Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Phytomedicine ; 127: 155494, 2024 May.
Article in English | MEDLINE | ID: mdl-38471370

ABSTRACT

BACKGROUND: Parkinson's disease (PD), a neurodegenerative disorder, is characterized by motor symptoms due to the progressive loss of dopaminergic neurons in the substantia nigra (SN) and striatum (STR), alongside neuroinflammation. Asiaticoside (AS), a primary active component with anti-inflammatory and neuroprotective properties, is derived from Centella asiatica. However, the precise mechanisms through which AS influences PD associated with inflammation are not yet fully understood. PURPOSE: This study aimed to explore the protective mechanism of AS in PD. METHODS: Targets associated with AS and PD were identified from the Swiss Target Prediction, Similarity Ensemble Approach, PharmMapper, and GeneCards database. A protein-protein interaction (PPI) network was constructed to identify potential therapeutic targets. Concurrently, GO and KEGG analyses were performed to predict potential signaling pathways. To validate these mechanisms, the effects of AS on 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD in mice were investigated. Furthermore, neuroinflammation and the activation of the NLRP3 inflammasome were assessed to confirm the anti-inflammatory properties of AS. In vitro experiments in BV2 cells were then performed to investigate the mechanisms of AS in PD. Moreover, CETSA, molecular docking, and molecular dynamics simulations (MDs) were performed for further validation. RESULTS: Network pharmacology analysis identified 17 potential targets affected by AS in PD. GO and KEGG analyses suggested the biological roles of these targets, demonstrating that AS interacts with 149 pathways in PD. Notably, the NOD-like receptor signaling pathway was identified as a key pathway mediating AS's effect on PD. In vivo studies demonstrated that AS alleviated motor dysfunction and reduced the loss of dopaminergic neurons in MPTP-induced PD mice. In vitro experiments demonstrated that AS substantially decreased IL-1ß release in BV2 cells, attributing this to the modulation of the NLRP3 signaling pathway. CETSA and molecular docking studies indicated that AS forms a stable complex with NLRP3. MDs suggested that ARG578 played an important role in the formation of the complex. CONCLUSION: In this study, we first predicted that the potential target and pathway of AS's effect on PD could be NLRP3 protein and NOD-like receptor signaling pathway by network pharmacology analysis. Further, we demonstrated that AS could alleviate symptoms of PD induced by MPTP through its interaction with the NLRP3 protein for the first time by in vivo and in vitro experiments. By binding to NLRP3, AS effectively inhibits the assembly and activation of the inflammasome. These findings suggest that AS is a promising inhibitor for PD driven by NLRP3 overactivation.


Subject(s)
MPTP Poisoning , Neuroprotective Agents , Parkinson Disease , Triterpenes , Mice , Animals , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , Neuroprotection , Neuroinflammatory Diseases , Molecular Docking Simulation , Microglia , Parkinson Disease/metabolism , Dopaminergic Neurons , Anti-Inflammatory Agents/therapeutic use , Mice, Inbred C57BL , Disease Models, Animal , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
2.
J Nutr Biochem ; 126: 109586, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38262563

ABSTRACT

Parkinson's disease (PD) is an incurable neurological disorder that causes typical motor deficits. In this study, we investigated the effects of creatine supplementation and exercise in the subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. We found that 2% creatine supplementation and/or exercise intervention for 4 weeks elicited neurobehavioral recovery and neuroprotective effects regarding dopaminergic cell loss in MPTP-treated mice; this effect implies functional preservation of dopaminergic cells in the substantia nigra, as reflected by tyrosine hydroxylase expression recovery. Creatine and exercise reduced necroptotic activity in dopaminergic cells by lowering mixed lineage kinase domain-like protein (MLKL) modification to active phenotypes (phosphorylation at Ser345 and oligomerization) and phosphorylated receptor-interacting protein kinase 1 (RIPK1) (Ser166-p) and RIPK3 (Ser232-p) levels. In addition, creatine and exercise reduced the MPTP-induced increase in pathogenic α-synuclein forms, such as Ser129 phosphorylation and oligomerization. Furthermore, creatine and exercise had anti-inflammatory and antioxidative effects in MPTP mice, as evidenced by a decrease in microglia activation, NF-κB-dependent pro-inflammatory molecule expression, and increase in antioxidant enzyme expression. These phenotypic changes were associated with the exercise/creatine-induced AMP-activated protein kinase (AMPK)/nuclear factor erythroid 2-related factor 2 (Nrf2) and sirtuin 3 (SIRT3)/forkhead box O3 (FoxO3a) signaling pathways. In all experiments, combining creatine with exercise resulted in considerable improvement over either treatment alone. Consequently, these findings suggest that creatine supplementation with exercise has anti-inflammatory, antioxidative, and anti-α-synucleinopathy effects, thereby reducing necroptotic cell death in a PD mouse model.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Mice , Animals , Parkinson Disease/drug therapy , alpha-Synuclein/metabolism , Creatine/pharmacology , Creatine/therapeutic use , Necroptosis , Dopaminergic Neurons/metabolism , Neuroprotective Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Dietary Supplements , Mice, Inbred C57BL , Disease Models, Animal , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism
3.
Phytomedicine ; 124: 155309, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38237261

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative condition characterized by the progressive loss of dopaminergic neurons within the substantia nigra. Neuroinflammation plays a pivotal role in the pathogenesis of PD, involving the activation of microglia cells, heightened production of proinflammatory cytokines, and perturbations in the composition of the gut microbiota. Rubusoside (Ru), the principal steviol bisglucoside present in Rubus chingii var. suavissimus (S.K.Lee) L.T.Lu (Rosaceae), has been documented for its anti-inflammatory properties in diverse disease models. Nonetheless, there is an imperative need to comprehensively assess and elucidate the protective and anti-inflammatory attributes of Ru concerning PD, as well as to uncover the underlying mechanism involved. OBJECTIVE: The aim of this study is to evaluate the neuroprotective and anti-inflammatory effects of Ru on PD and investigate its potential mechanisms associated with microbes. RESEARCH DESIGN AND METHODS: We pre-treated mice and cell lines with Ru in order to simulate the progression of PD and the neuroinflammatory state. The mouse model was induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), SN4741 cells were induced by 1-methyl-4-phenylpyridine (mpp+), and BV-2 cells were induced by lipopolysaccharide (LPS). We assessed the impact of Ru on motor function, neuroinflammation, neuron apoptosis, the composition of gut microbes, and their metabolites. RESULTS: Ru treatment reduces the release of pro-inflammatory mediators by inhibiting microglia activation. It also prevents neuronal apoptosis, thereby safeguarding dopaminergic neurons and ameliorating motor dysfunction. Furthermore, it induces alterations in the fecal microbiota composition and metabolites profile in PD mice. In vitro experiments have demonstrated that Ru inhibits neuronal apoptosis in SN4741 cells induced by mpp+, suppresses the production of pro-inflammatory mediators, and activates the c-Jun N-terminal kinase (JNK), mitogen-activated protein kinase (p38 MAPK), and nuclear factor kappa-B (NF-κB) signaling pathways. CONCLUSION: Ru exhibits inhibitory effects on the MPTP-induced PD model by mitigating neuroinflammation and neuronal apoptosis while also inducing changes in the gut microbiota and metabolite composition.


Subject(s)
Diterpenes, Kaurane , Gastrointestinal Microbiome , Glucosides , Neuroprotective Agents , Parkinson Disease , Mice , Animals , Parkinson Disease/metabolism , Neuroinflammatory Diseases , Anti-Inflammatory Agents/therapeutic use , 1-Methyl-4-phenylpyridinium , Apoptosis , Inflammation Mediators/metabolism , Dopaminergic Neurons , Mice, Inbred C57BL , Disease Models, Animal , Microglia , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
4.
Phytomedicine ; 116: 154866, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37209604

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is one of the most common neurodegenerative diseases in the world. Mitophagy has been implicated in PD etiology for decades and its pharmacological activation is recognized as a promising treatment strategy for PD. For mitophagy initiation, low mitochondrial membrane potential (ΔΨm) is essential. We identified a natural compound morin that could induce mitophagy without affecting ΔΨm. Morin is a flavonoid that can be isolated from fruits like mulberry. PURPOSE: To reveal the effect of morin on the PD mice model and their potential underlying molecular mechanism. METHODS: Mitophagy process induced by morin in N2a cells meditation were measured using flow cytometry and immunofluorescence. JC-1 fluorescence dye used to detect the mitochondrial membrane potential (ΔΨm). The TFEB nuclear translocation were examined by immunofluorescence staining and western blot assay. The PD mice model was induced by MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) intraperitoneal administration. RESULTS: We found that morin also promoted nuclear translocation of the mitophagy regulator TFEB and activated the AMPK-ULK1 pathway. In MPTP-induced PD in vivo models, morin protected DA neurons from MPTP neurotoxicity and ameliorated behavioral deficit. CONCLUSION: Although morin was previously reported to be neuroprotective in PD, the detailed molecular mechanisms remain to be elucidated. For the first time, we report morin served as a novel and safe mitophagy enhancer underlying AMPK-ULK1 pathway and exhibited anti-Parkinsonian effects indicating its potential as a clinical drug for PD treatment.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Mice , Animals , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Neuroprotective Agents/therapeutic use , AMP-Activated Protein Kinases/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Mitophagy , Flavonoids/pharmacology , Flavonoids/therapeutic use , Mice, Inbred C57BL , Disease Models, Animal , Dopaminergic Neurons
5.
J Nutr ; 153(1): 167-175, 2023 01.
Article in English | MEDLINE | ID: mdl-36913450

ABSTRACT

BACKGROUND: Circulating zinc (Zn) concentrations are lower than normal in patients with Parkinson disease (PD). It is unknown whether Zn deficiency increases the susceptibility to PD. OBJECTIVES: The study aimed to investigate the effect of dietary Zn deficiency on behaviors and dopaminergic neurons in a mouse model of PD and to explore potential mechanisms. METHODS: Male C57BL/6J mice aged 8-10 wk were fed Zn adequate (ZnA; 30 µg/g) or Zn deficient (ZnD; <5 µg/g) diet throughout the experiments. Six weeks later 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was injected to generate the PD model. Controls were injected with saline. Thus, 4 groups (Saline-ZnA, Saline-ZnD, MPTP-ZnA, and MPTP-ZnD) were formed. The experiment lasted 13 wk. Open field test, rotarod test, immunohistochemistry, and RNA sequencing were performed. Data were analyzed with t-test, 2-factor ANOVA, or Kruskal-Wallis test. RESULTS: Both MPTP and ZnD diet treatments led to a significant reduction in blood Zn concentrations (PMPTP = 0.012, PZn = 0.014), reduced total distance traveled (PMPTP < 0.001, PZn = 0.031), and affected the degeneration of dopaminergic neurons in the substantia nigra (PMPTP < 0.001, PZn = 0.020). In the MPTP-treated mice, the ZnD diet significantly reduced total distance traveled by 22.4% (P = 0.026), decreased latency to fall by 49.9% (P = 0.026), and reduced dopaminergic neurons by 59.3% (P = 0.002) compared with the ZnA diet. RNA sequencing analysis revealed a total of 301 differentially expressed genes (156 upregulated; 145 downregulated) in the substantia nigra of ZnD mice compared with ZnA mice. The genes were involved in a number of processes, including protein degradation, mitochondria integrity, and α-synuclein aggregation. CONCLUSIONS: Zn deficiency aggravates movement disorders in PD mice. Our results support previous clinical observations and suggest that appropriate Zn supplementation may be beneficial for PD.


Subject(s)
Malnutrition , Parkinson Disease , Mice , Male , Animals , Parkinson Disease/metabolism , Dopaminergic Neurons/metabolism , Mice, Inbred C57BL , Diet , Dopamine/metabolism , Zinc , Substantia Nigra/metabolism , Disease Models, Animal , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology
6.
Chem Biodivers ; 20(3): e202200926, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36661244

ABSTRACT

Qilong capsule (QLC) originates from the famous "Buyang Huanwu decoction" prescription. It is representative of drugs used in China during recovery from stroke, but its neuroprotective mechanism of action remains obscure. HPLC was used to evaluate the similarity of 10 batches of QLC samples. Then we used a zebrafish model to study the neuroprotective effect of QLC. At 24 hpf, embryos were treated with QLC and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and zebrafish were observed the neuronal length and the number of apoptotic cells in the brain at 72 hpf. At 120 hpf, we conduct zebrafish behavioural tests. We then also used qPCR to detect the expression of genes related to autophagy and apoptosis. The results showed that QLC significantly reduced the damage of dopaminergic neurons, the number of apoptotic cells in the brain, and alleviated motor disturbances induced by MPTP. We found that the mechanism of QLC activity involved decreased neuron cell death by inhibiting mitochondrial apoptosis and autophagy, promoting autophagy, degradation of alpha-synuclein, and neuron cell growth, and rescuing the function of neurons damaged by MPTP. The results indicated that QLC protected against MPTP-induced neuron injury and provided pharmacological evidence for clinical use of QLC.


Subject(s)
Drugs, Chinese Herbal , Neuroprotective Agents , Zebrafish , Animals , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Disease Models, Animal , Dopaminergic Neurons/metabolism , Drugs, Chinese Herbal/pharmacology , Neuroprotective Agents/therapeutic use
7.
Phytomedicine ; 108: 154512, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36288652

ABSTRACT

BACKGROUND: Feruloylated oligosaccharides (FOs) are natural esterification products of ferulic acid and oligosaccharides. STUDY DESIGN: In this study, we examined whether FOs contribute to the ensured survival of nigrostriatal dopamine neurons and inhibition of neuroinflammation in Parkinson's disease (PD). METHODS: 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP, 30 mg/kg) was injected intraperitoneally into mice to establish a Parkinson's disease (PD) mouse model. FOs (15 and 30 mg/kg) were orally administered daily to the MPTP-treated mice. The rotarod test, balance beam test, immunofluorescence, enzyme-linked immunosorbent assay (ELISA), quantitative PCR (qPCR), and western blot analyses were performed to examine the neuroprotective effects of FOs on MPTP-treated mice. RESULTS: Our study indicated that FOs increased the survival of dopamine neurons in the substantia nigra pars compacta (SNc) of the MPTP-treated mice. The neuroprotective effects of FOs were accompanied by inhibited glial activation and reduced inflammatory cytokine production. The mechanistic experiments revealed that the neuroprotective effects of FOs might be mediated through the activation of the ERK/CREB/BDNF/TrkB signalling pathway. CONCLUSION: This study provides new insights into the mechanism underlying the anti-neuroinflammatory effect of phytochemicals and may facilitate the development of dietary supplements for PD patients. Our results indicate that FOs can be used as potential modulators for the prevention and treatment of PD.


Subject(s)
MPTP Poisoning , Neuroprotective Agents , Parkinson Disease , Mice , Animals , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Mice, Inbred C57BL , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , MPTP Poisoning/prevention & control , Dopaminergic Neurons , Disease Models, Animal , Oligosaccharides/pharmacology
8.
Metab Brain Dis ; 38(2): 557-571, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36401682

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by both motor and non-motor features. The current treatment regimen for PD are dopamine enhancers which have been reported to worsen the disease prognosis after long term treatment, thus, the need for better treatment options. This study sought to investigate the protective action of Double Stem Cell® (DSC), a blend of stem cells extracts from Swiss apples (Malus Domestica) and Burgundy grapes (Vitis vinifera) on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinsonism in mice and genetic model of PD in Drosophila melanogaster. Male albino mice were pretreated with MPTP (4 × 20 mg/kg, i.p., two hourly in 8 h), twelve hours before administration of DSC (8, 40, or 200 mg/kg, p.o.). Thereafter, behavioural, biochemical and immunohistochemical assays were carried out. The impact of vehicle or DSC supplementation on α-synuclein aggregation was evaluated in Drosophila melanogaster using the UAS-Gal4 system, female DDC-Gal4 flies were crossed with male UAS-α-synuclein, the progenies were examined for fecundity, locomotion, memory, and lifespan. MPTP-induced motor deficits in open field test (OFT), working memory impairment (Y-maze test (YMT)) and muscle incoordination (rotarod test) were ameliorated by DSC (8, 40 or 200 mg/kg) through dose-dependent and significant improvements in motor, cognitive and motor coordination. Moreso, MPTP exposure caused significant increase in lipid peroxidation and decrease in antioxidant enzymes activities (glutathione, catalase and superoxide dismutase) in the midbrain which were attenuated by DSC. MPTP-induced expression of microglia (iba-1), astrocytes (glia fibrillary acidic protein; GFAP) as well as degeneration of dopamine neurons (tyrosine hydroxylase positive neurons) in the substantia nigra (SN) were reversed by DSC. Supplementation of flies feed with graded concentration of DSC (0.8, 4 or 20 mg/ml) did not affect fecundity but improved climbing activity and lifespan. Findings from this study showed that Double Stem Cell improved motor and cognitive functions in both mice and Drosophila through attenuation of neurotoxin-induced oxidative stress and neuroinflammation.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Plant Extracts , Animals , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , alpha-Synuclein/metabolism , Antioxidants/pharmacology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Drosophila melanogaster , Mice, Inbred C57BL , Models, Genetic , Neuroinflammatory Diseases , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Plant Extracts/pharmacology
9.
Phytomedicine ; 104: 154250, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35752074

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disorder involving the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Cellular clearance mechanisms, including the autophagy-lysosome pathway, are commonly affected in the pathogenesis of PD. The lysosomal Ca2+ channel mucolipin TRP channel 1 (TRPML1) is one of the most important proteins involved in the regulation of autophagy. Artemisia argyi Lev. et Vant., is a traditional Chinese herb, that has diverse therapeutic properties and is used to treat patients with skin diseases and oral ulcers. However, the neuroprotective effects of A. argyi are not explored yet. HYPOTHESIS: This study aims is to investigate the neuroprotective effects of A. argyi in promoting the TRPML1-mediated autophagy/mitophagy-enhancing effect METHODS: In this study, we used 1-methyl-4-phenyl-pyridinium (MPP+)-induced PD model established in an SH-SY5Y human neuroblastoma cell line as well as in a 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine (MPTP)-induced PD model in C57BL/6 J mice. MTT assay was conducted to measure the cell viability and further MitoSoX and DCFDA assay were used to measure the ROS. Western blot analysis was used to access levels of TRPML1, p-DRP1 (ser616), p-AKT, PI3K, and ß-catenin, Additionally, IF and IHC analysis to investigate the expression of TRPML1, LC3B, ß-catenin, TH+, α-synuclein. Mitotracker stain was used to check mitophagy levels and a lysosomal intracellular activity kit was used to measure the lysosomal dysfunction. Behavioral studies were conducted by rotarod and grip strength experiments to check motor functions. RESULTS: In our in vitro study, A. argyi rescued the MPP+-induced loss of cell viability and reduced the accumulation of mitochondrial and total reactive oxygen species (ROS). Subsequently, it increased the expression of TRPML1 protein, thereby inducing autophagy, which facilitated the clearance of toxic accumulation of α-synuclein. Furthermore, A. argyi played a neuroprotective role by activating the PI3K/AKT/ß-catenin cell survival pathway. MPP+-mediated mitochondrial damage was overcome by upregulation of mitophagy and downregulation of the mitochondrial fission regulator p-DRP1 (ser616) in SH-SY5Y cells. In the in vivo study, A. argyi ameliorated impaired motor function and rescued TH+ neurons in the SNpc region. Similar to the results of the in vitro study, TRPML1, LC3B, and ß-catenin expression was enhanced in the SNpc region in the A. argyi-treated mice brain. CONCLUSION: Thus, our results first demonstrate that A. argyi can exert neuroprotective effects by stimulating TRPML1 and rescuing neuronal cells by boosting autophagy/mitophagy and upregulating a survival pathway, suggesting that A. argyi can further be exploited to slow the progression of PD.


Subject(s)
Artemisia , Neuroblastoma , Neuroprotective Agents , Parkinson Disease , Transient Receptor Potential Channels/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Autophagy , Dopaminergic Neurons , Humans , Mice , Mice, Inbred C57BL , Mitophagy , Neuroblastoma/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , alpha-Synuclein/metabolism , beta Catenin/metabolism
10.
Phytomedicine ; 104: 154281, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35752080

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is the second most common neurodegenerative disease featured to mitochondrial dysfunction in neuronal cells. Dynamin-related protein 1 (Drp1) is an important regulator of mitochondrial fission and subsequent mitophagy. Mangiferin (MGF) is a glucosyl xanthone mainly derived from Mangifera indica L., possessing multifaceted properties, e.g., antioxidant, anti-inflammatory, and enhancement of cognitive ability. Besides, it can cross the blood-brain barrier, thereby exerting a neuroprotective effect. However, so far, MGF's effect in balancing mitochondrial homeostasis via regulation of Drp1 level and mitophagic pathway in PD remains rarely reported. PURPOSE: We aimed to investigate the neuroprotective effect of MGF against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD and examine the possible mechanisms. METHODS: We utilized C57BL/6 mice exposed to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP); Behavioral parameters, containing the open field test, balance beam, pole test, and rotarod test, assessed the locomotor activity; immunohistochemistry assessed the number of TH-positive neurons; transmission electron microscopy detected ultrastructural mitochondrial morphology in the dopaminergic neuron; complex I enzymatic activity microplate assay kit measured the mitochondrial complex I activity; ATP determination kit measured ATP levels in mitochondria isolated from cells or striatal tissues; western blot measured the levels of Drp1 and mitophagic proteins. RESULTS: We observed that MGF could mitigate motor deficiency and improve the expression of tyrosine hydroxylase in the substantia nigra of MPTP-induced PD mice. Furthermore, MGF not only ameliorated mitochondrial ultrastructure, but also improved mitochondrial ATP content. Within mitochondria, MGF could reduce Drp1 expression and reverse the expressions of mitophagic proteins, including PINK1, Parkin, NIX, BNIP3, FUNDC1, and p62. CONCLUSION: Present study indicates that MGF benefits mitochondrial networks by recovering mitochondrial ultrastructure and ATP contents, reducing mitochondrial Drp1, and modulating mitophagic proteins in the MPTP-induced PD mice model, which revealed a novel acting mechanism of MGF in PD's treatment.


Subject(s)
Neurodegenerative Diseases , Neuroprotective Agents , Parkinson Disease , Xanthones , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Disease Models, Animal , Dopaminergic Neurons , Dynamins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondria , Mitochondrial Proteins/metabolism , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Xanthones/pharmacology , Xanthones/therapeutic use
11.
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
12.
Oxid Med Cell Longev ; 2022: 1837278, 2022.
Article in English | MEDLINE | ID: mdl-36589679

ABSTRACT

A naphthoquinone molecule known as plumbagin (PL), which has a wide range of pharmacological properties including antitumor, antioxidation, anti-inflammation, and neuroprotective effects, is extracted from the roots of the medicinal herb Plumbago zeylanica L. Plumbagin has been studied for its potential to treat Parkinson's disease (PD). However, its effectiveness and mechanism are still unknown. This study intends to evaluate plumbagin's effectiveness against PD in vitro and in vivo. Plumbagin partially repaired the loss of dopaminergic neurons in the nigral substantia nigra and the resulting behavioural impairment caused by MPTP or MPTP/probenecid in mice. Furthermore, plumbagin treatment significantly inhibited the TLR/NF-κB pathways. It reduced the TNF-α, IL-6, and IL-1ß mRNA expression in PD mice induced by MPTP or MPTP/probenecid, which was consistent with the findings in the inflammatory model of BV2 cells induced by MPP+ or LPS. In addition, plumbagin treatment enhanced the microtubule-associated protein 1 light chain 3 beta (LC3) LC3-II/LC3-I levels while decreasing the p-mTOR and p62 protein accumulation in PD mice induced by MPTP or MPTP/probenecid, which was similar to the results obtained from the experiments in SH-SY5Y and PC12 cells induced by MPP+. Consequently, our results support the hypothesis that plumbagin, by promoting autophagy and inhibiting the activation of the TLR/NF-κB signaling pathway, is a promising treatment agent for treating Parkinson's disease (PD). However, to confirm plumbagin's anti-PD action more thoroughly, other animal and cell PD models must be used in future studies.


Subject(s)
Naphthoquinones , Neuroblastoma , Neuroprotective Agents , Parkinson Disease , Rats , Humans , Mice , Animals , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/metabolism , Probenecid/metabolism , Probenecid/pharmacology , Neuroblastoma/metabolism , Signal Transduction , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use , Naphthoquinones/metabolism , Dopaminergic Neurons/metabolism , Autophagy , Mice, Inbred C57BL , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Disease Models, Animal
13.
Brain Res Bull ; 135: 120-128, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29032054

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease which causes rigidity, resting tremor and postural instability. The neuroprotective effects of an ethanolic extract of Bacopa monnieri (BM) were evaluated in a Parkinsonian mice model induced by the MPTP. The present study investigates the mechanisms of neuroprotection elicited by BM, an herb traditionally recognized by the Indian system of medicine, Ayurveda. An ethanolic extract of BM was co-treated with the MPTP induced mouse model of PD and was shown to significantly rescue the motor behaviour (Rotarod, Grip Strength and Foot Printing test). Furthermore, on biochemical parameters too BM significantly showed protective effect as Catalase, LPO, Nitrite, SOD, GR, GPx parameters showed marked improvement and levels of Dopamine, DOPAC and HVA were enhanced significantly. There was a significant reduction in tyrosine hydroxylase (TH) immunoreactivity in the substantia nigra (SN) in MPTP treated group, which was considerably restored by the use of BM extract. BM also facilitated neuroprotection by creating an anti-apoptotic environment indicated by reduced apoptotic (Bax and caspase-3) and increased levels of anti-apoptotic (Bcl2) protein expression, respectively. Altogether, the present study suggests that BM treatment provides nigrostriatal dopaminergic neuroprotection against MPTP induced Parkinsonism by the modulation of oxidative stress and apoptotic machinery possibly accounting for the behavioural effects.


Subject(s)
Apoptosis/drug effects , Bacopa/metabolism , Dopaminergic Neurons/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Apoptosis/physiology , Bacopa/physiology , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Male , Mice , Motor Activity/drug effects , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Plant Extracts/pharmacology , Substantia Nigra/drug effects
14.
Pharmacol Res ; 115: 25-44, 2017 01.
Article in English | MEDLINE | ID: mdl-27838509

ABSTRACT

Pathogenesis of neurodegenerative diseases such as Parkinson's disease (PD) and Huntington's disease (HD) are closely related to the formation of protein aggregates and inclusion body. For instance, active autophagic components from Chinese herbal medicines (CHMs) are highlighted to modulate neurodegeneration via degradation of disease proteins. In this study, the neuroprotective effect of the purified Hedera helix (HH) fraction containing both hederagenin and α-hederin, is confirmed by the improvement of motor deficits in PD mice model. Furthermore, hederagenin and α-hederin derived from HH are confirmed as novel autophagic enhancers. Both compounds reduce the protein level of mutant huntingtin with 74 CAG repeats and A53T α-synuclein, and inhibit the oligomerization of α-synuclein and inclusion formation of huntingtin, via AMPK-mTOR dependent autophagy induction. Both hederagenin and α-hederin induce autophagy and promote the degradation of neurodegenerative mutant disease proteins in vitro, suggesting the therapeutic roles of HH in neurodegenerative disorders.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Neurodegenerative Diseases/drug therapy , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Adenylate Kinase/metabolism , Animals , Autophagy/drug effects , Cell Line, Tumor , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Female , Hedera/chemistry , Huntingtin Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Oleanolic Acid/pharmacology , PC12 Cells , Rats , TOR Serine-Threonine Kinases/metabolism , alpha-Synuclein/metabolism
15.
Neurotoxicology ; 33(3): 321-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22342763

ABSTRACT

Hypothalamic tuberoinfundibular dopamine (TIDA) neurons remain unaffected in Parkinson disease (PD) while there is significant degeneration of midbrain nigrostriatal dopamine (NSDA) neurons. A similar pattern of susceptibility is observed in acute and chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse and rotenone rat models of degeneration. It is not known if the resistance of TIDA neurons is a constitutive or induced cell-autonomous phenotype for this unique subset of DA neurons. In the present study, treatment with a single injection of MPTP (20 mg/kg; s.c.) was employed to examine the response of TIDA versus NSDA neurons to acute injury. An acute single dose of MPTP caused an initial loss of DA from axon terminals of both TIDA and NSDA neurons, with recovery occurring solely in TIDA neurons by 16 h post-treatment. Initial loss of DA from axon terminals was dependent on a functional dopamine transporter (DAT) in NSDA neurons but DAT-independent in TIDA neurons. The active metabolite of MPTP, 1-methyl, 4-phenylpyradinium (MPP+), reached higher concentration and was eliminated slower in TIDA compared to NSDA neurons, which indicates that impaired toxicant bioactivation or distribution is an unlikely explanation for the observed resistance of TIDA neurons to MPTP exposure. Inhibition of protein synthesis prevented TIDA neuron recovery, suggesting that the ability to recover from injury was dependent on an induced, rather than a constitutive cellular mechanism. Further, there were no changes in total tyrosine hydroxylase (TH) expression following MPTP, indicating that up-regulation of the rate-limiting enzyme in DA synthesis does not account for TIDA neuronal recovery. Differential candidate gene expression analysis revealed a time-dependent increase in parkin and ubiquitin carboxyl-terminal hydrolase-L1 (UCH-L1) expression (mRNA and protein) in TIDA neurons during recovery from injury. Parkin expression was also found to increase with incremental doses of MPTP. The increase in parkin expression occurred specifically within TIDA neurons, suggesting that these neurons have an intrinsic ability to up-regulate parkin in response to MPTP-induced injury. These data suggest that TIDA neurons have a compensatory mechanism to deal with toxicant exposure and increased oxidative stress, and this unique TIDA neuron phenotype provides a platform for dissecting the mechanisms involved in the natural resistance of central DA neurons following toxic insult.


Subject(s)
Basal Ganglia/drug effects , Dopaminergic Neurons/drug effects , Hypothalamus/drug effects , MPTP Poisoning/etiology , Striatonigral Degeneration/chemically induced , Substantia Nigra/drug effects , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Animals , Basal Ganglia/enzymology , Basal Ganglia/pathology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Hypothalamus/enzymology , Hypothalamus/pathology , Injections, Subcutaneous , MPTP Poisoning/enzymology , MPTP Poisoning/genetics , MPTP Poisoning/pathology , Male , Mice , Mice, Inbred C57BL , Phenotype , RNA, Messenger/metabolism , Recovery of Function , Striatonigral Degeneration/enzymology , Striatonigral Degeneration/genetics , Striatonigral Degeneration/pathology , Substantia Nigra/enzymology , Substantia Nigra/pathology , Time Factors , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin-Protein Ligases/genetics , Up-Regulation
16.
Exp Toxicol Pathol ; 64(7-8): 761-5, 2012 Nov.
Article in English | MEDLINE | ID: mdl-21324658

ABSTRACT

Intraperitoneal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration induces apoptosis of subventricular zone (SVZ) doublecortin (Dcx)-positive neural progenitor cells (migrating neuroblasts, A cells). Actually, a metabolite of MPTP, 1-methy-4-phenylpiridinium (MPP(+)), is responsible for neural progenitor cell toxicity. In the present study, to examine whether the MPTP-induced SVZ cell apoptosis is caused directly by MPP(+) metabolized through monoamine oxidase B (MAO-B), MPTP or MPP(+) was intracerebroventricularly (icv) injected into C57BL/6 mice. At Day 1 postinjection, many terminal deoxynucleotidyl transferase-mediated dUTP endlabeling (TUNEL)-positive cells were observed in the SVZ of both low (36 µg) and high (162 µg) dose MPTP- and MPP(+)-injected mice. The number of Dcx-positive A cells showed a significant decrease following high dose of MPTP- or MPP(+)-injection on Days 1 and 3, respectively, whereas that of EGFR-positive C cells showed no change in mice with any treatment. In addition, prior icv injection of a MAO-B inhibitor, R(-)-deprenyl (deprenyl), inhibited MPTP-induced apoptosis, but not MPP(+)-induced apoptosis. MAO-B- and GFAP-double positive cells were detected in the ependyma and SVZ in all mice. It is revealed from these results that icv injection of MPTP induces apoptosis of neural progenitor cells (A cells) in the SVZ via MPP(+) toxicity. In addition, it is suggested that the conversion from MPTP to MPP(+) is caused mainly by MAO-B located in ependymal cells and GFAP-positive cells in the SVZ.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenylpyridinium/metabolism , Apoptosis/drug effects , Cerebral Ventricles/drug effects , Monoamine Oxidase/metabolism , Neural Stem Cells/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Cerebral Ventricles/enzymology , Cerebral Ventricles/pathology , Dose-Response Relationship, Drug , Doublecortin Domain Proteins , Doublecortin Protein , Ependyma/drug effects , Ependyma/enzymology , Ependyma/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Neuropeptides/metabolism , Selegiline/pharmacology
17.
Basic Clin Pharmacol Toxicol ; 107(6): 958-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20629656

ABSTRACT

Previous reports have suggested that the herbal medicine Chunghyuldan (CHD, Qingxue-dan in Chinese and Daio-Orengedokuto in Japanese) has wide-ranging biological effects, including anti-hyperlipidaemic, anti-ischaemic, anti-inflammatory and antioxidant activities. Reactive oxygen species (ROS)-mediated mitochondrial dysfunction is thought to be one of the major pathological mechanisms responsible for Parkinson's disease (PD) and may underlie the selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) that is a hallmark of this disease. In this study, we examined the neuroprotective effects of CHD in PD models produced by treatment with neurotoxins that act via ROS-mediated mitochondrial dysfunction. In an in vitro PD model using 6-hydroxydopamine, CHD applied at concentrations of 10 and 100 µg/ml exhibited significant protective effects in PC12 cells by inhibiting intracellular ROS generation. CHD applied at 10 and 100 µg/ml also prevented 6-hydroxydopamine-induced mitochondrial depolarization and elevation of caspase-3 activity. At the same doses, CHD showed regulatory effects on the haem oxygenase-1 and gp91 phagocytic oxidase which have critical roles in generating ROS. In addition, CHD protected dopaminergic neurons in a primary mesencephalic culture against MPP+ neurotoxicity. In an in vivo PD model produced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment (20 mg/kg, 4 times, i.p.), co-administration of CHD (50 mg/kg, 5 days, p.o.) ameliorated PD-like behavioural symptoms (bradykinesia) and reduced dopaminergic neuronal damage in the SNpc and striatum as measured by immunocytochemistry. These results demonstrate the neuroprotective effects of CHD in PD models that are mediated through inhibition of ROS generation and associated mitochondrial dysfunction.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Reactive Oxygen Species/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Caspase 3/metabolism , Cell Death , Cells, Cultured , Disease Models, Animal , Dopamine/metabolism , Male , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Neurotoxins/metabolism , Oxidopamine/pharmacology , PC12 Cells , Parkinson Disease/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/pathology
18.
Pediatr Res ; 62(1): 14-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17515828

ABSTRACT

The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) inhibits the mitochondrial complex I of the respiratory chain. This results in ATP and ion homeostasis disturbances, which lead to selective death of the substantia nigra dopaminergic neurons. Well known as a Parkinson's disease model, the MPTP animal model also provides a potential paradigm of the energy deficiencies found in childhood. In these conditions, anticonvulsants may provide neuroprotection by limiting cellular energy consumption. We tested valproate, topiramate and lamotrigine in the MPTP mouse model. Dopamine transporter (DAT) density was assessed by quantitative autoradiography, tyrosine hydroxylase (TH) was evaluated by immunohistochemistry and dopamine (DA) levels by HPLC-ED whereas neuronal apoptosis was monitored through active caspase-3. Expectedly, the DAT density, TH immunoreactive neurons and DA content in the MPTP group were respectively reduced to 51%, 40% and 26% versus control animals. Unlike valproate and topiramate, lamotrigine provided a significant neuroprotection against MPTP in maintaining these levels at 99%, 74% and 58% respectively and reducing the induced apoptosis. Altogether, the data indicate that lamotrigine limits dopaminergic neuronal death in the substantia nigra and promotes striatal dendrites sprouting. Lamotrigine, a widely used and well-tolerated molecule in young patients, could represent a valuable adjuvant therapy in various energy deficiency conditions during childhood.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Anticonvulsants/metabolism , Dopamine Agents/metabolism , Energy Metabolism , Neuroprotective Agents/metabolism , Neurotoxins/metabolism , Triazines/metabolism , Animals , Anticonvulsants/chemistry , Apoptosis/physiology , Corpus Striatum/cytology , Corpus Striatum/metabolism , Dopamine/metabolism , Dopamine Agents/toxicity , Fructose/analogs & derivatives , Fructose/metabolism , Lamotrigine , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/chemistry , Neurotoxins/toxicity , Topiramate , Triazines/chemistry , Valproic Acid/metabolism
19.
Synapse ; 40(1): 47-54, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11170221

ABSTRACT

This is the first study to investigate the potential protective effects of the lipophilic kavapyrone (+/-)-kavain in the experimental MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) model of Parkinson's disease (PD). Male C57BL/6 mice were treated with (+/-)-kavain (50, 100, or 200 mg/kg i.p.) or vehicle 60 min before and 60 min after a single administration of MPTP (30 mg/kg s.c.) or saline, respectively. Mice were sacrificed after 7 days and the neostriatum was analyzed for dopamine and its metabolites using HPLC with electrochemical detection. Furthermore, nigral sections were processed for tyrosine hydroxylase (TH) immunocytochemistry. To determine the effects of (+/-)-kavain (200 mg/kg) on MPTP metabolism, HPLC analysis of striatal MPP(+) (1-methyl-4-phenylpyridinium) levels was performed. MPTP treatment alone led to a significant depletion of striatal dopamine levels to 12.61% of saline controls. The lower dosages of (+/-)-kavain (50 and 100 mg/kg) showed only a nonsignificant attenuation of MPTP-induced dopamine depletion, but a high dosage of (+/-)-kavain (200 mg/kg) significantly antagonized the dopamine depletion to 58.93% of saline control values. Remarkably, the MPTP-induced decrease of TH-immunoreactivity as well as the loss of nigral neurons was completely prevented by (+/-)-kavain (200 mg/kg). Striatal MPP(+) levels were not altered by (+/-)-kavain treatment. In conclusion, we found that MPTP metabolism was not influenced by (+/-)-kavain and postulate the antiglutamatergic effects of (+/-)-kavain for its protective effects against MPTP toxicity. (+/-)-Kavain may be a novel candidate for further preclinical studies in animal models of PD and other disorders with glutamatergic overactivity.


Subject(s)
Anticonvulsants/pharmacology , Brain/drug effects , Disease Models, Animal , Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Pyrones/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Brain/metabolism , Brain/physiopathology , Dopamine/metabolism , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Neostriatum/drug effects , Neostriatum/metabolism , Neostriatum/physiopathology , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neurons/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL