Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Pharmacother ; 174: 116442, 2024 May.
Article in English | MEDLINE | ID: mdl-38513596

ABSTRACT

Parkinson's disease (PD) is a complex neurodegenerative disorder with an unclear etiology. Despite significant research efforts, developing disease-modifying treatments for PD remains a major unmet medical need. Notably, drug repositioning is becoming an increasingly attractive direction in drug discovery, and computational approaches offer a relatively quick and resource-saving method for identifying testable hypotheses that promote drug repositioning. We used an artificial intelligence (AI)-based drug repositioning strategy to screen an extensive compound library and identify potential therapeutic agents for PD. Our AI-driven analysis revealed that efavirenz and nevirapine, approved for treating human immunodeficiency virus infection, had distinct profiles, suggesting their potential effects on PD pathophysiology. Among these, efavirenz attenuated α-synuclein (α-syn) propagation and associated neuroinflammation in the brain of preformed α-syn fibrils-injected A53T α-syn Tg mice and α-syn propagation and associated behavioral changes in the C. elegans BiFC model. Through in-depth molecular investigations, we found that efavirenz can modulate cholesterol metabolism and mitigate α-syn propagation, a key pathological feature implicated in PD progression by regulating CYP46A1. This study opens new avenues for further investigation into the mechanisms underlying PD pathology and the exploration of additional drug candidates using advanced computational methodologies.


Subject(s)
Alkynes , Artificial Intelligence , Benzoxazines , Cyclopropanes , Drug Repositioning , Parkinson Disease , alpha-Synuclein , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Alkynes/pharmacology , Benzoxazines/pharmacology , Drug Repositioning/methods , Animals , alpha-Synuclein/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Mice , Caenorhabditis elegans/drug effects , Mice, Transgenic , Humans , Nevirapine/pharmacology , Disease Models, Animal , Mice, Inbred C57BL
2.
Molecules ; 28(22)2023 Nov 08.
Article in English | MEDLINE | ID: mdl-38005205

ABSTRACT

Zaluzanin C (ZC), a sesquiterpene lactone isolated from Laurus nobilis L., has been reported to have anti-inflammatory and antioxidant effects. However, the mechanistic role of ZC in its protective effects in Kupffer cells and hepatocytes has not been elucidated. The purpose of this study was to elucidate the efficacy and mechanism of action of ZC in Kupffer cells and hepatocytes. ZC inhibited LPS-induced mitochondrial ROS (mtROS) production and subsequent mtROS-mediated NF-κB activity in Kupffer cells (KCs). ZC reduced mRNA levels of pro-inflammatory cytokines (Il1b and Tnfa) and chemokines (Ccl2, Ccl3, Ccl4, Cxcl2 and Cxcl9). Tumor necrosis factor (TNF)-α-induced hepatocyte mtROS production was inhibited by ZC. ZC was effective in alleviating mtROS-mediated mitochondrial dysfunction. ZC enhanced mitophagy and increased mRNA levels of fatty acid oxidation genes (Pparα, Cpt1, Acadm and Hadha) and mitochondrial biosynthetic factors (Pgc1α, Tfam, Nrf1 and Nrf2) in hepatocytes. ZC has proven its anti-lipid effect by improving lipid accumulation in hepatocytes by enhancing mitochondrial function to facilitate lipid metabolism. Therefore, our study suggests that ZC may be an effective compound for hepatoprotection by suppressing inflammation and lipid accumulation through regulating mtROS.


Subject(s)
Hepatocytes , Kupffer Cells , Humans , Kupffer Cells/metabolism , Reactive Oxygen Species/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mitochondria/metabolism , RNA, Messenger/metabolism , Lipids/pharmacology , Liver , Lipid Metabolism
3.
Exp Neurobiol ; 32(3): 147-156, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37403223

ABSTRACT

Parkinson's disease (PD) is characterized by the presence of α-synuclein (α-syn) inclusions in the brain and the degeneration of dopamine-producing neurons. There is evidence to suggest that the progression of PD may be due to the prion-like spread of α-syn aggregates, so understanding and limiting α-syn propagation is a key area of research for developing PD treatments. Several cellular and animal model systems have been established to monitor α-syn aggregation and propagation. In this study, we developed an in vitro model using A53T α-syn-EGFP overexpressing SH-SY5Y cells and validated its usefulness for high-throughput screening of potential therapeutic targets. Treatment with preformed recombinant α-syn fibrils induced the formation of aggregation puncta of A53T α-syn-EGFP in these cells, which were analyzed using four indices: number of dots per cell, size of dots, intensity of dots, and percentage of cells containing aggregation puncta. Four indices are reliable indicators of the effectiveness of interventions against α-syn propagation in a one-day treatment model to minimize the screening time. This simple and efficient in vitro model system can be used for high-throughput screening to discover new targets for inhibiting α-syn propagation.

4.
Int J Mol Sci ; 23(15)2022 Jul 27.
Article in English | MEDLINE | ID: mdl-35955451

ABSTRACT

The presence of protein inclusions, called Lewy bodies (LBs) and Lewy neurites (LNs), in the brain is the main feature of Parkinson's disease (PD). Recent evidence that the prion-like propagation of α-synuclein (α-syn), as a major component of LBs and LNs, plays an important role in the progression of PD has gained much attention, although the molecular mechanism remains unclear. In this study, we evaluated whether neuronal ApoE regulates the cell-to-cell transmission of α-syn and explored its molecular mechanism using in vitro and in vivo model systems. We demonstrate that neuronal ApoE deficiency attenuates both α-syn uptake and release by downregulating LRP-1 and LDLR expression and enhancing chaperone-mediated autophagy activity, respectively, thereby contributing to α-syn propagation. In addition, we observed that α-syn propagation was attenuated in ApoE knockout mice injected with pre-formed mouse α-syn fibrils. This study will help our understanding of the molecular mechanisms underlying α-syn propagation.


Subject(s)
Apolipoproteins E/metabolism , Parkinson Disease , alpha-Synuclein/metabolism , Animals , Apolipoproteins E/genetics , Lewy Bodies/metabolism , Mice , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , alpha-Synuclein/genetics
5.
Mol Brain ; 14(1): 122, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34321069

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, with aging being considered the greatest risk factor for developing PD. Caveolin-1 (Cav-1) is known to participate in the aging process. Recent evidence indicates that prion-like propagation of misfolded α-synuclein (α-syn) released from neurons to neighboring neurons plays an important role in PD progression. In the present study, we demonstrated that cav-1 expression in the brain increased with age, and considerably increased in the brain of A53T α-syn transgenic mice. Cav-1 overexpression facilitated the uptake of α-syn into neurons and formation of additional Lewy body-like inclusion bodies, phosphorylation of cav-1 at tyrosine 14 was found to be crucial for this process. This study demonstrates the relationship between age and α-syn spread and will facilitate our understanding of the molecular mechanism of the cell-to-cell transmission of α-syn.


Subject(s)
Aging/metabolism , Caveolin 1/metabolism , Neurons/metabolism , alpha-Synuclein/metabolism , Animals , Brain/pathology , Cell Line, Tumor , Cells, Cultured , Endocytosis , Humans , Inclusion Bodies/metabolism , Lewy Bodies/metabolism , Male , Membrane Microdomains , Mice, Inbred C57BL , Models, Biological , Phosphorylation , Phosphotyrosine/metabolism , Proteolysis , Rats, Sprague-Dawley
7.
EMBO Rep ; 21(7): e48950, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32372484

ABSTRACT

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons located in the substantia nigra pars compacta and the presence of proteinaceous inclusions called Lewy bodies and Lewy neurites in numerous brain regions. Increasing evidence indicates that Lewy pathology progressively involves additional regions of the nervous system as the disease advances, and the prion-like propagation of α-synuclein (α-syn) pathology promotes PD progression. Accordingly, the modulation of α-syn transmission may be important for the development of disease-modifying therapies in patients with PD. Here, we demonstrate that α-syn fibrils induce c-src activation in neurons, which depends on the FcγRIIb-SHP-1/-2-c-src pathway and enhances signals for the uptake of α-syn into neurons. Blockade of c-src activation inhibits the uptake of α-syn and the formation of Lewy body-like inclusions. Furthermore, the blockade of c-src activation also inhibits the release of α-syn via activation of autophagy. The brain-permeable c-src inhibitor, saracatinib, efficiently reduces α-syn propagation into neighboring regions in an in vivo model system. These results suggest a new therapeutic target against progressive PD.


Subject(s)
Parkinson Disease , alpha-Synuclein , Brain/metabolism , Dopaminergic Neurons/metabolism , Humans , Lewy Bodies/metabolism , Parkinson Disease/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...