Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 522
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Biophys Res Commun ; 729: 150358, 2024 10 15.
Article in English | MEDLINE | ID: mdl-38981401

ABSTRACT

Pterostilbene (PTE), a naturally occurring phenolic compound primarily found in blueberries, demonstrates neuroprotective properties. However, the role of PTE in Parkinson's disease (PD) remains unclear. This study aimed to investigate the neuroprotective role of PTE in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD animal model. Our findings demonstrate that administering PTE effectively reversed the diminished levels of dopamine in the striatum, thereby ameliorating motor impairments in the MPTP model. Moreover, PTE administration mitigated the loss of dopaminergic (DA) neurons and reduced the upregulation of α-synuclein (α-syn) induced by MPTP. Mechanistic analysis revealed that PTE administration inhibited the activation of microglia and astrocytes, as well as pro-inflammatory factors such as TNF-α and IL-1ß in the MPTP model. Additionally, PTE administration decreased MPTP-induced levels of reactive oxygen species (ROS) and malondialdehyde (MDA), while increasing total antioxidant capacity (TAOC) and superoxide dismutase (SOD) activity, thereby attenuating oxidative stress. Collectively, these findings demonstrate that PTE exerts neuroprotective effects in the MPTP mouse model of PD by suppressing neuroinflammation and oxidative stress. Thus, PTE holds promise as a therapeutic agent for PD.


Subject(s)
Mice, Inbred C57BL , Neuroinflammatory Diseases , Neuroprotective Agents , Oxidative Stress , Stilbenes , Animals , Oxidative Stress/drug effects , Stilbenes/pharmacology , Stilbenes/therapeutic use , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Reactive Oxygen Species/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , alpha-Synuclein/metabolism
2.
Environ Toxicol ; 39(7): 4022-4034, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38622810

ABSTRACT

Mitochondrial dysfunction, a common cellular hallmark in both familial and sporadic forms of Parkinson's disease (PD), is assumed to play a significant role in pathologic development and progression of the disease. Teaghrelin, a unique bioactive compound in some oolong tea varieties, has been demonstrated to protect SH-SY5Y cells against 1-methyl-4-phenylpyridinium induced neurotoxicity by binding to the ghrelin receptor to activate the AMPK/SIRT1/PGC-1α pathway. In this study, an animal model was established using a neurotoxin, 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine (MPTP), a byproduct of a prohibited drug, to evaluate the oral efficacy of teaghrelin on PD by monitoring motor dysfunction of mice in open field, pole, and bean walking tests. The results showed that MPTP-induced motor dysfunction of mice was significantly attenuated by teaghrelin supplementation. Tyrosine hydroxylase and dopamine transporter protein were found reduced in the striatum and midbrain of MPTP-treated mice, and significantly mitigated by teaghrelin supplementation. Furthermore, teaghrelin administration enhanced mitophagy and mitochondria biogenesis, which maintained cell homeostasis and prevented the accumulation of αSyn and apoptosis-related proteins. It seemed that teaghrelin protected dopaminergic neurons in MPTP-treated mice by increasing PINK1/Parkin-mediated mitophagy and AMPK/SIRT1/PGC-1α-mediated mitochondria biogenesis, highlighting its potential therapeutic role in maintaining dopaminergic neurons function in PD. Mitochondrial dysfunction, a common cellular hallmark in both familial and sporadic forms of Parkinson's disease (PD), is assumed to play a significant role in pathologic development and progression of the disease. Teaghrelin, a unique bioactive compound in some oolong tea varieties, has been demonstrated to protect SH-SY5Y cells against 1-methyl-4-phenylpyridinium induced neurotoxicity by binding to the ghrelin receptor to activate the AMPK/SIRT1/PGC-1α pathway. In this study, an animal model was established using a neurotoxin, 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine (MPTP), a byproduct of a prohibited drug, to evaluate the oral efficacy of teaghrelin on PD by monitoring motor dysfunction of mice in open field, pole, and bean walking tests. The results showed that MPTP-induced motor dysfunction of mice was significantly attenuated by teaghrelin supplementation. Tyrosine hydroxylase and dopamine transporter protein were found reduced in the striatum and midbrain of MPTP-treated mice, and significantly mitigated by teaghrelin supplementation. Furthermore, teaghrelin administration enhanced mitophagy and mitochondria biogenesis, which maintained cell homeostasis and prevented the accumulation of αSyn and apoptosis-related proteins. It seemed that teaghrelin protected dopaminergic neurons in MPTP-treated mice by increasing PINK1/Parkin-mediated mitophagy and AMPK/SIRT1/PGC-1α-mediated mitochondria biogenesis, highlighting its potential therapeutic role in maintaining dopaminergic neurons function in PD.


Subject(s)
AMP-Activated Protein Kinases , Dopaminergic Neurons , Ghrelin , Mice, Inbred C57BL , Mitophagy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Kinases , Sirtuin 1 , Ubiquitin-Protein Ligases , Animals , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Sirtuin 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Kinases/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Ghrelin/pharmacology , Male , Mitophagy/drug effects , Mice , AMP-Activated Protein Kinases/metabolism , Disease Models, Animal , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Neuroprotective Agents/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Organelle Biogenesis , Parkinson Disease/drug therapy , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology
3.
Toxicol Mech Methods ; 34(8): 908-919, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38847585

ABSTRACT

In this work, we have analyzed the neuroprotective activity of marrubiin against MPTP-induced Parkinson's disease (PD) in rat brains. MPTP (1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine) a neurotoxin was administered intraperitoneally (i.p.,) to rats and then treated using marrubiin. After marrubiin treatment, rats were trained, and tested for behavioral analyses like cognitive performance, open field test, rotarod test, grip strength test, beam walking test, the status of body weight, and striatal levels of neurotransmitters like dopamine, norepinephrine, serotonin, DOPAC, homovanillic acid, 5-hydroxy indole acetic acid, the status of oxidative stress markers like LPO, protein carbonyl content (PCC), Xanthine oxidase (XO), and status of antioxidant enzyme levels like SOD, CAT, GPX in the striatum and hippocampal tissues, status of neuroinflammatory markers like TNF-α, IL1ß, IL-6, and status of histological architecture in brain striatum were also analyzed. All these parameters were significantly (p < 0.05) abnormal in MPTP-induced rats. Marrubiin (MB) treated shows significant (p < 0.05) near normal behavioral restoration in cognitive performance, open field, rotarod, grip strength, and beam walking tests. Furthermore, the status of body weight, and levels of neurotransmitters, were also significantly (p < 0.05) reversed to near normalcy in marrubiin-treated rats. Similarly, oxidative stress, antioxidant enzyme levels in the striatum and hippocampal tissues, TNF-α, IL1ß, IL-6 levels, and histological architecture were noted to be restored to near normalcy in marrubiin-treated rats. Collectively, our preliminary results highlight the neuroprotective ability of marrubiin. However, the cellular and biochemical mechanisms of marrubiin's neuroprotective ability have to be studied in detail.


Subject(s)
Neuroprotective Agents , Oxidative Stress , Rats, Wistar , Animals , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Rats , Behavior, Animal/drug effects , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , MPTP Poisoning/prevention & control , MPTP Poisoning/metabolism
4.
Neurobiol Dis ; 180: 106067, 2023 05.
Article in English | MEDLINE | ID: mdl-36893901

ABSTRACT

Although Parkinson's disease (PD) key neuropathological hallmarks are well known, the underlying pathogenic mechanisms of the disease still need to be elucidated to identify innovative disease-modifying drugs and specific biomarkers. NF-κB transcription factors are involved in regulating several processes associated with neurodegeneration, such as neuroinflammation and cell death, that could be related to PD pathology. NF-κB/c-Rel deficient (c-rel-/-) mice develop a progressive PD-like phenotype. The c-rel-/- mice present both prodromal and motor symptoms as well as key neuropathological features, including nigrostriatal dopaminergic neurons degeneration, accumulation of pro-apoptotic NF-κB/RelA acetylated at the lysine 310 residue (Ac-RelA(lys310)) and progressive caudo-rostral brain deposition of alpha-synuclein. c-Rel inhibition can exacerbate MPTP-induced neurotoxicity in mice. These findings support the claim that misregulation of c-Rel protein may be implicated in PD pathophysiology. In this study, we aimed at evaluating c-Rel levels and DNA-binding activity in human brains and peripheral blood mononuclear cells (PBMCs) of sporadic PD patients. We analyzed c-Rel protein content and activity in frozen substantia nigra (SN) samples from post-mortem brains of 10 PD patients and 9 age-matched controls as well as in PBMCs from 72 PD patients and 40 age-matched controls. c-Rel DNA-binding was significantly lower and inversely correlated with Ac-RelA(lys310) content in post-mortem SN of sporadic PD cases, when compared to healthy controls. c-Rel DNA-binding activity was also reduced in PBMCs of followed-up PD subjects. The decrease of c-Rel activity in PBMCs from PD patients appeared to be independent from dopaminergic medication or disease progression, as it was evident even in early stage, drug-naïve patients. Remarkably, the levels of c-Rel protein were comparable in PD and control subjects, pointing out a putative role for post-translational modifications of the protein in c-Rel dysfunctions. These findings support that PD is characterized by the loss of NF-κB/c-Rel activity that potentially has a role in PD pathophysiology. Future studies will be aimed at addressing whether the reduction of c-Rel DNA-binding could constitute a novel biomarker for PD.


Subject(s)
MPTP Poisoning , Parkinson Disease , Humans , Mice , Animals , NF-kappa B/metabolism , Parkinson Disease/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Leukocytes, Mononuclear/metabolism , Substantia Nigra/metabolism , Dopaminergic Neurons/metabolism , MPTP Poisoning/pathology
5.
Neurobiol Dis ; 180: 106105, 2023 05.
Article in English | MEDLINE | ID: mdl-36977454

ABSTRACT

Parkinson's disease (PD) is the most common neurodegenerative movement disorder, characterized by the progressive loss of nigrostriatal dopaminergic neurons (DANs), involving the dysregulation of both neurons and glial cells. Cell type- and region-specific gene expression profiles can provide an effective source for revealing the mechanisms of PD. In this study, we adopted the RiboTag approach to obtain cell type (DAN, microglia, astrocytes)- and brain region (substantia nigra, caudate-putamen)-specific translatomes at an early stage in an MPTP-induced mouse model of PD. Through DAN-specific translatome analysis, the glycosphingolipid biosynthetic process was identified as a significantly downregulated pathway in the MPTP-treated mice. ST8Sia6, a key downregulated gene related to glycosphingolipid biosynthesis, was confirmed to be downregulated in nigral DANs from postmortem brains of patients with PD. Specific expression of ST8Sia6 in DANs exerts anti-inflammatory and neuroprotective effects in MPTP-treated mice. Through cell type (microglia vs. astrocyte) and brain region (substantia nigra vs. caudate-putamen) comparisons, nigral microglia showed the most intense immune responses. Microglia and astrocytes in the substantia nigra showed similar levels of activation in interferon-related pathways and interferon gamma (IFNG) was identified as the top upstream regulator in both cell types. This work highlights that the glycosphingolipid metabolism pathway in the DAN is involved in neuroinflammation and neurodegeneration in an MPTP mouse model of PD and provides a new data source for elucidating the pathogenesis of PD.


Subject(s)
MPTP Poisoning , Neurodegenerative Diseases , Neuroprotective Agents , Parkinson Disease , Mice , Animals , Parkinson Disease/metabolism , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Glycosphingolipids/metabolism , Mice, Inbred C57BL , Dopaminergic Neurons/metabolism , Disease Models, Animal , Substantia Nigra/metabolism , MPTP Poisoning/pathology
6.
J Pharmacol Sci ; 152(1): 30-38, 2023 May.
Article in English | MEDLINE | ID: mdl-37059489

ABSTRACT

Parkinson's disease (PD) is characterized by dopaminergic (DAergic) neuronal loss in the substantia nigra pars compacta (SNpc), resulting from α-synuclein (αSyn) toxicity. We previously reported that αSyn oligomerization and toxicity are regulated by the fatty-acid binding protein 3 (FABP3), and the therapeutic effects of the FABP3 ligand, MF1, was successfully demonstrated in PD models. Here, we developed a novel and potent ligand, HY-11-9, which has a higher affinity for FABP3 (Kd = 11.7 ± 8.8) than MF1 (Kd = 302.8 ± 130.3). We also investigated whether the FABP3 ligand can ameliorate neuropathological deterioration after the onset of disease in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism. Motor deficits were observed two weeks after MPTP treatment. Notably, oral administration of HY-11-9 (0.03 mg/kg) improved motor deficits in both beam-walking and rotarod tasks, whereas MF1 failed to improve the motor deficits in both tasks. Consistent with the behavioral tasks, HY-11-9 recovered dopamine neurons from MPTP toxicity in the substantia nigra and ventral tegmental areas. Furthermore, HY-11-9 reduced the accumulation of phosphorylated-serine129-α-synuclein (pS129-αSyn) and colocalization with FABP3 in tyrosine hydroxylase (TH)-positive DA neurons in the PD mouse model. Overall, HY-11-9 significantly improved MPTP-induced behavioral and neuropathological deterioration, suggesting that it may be a potential candidate for PD therapy.


Subject(s)
MPTP Poisoning , Parkinson Disease , Parkinsonian Disorders , Mice , Animals , alpha-Synuclein/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Ligands , Parkinsonian Disorders/drug therapy , Parkinson Disease/drug therapy , Substantia Nigra/metabolism , Substantia Nigra/pathology , Dopaminergic Neurons/metabolism , Mice, Inbred C57BL , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Disease Models, Animal , Fatty Acid Binding Protein 3/metabolism
7.
Int J Mol Sci ; 24(15)2023 Jul 29.
Article in English | MEDLINE | ID: mdl-37569538

ABSTRACT

Mutations in the GBA1 gene represent the major genetic risk factor for Parkinson's disease (PD). The lysosomal enzyme beta-glucocerebrosidase (GCase) encoded by the GBA1 gene participates in both the endolysosomal pathway and the immune response. Disruption of these mechanisms is involved in PD pathogenesis. However, molecular mechanisms of PD associated with GBA1 mutations (GBA-PD) are unknown today in particular due to the partial penetrance of GBA1 variants in PD. The modifiers of GBA1 penetrance have not been elucidated. We characterized the transcriptomic profiles of cells from the substantia nigra (SN) of mice with co-injection with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and selective inhibitor of GCase activity (conduritol-ß-epoxide, (CBE)) to mimic PD bearing GCase dysfunction (MPTP+CBE), mice treated with MPTP, mice treated with CBE and control mice treated with injection of sodium chloride (NaCl) (vehicle). Differential expression analysis, pathway enrichment analysis, and outlier detection were performed. Functional clustering of differentially represented transcripts revealed more processes associated with the functioning of neurogenesis, inflammation, apoptosis and autophagy in MPTP+CBE and MPTP mice than in vehicle mice, with a more pronounced alteration of autophagy processes in MPTP+CBE mice than in MPTP mice. The PI3K-Akt-mTOR signaling pathway may be considered a potential target for therapy in PD with GCase dysfunction.


Subject(s)
MPTP Poisoning , Parkinson Disease , Parkinsonian Disorders , Animals , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Disease Models, Animal , Gene Expression Profiling , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Mice, Inbred C57BL , MPTP Poisoning/pathology , Parkinson Disease/pathology , Parkinsonian Disorders/pathology , Phosphatidylinositol 3-Kinases/metabolism , Substantia Nigra/metabolism
8.
Drug Chem Toxicol ; 45(2): 947-954, 2022 Mar.
Article in English | MEDLINE | ID: mdl-32693643

ABSTRACT

1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxin that damages dopaminergic neurons. Zebrafish has been shown to be a suitable model organism to investigate the molecular pathways in the pathogenesis of Parkinson's disease and also for potential therapeutic agent research. Boron has been shown to play an important role in the neural activity of the brain. Boronic acids are used in combinatorial approaches in drug design and discovery. The effect of 3-pyridinylboronic acid which is an important sub-class of heterocyclic boronic acids has not been evaluated in case of MPTP exposure in zebrafish embryos. Accordingly, this study was designed to investigate the effects of 3-pyridinylboronic acid on MPTP exposed zebrafish embryos focusing on the molecular pathways related to neurodegeneration and apoptosis by RT-PCR. Zebrafish embryos were exposed to MPTP (800 µM); MPTP + Low Dose 3-Pyridinylboronic acid (50 µM) (MPTP + LB) and MPTP + High Dose 3-Pyridinylboronic acid (100 µM) (MPTP + HB) in well plates for 72 hours post fertilization. Results of our study showed that MPTP induced a P53 dependent and Bax mediated apoptosis in zebrafish embryos and 3-pyridinylboronic acid restored the locomotor activity and gene expressions related to mitochondrial dysfunction and oxidative stress due to the deleterious effects of MPTP, in a dose-dependent manner.


Subject(s)
MPTP Poisoning , Zebrafish , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Boronic Acids/metabolism , Boronic Acids/therapeutic use , Disease Models, Animal , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Mice , Mice, Inbred C57BL , Pyridines , Pyrrolidines/metabolism , Pyrrolidines/therapeutic use , Zebrafish/metabolism
9.
Int J Mol Sci ; 23(18)2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36142491

ABSTRACT

Neurodegenerative diseases such as Parkinson's disease (PD) are known to be related to oxidative stress and neuroinflammation, and thus, modulating neuroinflammation offers a possible means of treating PD-associated pathologies. Morin (2',3,4',5,7-pentahydroxy flavone) is a flavonol with anti-oxidative and anti-inflammatory effects found in wines, herbs, and fruits. The present study was undertaken to determine whether a morin-containing diet has protective effects in an MPTP-induced mouse model of PD. Mice were fed a control or morin diet for 34 days, and then MPTP (30 mg/kg, i.p.) was administered daily for 5 days to induce a PD-like pathology. We found that dietary morin prevented MPTP-induced motor dysfunction and ameliorated dopaminergic neuronal damage in striatum (STR) and substantia nigra (SN) in our mouse model. Furthermore, MPTP-induced neuroinflammation was significantly reduced in mice fed morin. In vitro studies showed that morin effectively suppressed glial activations in primary microglia and astrocytes, and biochemical analysis and a docking simulation indicated that the anti-inflammatory effects of morin were mediated by blocking the extracellular signal-regulated kinase (ERK)-p65 pathway. These findings suggest that morin effectively inhibits glial activations and has potential use as a functional food ingredient with therapeutic potential for the treatment of PD and other neurodegenerative diseases associated with neuroinflammation.


Subject(s)
Flavones , Food Ingredients , MPTP Poisoning , Neuroprotective Agents , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Dopaminergic Neurons/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavones/pharmacology , Flavonols/metabolism , Flavonols/pharmacology , Flavonols/therapeutic use , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/complications , Parkinson Disease/etiology
10.
J Neurochem ; 157(3): 727-751, 2021 05.
Article in English | MEDLINE | ID: mdl-33264426

ABSTRACT

Parkinson disease (PD) is the second most common neurodegenerative disorder, whose prevalence is 2~3% in the population over 65. α-Synuclein aggregation is the major pathological hallmark of PD. However, recent studies have demonstrated enhancing evidence of tau pathology in PD. Despite extensive considerations, thus far, the actual spreading mechanism of neurodegeneration has remained elusive in a PD brain. This study aimed to further investigate the development of α-synuclein and tau pathology. We employed various PD models, including cultured neurons treated with either 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or with recombinant α-synuclein. Also, we studied dopaminergic neurons of cytokine Interferon-ß knock-out. Moreover, we examined rats treated with 6-hydroxydopamine, Rhesus monkeys administrated with MPTP neurotoxin, and finally, human post-mortem brains. We found the α-synuclein phosphorylation triggers tau pathogenicity. Also, we observed more widespread phosphorylated tau than α-synuclein with prion-like nature in various brain areas. We optionally removed P-tau or P-α-synuclein from cytokine interferon-ß knock out with respective monoclonal antibodies. We found that tau immunotherapy suppressed neurodegeneration more than α-synuclein elimination. Our findings indicate that the pathogenic tau could be one of the leading causes of comprehensive neurodegeneration triggered by PD. Thus, we can propose an efficient therapeutic target to fight the devastating disorder.


Subject(s)
Brain/pathology , Parkinson Disease/pathology , Tauopathies/pathology , alpha-Synuclein/genetics , Animals , Autopsy , Behavior, Animal , Cell Survival/drug effects , Dopaminergic Neurons/drug effects , Female , Humans , Interferon-beta/genetics , MPTP Poisoning/pathology , Macaca mulatta , Male , Mice , Mice, Knockout , Parkinson Disease/psychology , Pregnancy , Rats , Rats, Wistar , Recombinant Proteins , tau Proteins/biosynthesis , tau Proteins/genetics
11.
J Biochem Mol Toxicol ; 35(1): e22631, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32926752

ABSTRACT

Parkinson's disease (PD) is a complex neurodegenerative illness associated with the loss or damage to neurons of the dopaminergic system in the brain. Few therapeutic approaches and considerable side effects of conventional drugs necessitate a new therapeutic agent to treat patients with PD. Rhaponticin is a natural hydroxystilbene, found in herbal plants such as Rheum rhaponticum, and known to have desirable biological activity including anti-inflammatory properties. However, the neuroinflammation on rhaponticin levels has only been investigated partially so far. So, the current study explored whether rhaponticin could ameliorate the pathophysiology observed in both the in vitro microglial BV-2 cells and the in vivo (1-methyl-4-phenyl-1,2,3,5-tetrahydropyridine [MPTP])-mediated PD model. The results show rhaponticin significantly attenuated lipopolysaccharide (LPS)-mediated microglial activation by suppressing nitric oxide synthase in conjunction with abridged reactive oxygen species production together with proinflammatory mediator reduction. In vivo rhaponticin treatment improves motor impairments as well as the loss of dopaminergic neurons in MPTP-treated mice possibly through suppression via mediators of inflammation. Taken together, these results offer evidence that rhaponticin exerts anti-inflammatory effects and neuroprotection in an LPS-induced microglial model and the MPTP-induced mouse models of PD.


Subject(s)
Dopaminergic Neurons/metabolism , MPTP Poisoning/prevention & control , Neuroprotective Agents/pharmacology , Stilbenes/pharmacology , Animals , Cell Line, Transformed , Dopaminergic Neurons/pathology , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Mice
12.
J Biochem Mol Toxicol ; 35(3): e22685, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33368846

ABSTRACT

Parkinsonism is a neurodegenerative disease that is common all over the world. This study aimed at exploring the neuroprotective effect of tiron against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism. MPTP (30 mg/kg, intraperitoneally [ip]) was injected in mice daily for 5 consecutive days. Mice were treated with tiron (140 and 280 mg/kg, ip) or levodopa (8.4 mg/kg, orally) for 10 consecutive days starting 5 days before MPTP injection. At the end of the experiment, behavioral tests were conducted to assess the neuroprotective effect of tiron. Moreover, oxidative stress was assessed via measuring antioxidant enzyme, such as catalase, and lipid peroxidation was evaluated as malondialdehyde. Neuronal damage was also detected by histopathological examination and via estimating hippocampal levels of dopamine, γ-aminobutyric acid, and nuclear factor erythroid-derived 2-like 2. In addition, the expression of Kelch-like ECH-associated protein 1 and heme oxygenase-1 was assessed by immunohistochemistry. Compared with the blank control group and the positive control group, the inhibitory effect of tiron on MPTP-induced neurodegenerative injury was statistically significant.


Subject(s)
1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , MPTP Poisoning , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Animals , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Mice , Oxidative Stress/drug effects
13.
Acta Biochim Biophys Sin (Shanghai) ; 53(7): 870-882, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33984130

ABSTRACT

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.


Subject(s)
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
14.
Proc Natl Acad Sci U S A ; 115(30): E7193-E7201, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29987020

ABSTRACT

Parkin functions as a multipurpose E3 ubiquitin ligase, and Parkin loss of function is associated with both sporadic and familial Parkinson's disease (PD). We report that the Bin/Amphiphysin/Rvs (BAR) domain of protein interacting with PRKCA1 (PICK1) bound to the really interesting new gene 1 (RING1) domain of Parkin and potently inhibited the E3 ligase activity of Parkin by disrupting its interaction with UbcH7. Parkin translocated to damaged mitochondria and led to their degradation in neurons, whereas PICK1 robustly inhibited this process. PICK1 also impaired the protective function of Parkin against stresses in SH-SY5Y cells and neurons. The protein levels of several Parkin substrates were reduced in young PICK1-knockout mice, and these mice were resistant to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated toxicity. Taken together, the results indicate that PICK1 is a potent inhibitor of Parkin, and the reduction of PICK1 enhances the protective effect of Parkin.


Subject(s)
Carrier Proteins/metabolism , MPTP Poisoning/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line, Tumor , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Protein Domains , Ubiquitin-Protein Ligases/genetics
15.
Proc Natl Acad Sci U S A ; 115(25): E5815-E5823, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29735655

ABSTRACT

Parkinson's disease (PD) is characterized as a chronic and progressive neurodegenerative disorder, and the deposition of specific protein aggregates of α-synuclein, termed Lewy bodies, is evident in multiple brain regions of PD patients. Although there are several available medications to treat PD symptoms, these medications do not prevent the progression of the disease. Soluble epoxide hydrolase (sEH) plays a key role in inflammation associated with the pathogenesis of PD. Here we found that MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced neurotoxicity in the mouse striatum was attenuated by subsequent repeated administration of TPPU, a potent sEH inhibitor. Furthermore, deletion of the sEH gene protected against MPTP-induced neurotoxicity, while overexpression of sEH in the striatum significantly enhanced MPTP-induced neurotoxicity. Moreover, the expression of the sEH protein in the striatum from MPTP-treated mice or postmortem brain samples from patients with dementia of Lewy bodies (DLB) was significantly higher compared with control groups. Interestingly, there was a positive correlation between sEH expression and phosphorylation of α-synuclein in the striatum. Oxylipin analysis showed decreased levels of 8,9-epoxy-5Z,11Z,14Z-eicosatrienoic acid in the striatum of MPTP-treated mice, suggesting increased activity of sEH in this region. Interestingly, the expression of sEH mRNA in human PARK2 iPSC-derived neurons was higher than that of healthy control. Treatment with TPPU protected against apoptosis in human PARK2 iPSC-derived dopaminergic neurons. These findings suggest that increased activity of sEH in the striatum plays a key role in the pathogenesis of neurodegenerative disorders such as PD and DLB. Therefore, sEH may represent a promising therapeutic target for α-synuclein-related neurodegenerative disorders.


Subject(s)
Epoxide Hydrolases/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Line , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , HEK293 Cells , Humans , Lewy Bodies/drug effects , Lewy Bodies/metabolism , Lewy Bodies/pathology , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , RNA, Messenger/metabolism , alpha-Synuclein/metabolism
16.
Int J Mol Sci ; 22(9)2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33919373

ABSTRACT

The diurnal rodent Octodon degus (O. degus) is considered an attractive natural model for Alzheimer's disease and other human age-related features. However, it has not been explored so far if the O. degus could be used as a model to study Parkinson's disease. To test this idea, 10 adult male O. degus were divided into control group and MPTP-intoxicated animals. Motor condition and cognition were examined. Dopaminergic degeneration was studied in the ventral mesencephalon and in the striatum. Neuroinflammation was also evaluated in the ventral mesencephalon, in the striatum and in the dorsal hippocampus. MPTP animals showed significant alterations in motor activity and in visuospatial memory. Postmortem analysis revealed a significant decrease in the number of dopaminergic neurons in the ventral mesencephalon of MPTP animals, although no differences were found in their striatal terminals. We observed a significant increase in neuroinflammatory responses in the mesencephalon, in the striatum and in the hippocampus of MPTP-intoxicated animals. Additionally, changes in the subcellular expression of the calcium-binding protein S100ß were found in the astrocytes in the nigrostriatal pathway. These findings prove for the first time that O. degus are sensitive to MPTP intoxication and, therefore, is a suitable model for experimental Parkinsonism in the context of aging.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Behavior, Animal/drug effects , Disease Models, Animal , Inflammation/pathology , MPTP Poisoning/pathology , Neurotoxins/toxicity , Parkinsonian Disorders/pathology , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Inflammation/etiology , MPTP Poisoning/etiology , Male , Neostriatum/drug effects , Neostriatum/pathology , Octodon , Parkinsonian Disorders/etiology
17.
Molecules ; 26(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34208814

ABSTRACT

Parkinson's disease (PD) is one of the most common neurodegenerative diseases and affects approximately 6.3 million people worldwide. To date, the treatment of PD remains a challenge, as available treatment options are known to be associated with serious side effects; hence, the search for new treatment strategies is critical. Extracts from the Amaryllidaceae plant family as well as their alkaloids have been reported to have neuroprotective potentials. This study, therefore, investigated the biological activities of Crossyne flava and its isolated alkaloids in an in vitro MPP+ (1-methyl-4-phenylpyridinium) PD model using SH-SY5Y cells. The effects of the total extract as well as the four compounds isolated from Crossyne flava (i.e., pancratinine B (1), bufanidrine (2), buphanisine (3), and epibuphanisine (4)) were evaluated for cell viability, neuroprotection, levels of reactive oxygen species (ROS), adenosine triphosphate activity (ATP), and caspase 3/7 activity in SH-SY5Y cells. The results obtained showed that pre-treatment with both the extract and the isolated compounds was effective in protecting the SH-SY5Y cells from MPP+-induced neurotoxicity and inhibited ROS generation, ATP depletion as well as apoptosis induction in the SH-SY5Y cells. The results of this study show that the Amaryllidaceae plant family may be a source of novel compounds for the treatment of neurodegenerative diseases, which validates the reported traditional uses.


Subject(s)
Amaryllidaceae Alkaloids/pharmacology , Amaryllidaceae/chemistry , MPTP Poisoning/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Amaryllidaceae Alkaloids/chemistry , Amaryllidaceae Alkaloids/isolation & purification , Cell Line, Tumor , Humans , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Neuroprotection/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification
18.
Cell Mol Life Sci ; 76(23): 4725-4743, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31359086

ABSTRACT

Targeted cell ablation is a powerful approach for studying the role of specific cell populations in a variety of organotypic functions, including cell differentiation, and organ generation and regeneration. Emerging tools for permanently or conditionally ablating targeted cell populations and transiently inhibiting neuronal activities exhibit a diversity of application and utility. Each tool has distinct features, and none can be universally applied to study different cell types in various tissue compartments. Although these tools have been developed for over 30 years, they require additional improvement. Currently, there is no consensus on how to select the tools to answer the specific scientific questions of interest. Selecting the appropriate cell ablation technique to study the function of a targeted cell population is less straightforward than selecting the method to study a gene's functions. In this review, we discuss the features of the various tools for targeted cell ablation and provide recommendations for optimal application of specific approaches.


Subject(s)
Bacteriocins/metabolism , Clodronic Acid/chemistry , Diphtheria Toxin/genetics , Optogenetics/methods , Simplexvirus/physiology , Animals , Clodronic Acid/toxicity , Diphtheria Toxin/metabolism , Humans , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , Neurons/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Simplexvirus/enzymology
19.
J Cell Physiol ; 234(12): 23379-23387, 2019 12.
Article in English | MEDLINE | ID: mdl-31232472

ABSTRACT

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.


Subject(s)
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.
J Neurochem ; 148(1): 63-79, 2019 01.
Article in English | MEDLINE | ID: mdl-30295916

ABSTRACT

Glial pathology precedes symptoms of Parkinson's disease and multiple other neurodegenerative diseases. Prolonged impairment of astrocytic functions could increase the vulnerability of dopaminergic neurons in the substantia nigra (SN), accelerate their degeneration and affect ability to compensate for partial degeneration at the presymptomatic stages of the disease. The aim of this study was to investigate the astrocyte depletion in the SN, its impact on the dopaminergic system functioning and multiple markers of energy metabolism during the early stages of neurodegeneration and compensation. We induced death of 30% of astrocytes by chronic infusion of fluorocitrate (FC) into the SN, simultaneously activating microglia response but sparing the dopaminergic neurons. The FC effect was reversible after toxin withdrawal. Dopaminergic neurons were killed by 6-hydroxydopamine causing transient locomotor disability, reversed with time showing compensatory potential. Death of astrocytes diminished the capability of the dopaminergic system to compensate for the degeneration of neurons and caused a local energy deprivation by decreasing lactate and glycogen amount. Studied markers suggest a shift in the usage of energy substrates, via increased glycogenolysis and glycolysis markers, ketone bodies availability and fatty acid transport in remaining cells. Peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1alpha) and AMP-activated protein kinase (AMPK), the energy sensors, showed different regulation between the cell-types. Increased neuronal expression of carnitine palmitoyltransferase 1c could play a role in the adaptation to metabolic stress in response to glia dysfunction. Astrocyte energetic support is one of the essential factors for neuronal compensatory mechanisms of dopaminergic system and might have a leading role in the presymptomatic Parkinson's disease stages. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Subject(s)
Astrocytes/metabolism , MPTP Poisoning/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Substantia Nigra/metabolism , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Energy Metabolism/physiology , MPTP Poisoning/pathology , Male , Nerve Degeneration/pathology , Neurons/pathology , Rats , Rats, Wistar , Substantia Nigra/pathology
SELECTION OF CITATIONS
SEARCH DETAIL