Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 108
1.
Int J Mol Sci ; 23(18)2022 09 15.
Article En | MEDLINE | ID: mdl-36142685

Parkinson's disease (PD) is a neurodegenerative disease with an impairment of movement execution that is related to age and genetic and environmental factors. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxin widely used to induce PD models, but the effect of MPTP on the cells and genes of PD has not been fully elucidated. By single-nucleus RNA sequencing, we uncovered the PD-specific cells and revealed the changes in their cellular states, including astrocytosis and endothelial cells' absence, as well as a cluster of medium spiny neuron cells unique to PD. Furthermore, trajectory analysis of astrocyte and endothelial cell populations predicted candidate target gene sets that might be associated with PD. Notably, the detailed regulatory roles of astrocyte-specific transcription factors Dbx2 and Sox13 in PD were revealed in our work. Finally, we characterized the cell-cell communications of PD-specific cells and found that the overall communication strength was enhanced in PD compared with a matched control, especially the signaling pathways of NRXN and NEGR. Our work provides an overview of the changes in cellular states of the MPTP-induced mouse brain.


MPTP Poisoning , Neurodegenerative Diseases , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Disease Models, Animal , Endothelial Cells/metabolism , MPTP Poisoning/genetics , MPTP Poisoning/metabolism , Mice , Mice, Inbred C57BL , Neurotoxins/adverse effects , Parkinson Disease/genetics , Parkinson Disease/metabolism , Sequence Analysis, RNA , Transcription Factors/genetics
2.
J Environ Pathol Toxicol Oncol ; 40(3): 75-85, 2021.
Article En | MEDLINE | ID: mdl-34587406

BACKGROUND: Parkinson's disease (PD) is the most prevalent disease linked with age-associated neuronal degeneration. Phytotherapeutic compounds or agents have gained increased importance because of their increased specificity and minimal side effects. Isopulegol, a monoterpene, was utilized in the present study because of its wide range of therapeutic properties. Our aim was to examine the underlying mechanism of anti-neuroinflammatory action and neuroprotective efficacy of isopulegol in cell lines and in an experimental animal model of PD. METHODS: The MTT assay was performed in microglial BV-2 cells subjected to lipopolysaccharides (LPS). The release of NO and synthesis of ROS intracellularly in BV-2 cells were detected. C57BL/6 mice induced with MPTP were examined for motor function and coordination. Expression of proinflammatory mediators was also assessed both in vivo and in vitro. Histopathological sections of brain and expression of iNOS and COX-2 were also analyzed. RESULTS: BV-2 cells did not exhibit noticeable toxicity at selected concentrations and LPS-incubated cells showed marked elevation of NO levels and increased production of intracellular ROS. Increased expression of proinflammatory cytokines was also observed. Motor function and coordination deficits were observed in mice induced with MPTP. Histopathological abnormalities and increased iNOS and COX-2 expression were noted in MPTP-induced mice. Administration of isopulegol reversed the changes brought about by LPS and MPTP. CONCLUSION: The study indicated that isopulegol is a potential therapeutic drug against clinical complications of PD.


Cyclohexane Monoterpenes/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Animals , Brain/drug effects , Brain/pathology , Cell Line , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , MPTP Poisoning/drug therapy , MPTP Poisoning/genetics , MPTP Poisoning/physiopathology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism
3.
Genes Cells ; 26(9): 684-697, 2021 Sep.
Article En | MEDLINE | ID: mdl-34086379

Autophagy in dopamine (DA) neurons is concerned to be associated with Parkinson's disease (PD), but the detailed mechanism remains unknown. Herein, we aimed to investigate the function of microRNA (miR)-29c-3p in autophagy in PD models. Intraperitoneal injection of MPTP (20 mg/kg) was given to C57BL/6 mice to establish PD mouse model. SH-SY5Y cells were treated with MPP+ (1 mmol/L) to establish in vitro PD model. The results indicated that in the substantia nigra pars compacta (SNpc) DA neurons of PD mice, autophagy was activated accompanied by down-regulated miR-29c-3p and up-regulated ten-eleven translocation 2 (TET2) expression. Up-regulation of miR-29c-3p inhibited TET2 expression and SNpc (including DA neurons) autophagy in PD mice. In vitro PD model confirmed that MPP+ treatment markedly down-regulated miR-29c-3p expression and up-regulated TET2 expression in SH-SY5Y cells in a dose/time-dependent manner. Moreover, miR-29c-3p up-regulation also inhibited autophagy and TET2 expression in vitro. Additionally, TET2 was proved to be targeted and down-regulated by miR-29c-3p. TET2 knockdown inhibited MPP+ -induced autophagy, whereas TET2 over-expression reversed the effects of miR-29c-3p over-expression on SH-SY5Y cell autophagy. Overall, miR-29c-3p over-expression inhibits autophagy in PD models, which may be mediated by TET2. Our finding may provide new insights for regulating autophagy to improve PD progression.


Autophagy , DNA-Binding Proteins/genetics , Dioxygenases/genetics , MPTP Poisoning/metabolism , MicroRNAs/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Humans , MPTP Poisoning/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neurons/metabolism
4.
Acta Biochim Biophys Sin (Shanghai) ; 53(7): 870-882, 2021 Jul 05.
Article En | MEDLINE | ID: mdl-33984130

Long non-coding RNAs (lncRNAs) play biological roles in brain disorder and neurodegenerative diseases. As the functions of lncRNA NEAT1 in Parkinson's disease (PD) remain unknown, in the present study, we aimed to explore the roles and underlying molecular mechanisms of NEAT1 in PD. A PD mouse model induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and a cell model of SH-SY5Y induced by N-methyl-4-phenylpyridinium (MPP+) were established. The ratio of tyrosine hydroxylase (TH+) cells was determined by immunofluorescence assay, and the behavioral changes in mice were observed using pole tests and rotarod tests. The cellular viability and apoptosis of SH-SY5Y were detected by MTT assay and flow cytometric analysis, respectively, and the number of autophagosomes was subsequently measured by transmission electron microscopy. High-performance liquid chromatography was performed to detect the content of dopamine, and a dual-luciferase reporter assay was used to clarify the target of NEAT1 simultaneously. The results demonstrated that the level of NEAT1 was upregulated in the MPTP-induced PD mice, dopamine neurons, and the SH-SY5Y cells treated with MPP+, whereas the level of miR-374c-5p was downregulated. NEAT1 level was positively correlated with MPP+ in a concentration-dependent manner. NEAT1 inhibition efficiently facilitated cell proliferation but inhibited apoptosis and autophagy in the MPP+-treated SH-SY5Y cells. Additionally, silencing of NEAT1 increased the TH+ rate of neurons and suppressed autophagy greatly in PD mice. As a possible target of NEAT1, miR-374c-5p could impact on the apoptosis and autophagy of the SH-SY5Y cells. NEAT1 inhibition upregulated the expression of miR-374c-5p, enhanced SH-SY5Y cell viability, and repressed autophagy and apoptosis in MPTP-induced PD mice. These findings indicated a potential therapeutic role of NEAT1 in treating PD.


Apoptosis , Autophagy , Dopaminergic Neurons/metabolism , MPTP Poisoning/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Animals , Dopaminergic Neurons/pathology , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Male , Mice , MicroRNAs/genetics , RNA, Long Noncoding/genetics
5.
Mol Biol (Mosk) ; 55(1): 152-163, 2021.
Article Ru | MEDLINE | ID: mdl-33566034

Aggregated forms of α-synuclein are core components of pathohistological inclusions known as Lewy bodies in substantia nigra (SN) neurons of patients with Parkinson's disease (PD). The role of α-synuclein in selective loss of SN dopaminergic neurons (DNs) in PD is studied in mice knocked out in the α-synuclein gene. The new mouse strain delta flox KO with a constitutive knockout of the α-synuclein gene models the end point of in vivo deletion of the α-synuclein gene in mice with a conditional knockout and has no foreign sequence in the modified genomic locus, thus differing from all other α-synuclein knockout mouse strains. The effect of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which is used to model PD, was compared between delta flox KO mice and mice of the well-known α-synuclein knockout strain AbKO. Subchronic MPTP administration, which models early PD, was found to reduce the dopamine content and to change the ratio of dopamine metabolites in the striatum to the same levels in delta flox KO, АbKO, and wild-type mice. Overt locomotor defects were not observed after MPTP treatment, but gait testing in a CatWalk XT (Noldus) system revealed identical gait deviations in mice of the two strains and control wild-type mice. Based on the findings, a similar mechanism of neurotoxic damage to DNs was assumed for delta flox KO and AbKO mice.


MPTP Poisoning , alpha-Synuclein , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Humans , MPTP Poisoning/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Substantia Nigra/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
6.
Commun Biol ; 4(1): 232, 2021 02 19.
Article En | MEDLINE | ID: mdl-33608634

Convincing evidence supports the premise that reducing α-synuclein levels may be an effective therapy for Parkinson's disease (PD); however, there has been lack of a clinically applicable α-synuclein reducing therapeutic strategy. This study was undertaken to develop a blood-brain barrier and plasma membrane-permeable α-synuclein knockdown peptide, Tat-ßsyn-degron, that may have therapeutic potential. The peptide effectively reduced the level of α-synuclein via proteasomal degradation both in cell cultures and in animals. Tat-ßsyn-degron decreased α-synuclein aggregates and microglial activation in an α-synuclein pre-formed fibril model of spreading synucleinopathy in transgenic mice overexpressing human A53T α-synuclein. Moreover, Tat-ßsyn-degron reduced α-synuclein levels and significantly decreased the parkinsonian toxin-induced neuronal damage and motor impairment in a mouse toxicity model of PD. These results show the promising efficacy of Tat-ßsyn-degron in two different animal models of PD and suggest its potential use as an effective PD therapeutic that directly targets the disease-causing process.


Antiparkinson Agents/pharmacology , Brain/drug effects , MPTP Poisoning/drug therapy , Neurons/drug effects , Parkinson Disease/drug therapy , Peptides/pharmacology , alpha-Synuclein/metabolism , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Down-Regulation , HEK293 Cells , Humans , MPTP Poisoning/genetics , MPTP Poisoning/metabolism , MPTP Poisoning/physiopathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Mutation , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats, Sprague-Dawley , alpha-Synuclein/genetics
7.
Int J Mol Med ; 47(2): 751-760, 2021 02.
Article En | MEDLINE | ID: mdl-33416093

Aldose reductase (AR) is known to detoxify aldehydes and prevent oxidative stress. Although AR exerts antioxidant effects, the role of AR in Parkinson's disease (PD) remains unclear. The objective of the present study was to investigate the protective effects of AR protein against 1­methyl­4­phenylpyridinium (MPP+)­induced SH­SY5Y cell death and 1­methyl­4­phenyl­1,2,3,6­tetrahydropyridine (MPTP)­induced PD in a mouse model using the cell permeable Tat­AR fusion protein. The results revealed that when Tat­AR protein was transduced into SH­SY5Y cells, it markedly protected the cells against MPP+­induced death and DNA fragmentation. It also reduced the activation of mitogen-activated protein kinase (MAPKs) and regulated the expression levels of Bcl­2, Bax and caspase­3. Immunohistochemical analysis revealed that when Tat­AR protein was transduced into the substantia nigra (SN) of mice with PD, it markedly inhibited dopaminergic neuronal cell death. Therefore, Tat­AR may be useful as a therapeutic protein for PD.


Aldehyde Reductase/metabolism , Dopaminergic Neurons/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Oxidative Stress , Substantia Nigra/enzymology , Aldehyde Reductase/genetics , Animals , Cell Death , Cell Line, Tumor , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , MPTP Poisoning/enzymology , MPTP Poisoning/genetics , Male , Mice
8.
Mol Neurobiol ; 58(2): 603-616, 2021 Feb.
Article En | MEDLINE | ID: mdl-32997292

Parkinson's disease (PD) is a neurodegenerative disease caused by a variety of unclear complex pathogenic factors. The 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine/probenecid (MPTP/p)-induced progressive PD mice is a well-recognized classic model for studying PD, but the molecular toxicology of this model is still unclear. Here, for the first time, we report gradual neurodegenerative processes in MPTP/p-induced progressive PD mice model using RNA-seq. Transcriptional responses are orchestrated to regulate the expression of many genes in substantia nigra, such as Ntf3, Pitx3, Th, and Drd2, leading to the degeneration of dopaminergic neurons at last. We proposed that the established model could be divided into three phases based on their molecular toxicological features: "the stress response phase" which maintained the microenvironment homeostasis, "the pre-neurodegenerative phase" which demonstrated observed MPTP/p cytotoxicity and gradual degeneration of dopaminergic neurons, and "the neurodegenerative phase" which reflected distinct damage and dopaminergic neuron apoptotic process. Glia cells exhibited a certain protective effect on dopaminergic neurons in 3rd and 6th MPTP/p-induced cytotoxicity. But in 10th MPTP/p injection, glia cells play a promoting role in PD and tissue damages caused by oxidative stress. This study also indicated that the substantia nigra of PD mice showed unique patterns of changes at each stage. Moreover, neurotrophic signaling pathway, ECM-receptor interaction, oxidative phosphorylation, apoptosis and necroptosis were enriched at 3rd and 6th MPTP/p injection, which might be associated with the PD progress. This study provided an extensive data set of molecular toxicology for elucidating of PD progression and offered comprehensive theoretical knowledge for the development of new therapy.


Gene Expression Regulation , MPTP Poisoning/genetics , Nerve Degeneration/genetics , Parkinson Disease/genetics , Probenecid/toxicity , Transcriptome/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Behavior, Animal , Disease Models, Animal , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Nerve Degeneration/pathology , Reproducibility of Results , Substantia Nigra/pathology , Transcriptome/drug effects , Tyrosine 3-Monooxygenase/metabolism
9.
J Neurosci ; 40(43): 8262-8275, 2020 10 21.
Article En | MEDLINE | ID: mdl-32928885

A subset of adult ventral tegmental area dopamine (DA) neurons expresses vesicular glutamate transporter 2 (VGluT2) and releases glutamate as a second neurotransmitter in the striatum, while only few adult substantia nigra DA neurons have this capacity. Recent work showed that cellular stress created by neurotoxins such as MPTP and 6-hydroxydopamine can upregulate VGluT2 in surviving DA neurons, suggesting the possibility of a role in cell survival, although a high level of overexpression could be toxic to DA neurons. Here we examined the level of VGluT2 upregulation in response to neurotoxins and its impact on postlesional plasticity. We first took advantage of an in vitro neurotoxin model of Parkinson's disease and found that this caused an average 2.5-fold enhancement of Vglut2 mRNA in DA neurons. This could represent a reactivation of a developmental phenotype because using an intersectional genetic lineage-mapping approach, we find that >98% of DA neurons have a VGluT2+ lineage. Expression of VGluT2 was detectable in most DA neurons at embryonic day 11.5 and was localized in developing axons. Finally, compatible with the possibility that enhanced VGluT2 expression in DA neurons promotes axonal outgrowth and reinnervation in the postlesional brain, we observed that DA neurons in female and male mice in which VGluT2 was conditionally removed established fewer striatal connections 7 weeks after a neurotoxin lesion. Thus, we propose here that the developmental expression of VGluT2 in DA neurons can be reactivated at postnatal stages, contributing to postlesional plasticity of dopaminergic axons.SIGNIFICANCE STATEMENT A small subset of dopamine neurons in the adult, healthy brain expresses vesicular glutamate transporter 2 (VGluT2) and thus releases glutamate as a second neurotransmitter in the striatum. This neurochemical phenotype appears to be plastic as exposure to neurotoxins, such as 6-OHDA or MPTP, that model certain aspects of Parkinson's disease pathophysiology, boosts VGluT2 expression in surviving dopamine neurons. Here we show that this enhanced VGluT2 expression in dopamine neurons drives axonal outgrowth and contributes to dopamine neuron axonal plasticity in the postlesional brain. A better understanding of the neurochemical changes that occur during the progression of Parkinson's disease pathology will aid the development of novel therapeutic strategies for this disease.


Corpus Striatum/physiology , Dopaminergic Neurons/metabolism , Vesicular Glutamate Transport Protein 2/biosynthesis , Animals , Animals, Newborn , Axons/physiology , Cell Lineage/genetics , Cell Survival/genetics , Corpus Striatum/embryology , Corpus Striatum/growth & development , Female , MPTP Poisoning/genetics , MPTP Poisoning/metabolism , Mesencephalon/embryology , Mesencephalon/growth & development , Mesencephalon/physiology , Mice , Mice, Knockout , Neural Pathways/embryology , Neural Pathways/growth & development , Neural Pathways/physiology , Neurotoxins/toxicity , Pregnancy , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Vesicular Glutamate Transport Protein 2/genetics
10.
Parkinsonism Relat Disord ; 79: 88-90, 2020 10.
Article En | MEDLINE | ID: mdl-32906083

Susceptibility to Parkinson's disease (PD) increases more than threefold in first-degree relatives of PD patients. Using a mouse model, we investigated in a proof-of-principle approach whether toxin exposure of F0 affects the F1 generation. We provide first evidence that disturbance of the nigrostriatal pathway can be transferred to the next generation.


Epigenesis, Genetic/genetics , Genetic Predisposition to Disease/genetics , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Animals , Corpus Striatum/pathology , Disease Models, Animal , Mice , Neural Pathways/pathology , Proof of Concept Study , Substantia Nigra/pathology
11.
Pharmacol Res ; 160: 105197, 2020 10.
Article En | MEDLINE | ID: mdl-32942015

Parkin is a crucial protein that promotes the clearance of damaged mitochondria via mitophagy in neuron, and parkin mutations result in autosomal-recessive Parkinson's disease (AR-PD). However, the exact mechanisms underlying the regulation of Parkin-mediated mitophagy in PD remain unclear. In this study, PD models were generated through incubation of SH-SY5Y cells with 1-methyl-4-phenylpyridinium ion (MPP+, 1.5 mM for 24 h) and intraperitoneal injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, 30 mg/kg for five consecutive days) in mice. A Bioinformatics database was used to identify Parkin-targeting microRNAs (miRNAs). Then, miR-103a-3p agomir, miR-103a-3p antagomir and Parkin siRNA were used to assess the effects of miR-103a-3p/Parkin/Ambra1 signaling-mediated mitophagy in PD in vitro and in vivo. The protein and mRNA levels of Parkin and Ambra1 were significantly decreased, while miR-103a-3p, which is a highly expressed miRNA in the human brain, was obviously increased in PD mouse and SH-SY5Y cell models. Moreover, miR-103a-3p suppressed Parkin expression by targeting a conserved binding site in the 3'-untranslated region (UTR) of Parkin mRNA. Importantly, miR-103a-3p inhibition resulted in neuroprotective effects and improved mitophagy in vitro and in vivo, whereas Parkin siRNA strongly abolished these effects. These findings suggested that miR-103a-3p inhibition has neuroprotective effects in PD, which may be involved in regulating mitophagy through the Parkin/Ambra1 pathway. Modulating miR-103a-3p levels may be an applicable therapeutic strategy for PD.


Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , MicroRNAs/genetics , Mitophagy/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions/genetics , Animals , Cell Line , Computational Biology , Dopamine/metabolism , Humans , MPTP Poisoning/drug therapy , MPTP Poisoning/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Point Mutation , RNA, Small Interfering/pharmacology
12.
Neuropharmacology ; 181: 108335, 2020 12 15.
Article En | MEDLINE | ID: mdl-32979381

Parkinson's disease (PD) is one of the common complex neurodegenerative diseases and characterized by abnormal metabolic brain networks. Fibroblast growth factor 21 (FGF21), an endocrine hormone that belongs to the fibroblast growth factor superfamily, plays an extensive role in the regulation of metabolism. However, our understandings of the specific function and mechanisms of FGF21 on PD are still quite limited. Here we aimed to elucidate the actions and the underlying mechanisms of FGF21 on dopaminergic neurodegeneration using cellular and animal models of parkinsonism. To investigate the effects of FGF21 on dopaminergic neurodegeneration in vivo and in vitro, 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine models of PD were utilized, and animals were treated with recombinant FGF21 protein or FGF21 gene delivered via an adeno-associated virus. In the present study, systemic and continuous intracerebroventricular recombinant FGF21 protein administration to mice both prevented behavioral deficits, protected dopaminergic neurons against degeneration, and ameliorated α-synuclein pathology in PD models; and in vivo gene delivery of FGF21 improved PD-like symptoms and pathologies suggesting a potential implication of FGF21 in gene therapy for PD. In vitro evidence confirmed FGF21 mediated neuroprotective benefits against PD pathologies. Further, our data suggested that enhanced autophagy was involved in the FGF21 neuroprotection in PD models, and silent information regulator 2 homolog 1 may play a crucial role in molecular mechanisms underlying anti-PD activities of FGF21.


Fibroblast Growth Factors/genetics , Genetic Therapy/methods , Neurodegenerative Diseases/genetics , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/therapy , Sirtuin 1/genetics , Animals , Autophagy/drug effects , Behavior, Animal/drug effects , Dopaminergic Neurons/pathology , Humans , MPTP Poisoning/genetics , MPTP Poisoning/pathology , MPTP Poisoning/therapy , Male , Mice , Mice, Inbred C57BL , Neuroprotection , Parkinson Disease, Secondary/pathology , Recombinant Proteins/therapeutic use , Sirtuin 1/drug effects , alpha-Synuclein/genetics
13.
Hum Gene Ther ; 31(23-24): 1274-1287, 2020 12.
Article En | MEDLINE | ID: mdl-32808542

Long-noncoding RNA taurine upregulated gene 1 (TUG1) participates in nervous system diseases, but its function in Parkinson's disease (PD) remains unclear. This study explored the function and mechanism of TUG1 in PD. A PD model was constructed using SH-SY5Y cells induced by 1-methyl-4-phenylpyridinium (MPP+) in vitro and mice treated by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in vivo. The expressions of TUG1, miR-152-3p, phosphatase and tensin homologue (PTEN), tyrosine hydroxylase (TH), and Bcl-2, and cleaved caspase-3 expressions were determined by quantitative reverse transcription-PCR and Western blotting. The viability, apoptosis, reactive oxygen species, and release of inflammatory factors from SH-SY5Y cells and substantia nigra tissues were detected by commercial kits. The interaction between TUG1 and miR-152-3p was analyzed by dual-luciferase reporter assay. Hematoxylin/eosin and immunohistochemical staining was performed for assessing the pathological damage and proportion of TH-positive cells. In PD cell model and mice model, TUG1 expression was upregulated and that of miR-152-3p was downregulated. Further research showed that TUG1 sponged and regulated miR-152-3p expression. Silencing of TUG1 not only protected SH-SY5Y cells against cell apoptosis, oxidative stress, and neuroinflammation in vitro, pathological damage and neuroinflammation in vivo, but also suppressed the expressions of PTEN and cleaved caspase-3, and increased the expressions of TH and Bcl-2 in MPP+-treated SH-SY5Y cells. However, the protective role of siTUG1 in SH-SY5Y cells was significantly inhibited by the miR-152-3p inhibitor. Thus, knocking down TUG1 might have a protective effect on PD through the miR-152-3p/PTEN pathway.


MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Parkinson Disease, Secondary/genetics , Parkinson Disease/genetics , RNA, Long Noncoding/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Gene Expression Regulation , Humans , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Mice , Parkinson Disease/pathology , Parkinson Disease, Secondary/chemically induced , Reactive Oxygen Species/metabolism , Signal Transduction
14.
Learn Mem ; 27(9): 372-379, 2020 09.
Article En | MEDLINE | ID: mdl-32817303

Parkinson's disease (PD) is characterized clinically by progressive motor dysfunction; overt parkinsonism is often preceded by prodromal symptoms including disturbances in the sleep-wake cycle. Up to 80% of patients with PD also develop dementia. In humans, there are three major apolipoprotein E isoforms: E2, E3, and E4. Increased rate of dementia in PD may be associated with E4 isoform. To better understand prodromal changes associated with E4, we exposed young (3-5 mo) male and female mice expressing E3 or E4 via targeted replacement to a subchronic dosage of the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We hypothesized that E4 mice would be more susceptible to MPTP-related behavioral and cognitive changes. MPTP-treated E4 mice explored novel objects longer than genotype-matched saline-treated mice. In contrast, saline-treated E3 mice preferentially explored the novel object whereas MPTP-treated E3 mice did not and showed impaired object recognition. MPTP treatment altered swim speed of E4, but not E3, mice in the water maze compared to controls. Thus, E4 carriage may influence the preclinical symptoms associated with PD. Increased efforts are warranted to study early time points in this disease model.


Apolipoprotein E3 , Apolipoprotein E4 , Behavior, Animal , MPTP Poisoning/genetics , MPTP Poisoning/physiopathology , Motor Activity , Recognition, Psychology , Spatial Learning , Animals , Behavior, Animal/physiology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Recognition, Psychology/physiology , Spatial Learning/physiology
15.
J Mol Neurosci ; 70(12): 1977-1986, 2020 Dec.
Article En | MEDLINE | ID: mdl-32617872

The objective of this study was to understand if RNF13 can affect Parkinson's disease (PD) model mice by modulating the endoplasmic reticulum stress (ERS)-mediated IRE1α-TRAF2-ASK1-JNK pathway. C57BL/6 mice injected with MPTP to establish PD mice models were divided into Control, MPTP, MPTP + sh-RNF13, and MPTP + sh-NC groups. Rotarod, balance beam, and open-field tests were used to assess the behavioral changes of experimental mice. Immunofluorescence assay was used to determine TH-positive expression in substantia nigra, TUNEL staining to detect apoptosis, and Western blotting to measure the expression of IRE1α-TRAF2-ASK1-JNK pathway. Besides, SH-SY5Y cells treated with MPP+ were assigned into Control, MPP+, MPP+ + sh-RNF13, and MPP+ + sh-NC groups in vitro to detect cell viability, apoptosis and Ca2+ level. When compared with those Control mice, MPTP mice showed decreased retention time spent on rotarod performance and prolonged time on balance beam test, as well as evident reductions in floor plane (FP) movements, moving time, moving distance, and mean velocity in open-field test, which had an obvious increase of TUNEL-positive cells, significant decrease of TH-positive cells, and remarkable up-regulations of RNF13, p-IRE1α/IRE1α, TRAF2, ASK1, and p-JNK/JNK. Meanwhile, MPTP mice treated with sh-RNF13 were improved in all above indexes. In vitro, MPP+ treated SH-SY5Y cells had decreased cell viability and increased cell apoptosis, as well as the upregulated IRE1α-TRAF2-ASK1-JNK pathway proteins and Ca2+ level. RNF13 knockdown improved all above indexes in SH-SY5Y cells treated with MPP+. Silencing RNF13 can alleviate motor dysfunction and dopamine neuronal damage in PD mice by inhibiting ERS-mediated IRE1α-TRAF2-ASK1-JNK pathway.


Gene Silencing , MPTP Poisoning/genetics , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis , Calcium/metabolism , Cell Line, Tumor , Dopaminergic Neurons/metabolism , Endoplasmic Reticulum Stress , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinase 5/metabolism , MPTP Poisoning/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Movement , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin-Protein Ligases/genetics
16.
Mol Neurobiol ; 57(7): 2920-2933, 2020 Jul.
Article En | MEDLINE | ID: mdl-32436108

Parkinson's disease (PD) is a progressive neurodegenerative disease highlighted by a marked loss of dopaminergic cell loss and motor disturbances. Currently, there are no drugs that slow the progression of the disease. A myriad of factors have been implicated in the pathogenesis and progression of PD including neuroinflammation. Although anti-inflammatory agents are being evaluated as potential disease-modifying therapies for PD, none has proven effective to date, suggesting that new and novel targets are needed. Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a transmembrane glycoprotein that has recently been shown to reduce inflammation in astrocytes and to be increased in post-mortem PD brain samples. Here we show that transgenic overexpression of GPNMB protects against dopaminergic neurodegeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropridine mouse model of Parkinson's disease. Furthermore, GPNMB overexpression reduces gliosis and prevented microglial morphological changes following MPTP treatment compared with wild-type MPTP-treated mice. Additionally, recombinant GPNMB attenuates LPS-induced inflammation in primary mouse microglia. These results suggest a neuroprotective and anti-inflammatory role for GPNMB and warrant further investigation for GPNMB as a novel therapy for PD.


Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Eye Proteins/genetics , MPTP Poisoning/genetics , Membrane Glycoproteins/genetics , Microglia/metabolism , Animals , Corpus Striatum/pathology , Dopaminergic Neurons/pathology , Eye Proteins/metabolism , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Microglia/pathology
17.
J Mol Neurosci ; 69(2): 197-214, 2019 Oct.
Article En | MEDLINE | ID: mdl-31270675

Parkinson's disease (PD), one of the motor system disorders, is characterized by the loss of dopamine-producing brain cells. Accumulating evidence has highlighted the involvement of microRNAs (miRs) in the development and progression of PD. Hence, we aimed at exploring possible effects of miR-326 on the progression of PD in mice in an attempt to elucidate the underlying mechanism associated with the kallikrein-related peptidase 7 (KLK7)-mediated mitogen-activated protein kinase (MAPK) signaling pathway. In order to identify the regulatory relationship between miR-326 and KLK7 and its biological significance in PD, PD mouse models were established and subsequently treated with mimics or inhibitors of miR-326 or siRNA-KLK7. The content of striatal dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), 3-methoxytyrosine (3-MT), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA); positive expression of tyrosine hydroxylase (TH) and inducible nitric oxide synthase (iNOS); and the levels of IL-1, IL-6, TNF-α, INF-γ, and MAPK signaling pathway-related genes were determined accordingly. The results obtained indicated that KLK7 was negatively targeted by miR-326, with lower miR-326 and higher KLK7 detected among PD mice. The overexpression of miR-326 or silencing of KLK7 was demonstrated to increase the content of DA, DOPAC, HVA, 3-MT, SOD, GSH-Px, and TH positive expression, while reducing iNOS positive expression, MDA content and cell apoptosis, as well as inhibited levels of IL-1, IL-6, TNF-α, INF-γ, and mRNA and protein levels of p38, ERK, JNK, and caspase-3. Taken together, these results provided evidence suggesting that miR-326 could inhibit iNOS activation and apoptosis of dopaminergic neurons through inhibiting the MAPK signaling pathway and negatively regulating KLK7 in mice with PD. These findings highlight the potential of miR-326 as a novel target for future PD treatment.


Apoptosis , Dopaminergic Neurons/metabolism , Kallikreins/genetics , MAP Kinase Signaling System , MPTP Poisoning/metabolism , MicroRNAs/genetics , Animals , Cell Proliferation , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/metabolism , Interleukins/genetics , Interleukins/metabolism , Kallikreins/metabolism , MPTP Poisoning/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley
18.
Neurosci Lett ; 709: 134352, 2019 09 14.
Article En | MEDLINE | ID: mdl-31283965

This study aims to investigate the function and molecular mechanisms of Tribbles homolog 3 (TRB3) on the MPP+/MPTP-induced Parkinson's disease (PD). In this study, MPP+-induced PD cellular model and MPTP-caused PD mice model were established. Following the transfection with TRB3-shRNA, cell viability, cell apoptosis, ROS level, and the ratio of p-p38/ p38, p-JNK/JNK, p-AKT/AKT were examined. At the same time, behavior assessment of wild type female C57BL/6 mice and whole-body TRB3 knockout mice PD models caused by MPTP were performed by Rotarod test and Open-field test. The results showed that TRB3 was markedly upregulated in MPP+-induced cellular model through ATF4/CHOP pathway. Knockdown of TRB3 significantly decreased the MPP+-induced reduction of cell viability, augment of cell apoptosis and accumulation of ROS, inhibited the phosphorylation of p38 and JNK, and promoted the phosphorylation of AKT, in vitro. Further, knockout of TRB3 improved the behavior impairment of PD mice induced by MPTP, in vivo. In conclusion, knockdown of TRB3 has a neuroprotective effect on MPTP/MPP+-induced PD cellular and mice models, through regulating MAPK and AKT signaling pathways.


Cell Cycle Proteins/deficiency , MAP Kinase Signaling System/physiology , MPTP Poisoning/genetics , MPTP Poisoning/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/physiology , Female , Humans , MPTP Poisoning/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics
19.
J Cell Physiol ; 234(12): 23379-23387, 2019 12.
Article En | MEDLINE | ID: mdl-31232472

Parkinson's disease (PD) is neurodegenerative dyskinesia characterized by loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Although neuroinflammation is one of the pathological features of PD, its mechanism of promoting PD is still not fully understood. Recently, the microRNA (miR) is considered to play a critical regulatory role in inflammatory responses. In this study, we examined the anti-inflammatory activity, antineuronal injury, and the underlying target of miR-190 with MPTP-induced PD mouse model and BV2 cells. The results showed that miR-190 is downregulated in lipopolysaccharide (LPS)-induced BV2 cells; however, when the miR-190 overexpressed, the expression of proinflammatory mediators, such as iNOS, IL-6, TNF-α, and TGF-ß1, were inhibited and the anti-inflammatory mediator such IL-10 was increased. In addition, we predicted the potential target of miR-190 to be Nlrp3 and verified by luciferase reporter assay. The results also showed that Nlrp3 was upregulated in LPS-induced BV2 cells, whereas knockdown of Nlrp3 inhibited the LPS-induced inflammatory response in BV2 cells. Furthermore, upregulation of miR-190 or knockdown of Nlrp3 inhibited LPS-induced apoptosis in BV2 cells. However, the apoptosis inhibition effect of miR-190 was abrogated by overexpression of Nlrp3. Finally, upregulation of miR-190 inhibited the activation of microglial cells and inflammation and attenuated the tyrosine hydroxylase loss in SNpc in MPTP-induced PD mice. In conclusion, we demonstrated that miR-190 alleviates neuronal damage and inhibits inflammation via negatively regulating the expression and activation of Nlrp3 in MPTP-induced PD mouse model.


Apoptosis , Brain/metabolism , Encephalitis/metabolism , Inflammasomes/metabolism , MPTP Poisoning/metabolism , MicroRNAs/metabolism , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neurons/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Brain/pathology , Cell Line, Tumor , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/genetics , Encephalitis/pathology , Humans , Inflammasomes/genetics , Inflammation Mediators/metabolism , MPTP Poisoning/chemically induced , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neurons/pathology , Signal Transduction
20.
Dig Dis Sci ; 64(12): 3502-3517, 2019 12.
Article En | MEDLINE | ID: mdl-31187328

BACKGROUND: Gastrointestinal (GI) motility dysfunction is the most common non-motor symptom of Parkinson's disease (PD). Studies have indicated that GI motility functions are impaired before the onset of PD. AIMS: To investigate the underlying mechanism of PD-induced GI dysmotility in MPTP (1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine)-induced animal model. METHODS: C57BL/6 mice were administered with or without a selective dopamine neurotoxin, MPTP, to induce parkinsonian symptoms. In addition to in vivo studies, in vitro experiments were also conducted in colon specimens using l-methyl-4-phenylpyridinium (MPP+), a metabolic product of MPTP. Gastric emptying, colon motility, nitrergic relaxation, and western blot experiments were performed as reported. RESULTS: MPTP-induced PD mice showed decreased expression of nuclear factor erythroid 2-related factor (Nrf2) and its target phase II genes in gastric and colon neuromuscular tissues. Decreased levels of tetrahydrobiopterin (BH4, a critical cofactor for nNOS dimerization) associated with uncoupling of nNOS in gastric and colon tissues exposed to MPTP. Impaired enteric nitrergic system led to delayed gastric emptying and slower colonic motility compared to the control mice. In vitro results in colon specimens confirm that activation of Nrf2 restored MPP+-induced suppression of alpha-synuclein, tyrosine hydroxylase (TH), Nrf2, and heme oxygenase-1. In vitro exposure to L-NAME [N(w)-nitro-L-arginine methyl ester], a NOS synthase inhibitor, reduced protein expression of TH in colon tissue homogenates. CONCLUSIONS: Loss of Nrf2/BH4/nNOS expression in PD impairs antioxidant gene expression, which deregulates NO synthesis, thereby contributing to the development of GI dysmotility and constipation. Nitric oxide appears to be important to maintain dopamine synthesis in the colon.


Gastrointestinal Motility/physiology , MPTP Poisoning/genetics , NF-E2-Related Factor 2/genetics , Nitric Oxide Synthase Type I/genetics , Nitric Oxide/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Biopterins/analogs & derivatives , Biopterins/metabolism , Blotting, Western , Colon/drug effects , Colon/metabolism , Colon/physiopathology , Constipation , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gastric Emptying/physiology , Gene Expression Regulation , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , MPTP Poisoning/metabolism , MPTP Poisoning/physiopathology , Male , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/metabolism , Parkinson Disease/physiopathology , Parkinsonian Disorders , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/drug effects , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
...