and Wang, Tao
- Published
- 2022
- Full Text
- View/download PDF
16. Exosome-mediated delivery of antisense oligonucleotides targeting α-synuclein ameliorates the pathology in a mouse model of Parkinson's disease.
- Author
-
Yang, Jiaolong, Luo, Shilin, Zhang, Jichun, Yu, Ting, Fu, Zhihui, Zheng, Yongfa, Xu, Ximing, Liu, Chaoyang, Fan, Mingxia, and Zhang, Zhentao
- Subjects
- *
PARKINSON'S disease , *OLIGONUCLEOTIDES , *ANIMAL disease models , *DOPAMINERGIC neurons , *PATHOLOGY , *MULTIPLE system atrophy - Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. Pathologically, PD is characterized by the formation of Lewy bodies (LBs) in the brain, which mainly comprises phosphorylated and aggregated α-synuclein (α-syn). The aberrant aggregation of α-syn is believed to play a key role in the pathogenesis of PD. While α-syn expression can be reduced by antisense oligonucleotides (ASOs), the challenge to deliver ASOs safely and effectively into the neurons remains unresolved. Here, we developed a safe and highly effective ASO delivery method by using exosomes. We first identified the ASO sequence that selectively reduced α-syn expression: ASO4. Exosome-mediated delivery of ASO4 (exo -ASO4) showed high cellular uptake and low toxicity in primary neuronal cultures. Exo-ASO4 also significantly attenuated α-syn aggregation induced by pre-formed α-syn fibrils in vitro. Exo-ASO4 intracerebroventricular injection into the brains of α-syn A53T mice, a transgenic model of PD, significantly decreased the expression of α-syn and attenuated its aggregation. Furthermore, exo -ASO4 ameliorated the degeneration of dopaminergic neurons in these mice. Finally, the α-syn A53T mice showed significantly improved locomotor functions after exo-ASO4 injection. Overall, this study demonstrates that exosome-mediated ASO4 delivery may be an effective treatment option for PD. • Exosomes are suitable carrier for the ASO delivery into the mouse neurons. • Exosome-mediated ASO4 delivery (exo -ASO4) can both reduce the expression level of α-synuclein and phosphorylated α-synuclein. • Exo-ASO4 attenuates α-syn pathology and thereby improves motor function in the α-syn A53T mice. • Exo-ASO4 may be an effective treatment option for PD. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. Asparagine endopeptidase inhibitor protects against fenpropathrin-induced neurodegeneration via suppressing α-synuclein aggregation and neuroinflammation.
- Author
-
Yu, Ting, Wan, Fang, Liu, Chaoyang, Zhang, Xingyu, Liu, Zehua, Zhang, Jichun, Xiong, Jing, Wang, Tao, and Zhang, Zhentao
- Subjects
- *
INFLAMMATION , *ASPARAGINE , *DOPAMINERGIC neurons , *PARKINSON'S disease , *NEURODEGENERATION , *CELL aggregation - Abstract
Exposure to fenpropathrin (Fen), one of the most widely used pyrethroid pesticides, has been reported to increase the incidence of Parkinson's disease (PD). However, the molecular mechanisms underlying Fen-induced Parkinsonism remain unknown. Here we investigated the role of the lysosomal protease asparagine endopeptidase (AEP) in Fen-induced neurodegeneration and tested the protective effect of an AEP inhibitor Compound #11 (CP11). Fen induced AEP activation, α-synuclein aggregation, and dopaminergic neuronal degeneration both in vitro and in vivo. CP11 alleviated Fen-induced cell injury in cultured SH-SY5Y cells and A53T α-synuclein transgenic mice. CP11 protected SH-SY5Y cells against Fen-induced toxicity and decreased α-synuclein aggregation in HEK293 cells stably transfected with α-synuclein. In Fen-treated mice, CP11 attenuated the degeneration of dopaminergic neurons and reduced neuroinflammation. Our findings demonstrate that neurodegeneration in Fen-treated models might be attributed to the activation of AEP. AEP might be a novel therapeutic target in PD induced by Fen and other environmental factors. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.