Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.679
1.
Neuromolecular Med ; 26(1): 19, 2024 May 04.
Article En | MEDLINE | ID: mdl-38703217

Parkinson's disease (PD) is a neurodegenerative disorder associated with mitochondrial dysfunctions and oxidative stress. However, to date, therapeutics targeting these pathological events have not managed to translate from bench to bedside for clinical use. One of the major reasons for the lack of translational success has been the use of classical model systems that do not replicate the disease pathology and progression with the same degree of robustness. Therefore, we employed a more physiologically relevant model involving alpha-synuclein-preformed fibrils (PFF) exposure to SH-SY5Y cells and Sprague Dawley rats. We further explored the possible involvement of transient receptor potential canonical 5 (TRPC5) channels in PD-like pathology induced by these alpha-synuclein-preformed fibrils with emphasis on amelioration of oxidative stress and mitochondrial health. We observed that alpha-synuclein PFF exposure produced neurobehavioural deficits that were positively ameliorated after treatment with the TRPC5 inhibitor clemizole. Furthermore, Clemizole also reduced p-alpha-synuclein and diminished oxidative stress levels which resulted in overall improvements in mitochondrial biogenesis and functions. Finally, the results of the pharmacological modulation were further validated using siRNA-mediated knockdown of TRPC5 channels, which also decreased p-alpha-synuclein expression. Together, the results of this study could be superimposed in the future for exploring the beneficial effects of TRPC5 channel modulation for other neurodegenerative disorders and synucleopathies.


Mitochondria , Oxidative Stress , Rats, Sprague-Dawley , TRPC Cation Channels , alpha-Synuclein , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Animals , Rats , Oxidative Stress/drug effects , Humans , TRPC Cation Channels/genetics , TRPC Cation Channels/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/metabolism , Cell Line, Tumor , Male , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/chemically induced , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy
2.
Int J Mol Sci ; 25(10)2024 May 13.
Article En | MEDLINE | ID: mdl-38791326

Chronic environmental exposure to toxic heavy metals, which often occurs as a mixture through occupational and industrial sources, has been implicated in various neurological disorders, including Parkinsonism. Vanadium pentoxide (V2O5) typically presents along with manganese (Mn), especially in welding rods and high-capacity batteries, including electric vehicle batteries; however, the neurotoxic effects of vanadium (V) and Mn co-exposure are largely unknown. In this study, we investigated the neurotoxic impact of MnCl2, V2O5, and MnCl2-V2O5 co-exposure in an animal model. C57BL/6 mice were intranasally administered either de-ionized water (vehicle), MnCl2 (252 µg) alone, V2O5 (182 µg) alone, or a mixture of MnCl2 (252 µg) and V2O5 (182 µg) three times a week for up to one month. Following exposure, we performed behavioral, neurochemical, and histological studies. Our results revealed dramatic decreases in olfactory bulb (OB) weight and levels of tyrosine hydroxylase, dopamine, and 3,4-dihydroxyphenylacetic acid in the treatment groups compared to the control group, with the Mn/V co-treatment group producing the most significant changes. Interestingly, increased levels of α-synuclein expression were observed in the substantia nigra (SN) of treated animals. Additionally, treatment groups exhibited locomotor deficits and olfactory dysfunction, with the co-treatment group producing the most severe deficits. The treatment groups exhibited increased levels of the oxidative stress marker 4-hydroxynonenal in the striatum and SN, as well as the upregulation of the pro-apoptotic protein PKCδ and accumulation of glomerular astroglia in the OB. The co-exposure of animals to Mn/V resulted in higher levels of these metals compared to other treatment groups. Taken together, our results suggest that co-exposure to Mn/V can adversely affect the olfactory and nigral systems. These results highlight the possible role of environmental metal mixtures in the etiology of Parkinsonism.


Manganese Compounds , Manganese , Mice, Inbred C57BL , Vanadium , Animals , Mice , Manganese/toxicity , Vanadium/toxicity , Male , Olfactory Bulb/metabolism , Olfactory Bulb/drug effects , Olfactory Bulb/pathology , Dopamine/metabolism , Vanadium Compounds , Oxidative Stress/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/chemically induced , alpha-Synuclein/metabolism , Chlorides/toxicity , Chlorides/metabolism , Tyrosine 3-Monooxygenase/metabolism , Aldehydes/metabolism , Substantia Nigra/metabolism , Substantia Nigra/drug effects , Substantia Nigra/pathology , Disease Models, Animal , 3,4-Dihydroxyphenylacetic Acid/metabolism
3.
Physiol Behav ; 281: 114563, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38723388

Parkinson's Disease (PD) is a neurodegenerative movement disorder characterized by dopamine (DA) cell loss in the substantia nigra pars compacta (SNc). As PD progresses, patients display disruptions in gait such as changes in posture, bradykinesia, and shortened stride. DA replacement via L-DOPA alleviates many PD symptoms, though its effects on gait are not well demonstrated. This study aimed to assess the relationship between DA lesion, gait, and deficit-induced reversal with L-DOPA. To do so, Sprague-Dawley rats (N = 25, 14 males, 11 females) received unilateral medial forebrain bundle (MFB) DA lesions with 6-hydroxydopamine (6-OHDA). An automated gait analysis system assessed spatiotemporal gait parameters pre- and post-lesion, and after various doses of L-DOPA (0, 3, or 6 mg/kg; s.c.). The forepaw adjusting steps (FAS) test was implemented to evaluate lesion efficacy while the abnormal involuntary movements (AIMs) scale monitored the emergence of L-DOPA-induced dyskinesia (LID). High performance liquid chromatography (HPLC) assessed changes in brain monoamines on account of lesion and treatment. Results revealed lesion-induced impairments in gait, inclusive of max-contact area and step-sequence alterations that were not reversible with L-DOPA. However, the emergence of AIMs were observed at higher doses. Post-mortem, 6-OHDA lesions induced a loss of striatal DA and norepinephrine (NE), while prefrontal cortex (PFC) displayed noticeable reduction in NE but not DA. Our findings indicate that hemiparkinsonian rats display measurable gait disturbances similar to PD patients that are not rescued by DA replacement. Furthermore, non-DA mechanisms such as attention-related NE in PFC may contribute to altered gait and may constitute a novel target for its treatment.


Gait Disorders, Neurologic , Levodopa , Oxidopamine , Rats, Sprague-Dawley , Animals , Levodopa/pharmacology , Levodopa/adverse effects , Male , Female , Rats , Gait Disorders, Neurologic/chemically induced , Gait Disorders, Neurologic/drug therapy , Gait Disorders, Neurologic/etiology , Antiparkinson Agents/pharmacology , Disease Models, Animal , Medial Forebrain Bundle/drug effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , Parkinsonian Disorders/pathology , Dopamine/metabolism , Dose-Response Relationship, Drug , Functional Laterality/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Gait/drug effects , Dyskinesia, Drug-Induced
4.
Brain Res ; 1835: 148918, 2024 Jul 15.
Article En | MEDLINE | ID: mdl-38588847

The lateral habenula (LHb) projects to the ventral tegmental area (VTA) and dorsal raphe nuclei (DRN) that deliver dopamine (DA) and serotonin (5-HT) to cortical and limbic regions such as the medial prefrontal cortex (mPFC), hippocampus and basolateral amygdala (BLA). Dysfunctions of VTA-related mesocorticolimbic dopaminergic and DRN-related serotonergic systems contribute to non-motor symptoms in Parkinson's disease (PD). However, how the LHb affects the VTA and DRN in PD remains unclear. Here, we used electrophysiological and neurochemical approaches to explore the effects of LHb lesions on the firing activity of VTA and DRN neurons, as well as the levels of DA and 5-HT in related brain regions in unilateral 6-hydroxydopamie (6-OHDA)-induced PD rats. We found that compared to sham lesions, lesions of the LHb increased the firing rate of DA neurons in the VTA and 5-HT neurons in the DRN, but decreased the firing rate of GABAergic neurons in the same nucleus. In addition, lesions of the LHb increased the levels of DA and 5-HT in the mPFC, ventral hippocampus and BLA compared to sham lesions. These findings suggest that lesions of the LHb enhance the activity of mesocorticolimbic dopaminergic and serotonergic systems in PD.


Dopamine , Dopaminergic Neurons , Dorsal Raphe Nucleus , Habenula , Rats, Sprague-Dawley , Serotonergic Neurons , Serotonin , Ventral Tegmental Area , Animals , Ventral Tegmental Area/metabolism , Habenula/metabolism , Male , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Dorsal Raphe Nucleus/metabolism , Serotonergic Neurons/metabolism , Serotonergic Neurons/physiology , Rats , Serotonin/metabolism , Dopamine/metabolism , Oxidopamine/toxicity , Parkinsonian Disorders/physiopathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Prefrontal Cortex/metabolism , Neural Pathways/metabolism , Neural Pathways/physiopathology
5.
Exp Brain Res ; 242(5): 1175-1190, 2024 May.
Article En | MEDLINE | ID: mdl-38499659

Parkinson's disease is a degenerative, chronic and progressive disease, characterized by motor dysfunctions. Patients also exhibit non-motor symptoms, such as affective and sleep disorders. Sleep disorders can potentiate clinical and neuropathological features and lead to worse prognosis. The goal of this study was to evaluate the effects of sleep deprivation (SD) in mice submitted to a progressive pharmacological model of Parkinsonism (chronic administration with a low dose of reserpine). Male Swiss mice received 20 injections of reserpine (0.1 mg/kg) or vehicle, on alternate days. SD was applied before or during reserpine treatment and was performed by gentle handling for 6 h per day for 10 consecutive days. Animals were submitted to motor and non-motor behavioral assessments and neurochemical evaluations. Locomotion was increased by SD and decreased by reserpine treatment. SD during treatment delayed the onset of catalepsy, but SD prior to treatment potentiated reserpine-induced catalepsy. Thus, although SD induced an apparent beneficial effect on motor parameters, a delayed deleterious effect on alterations induced by reserpine was found. In the object recognition test, both SD and reserpine treatment produced cognitive deficits. In addition, the association between SD and reserpine induced anhedonic-like behavior. Finally, an increase in oxidative stress was found in hippocampus of mice subjected to SD, and tyrosine hydroxylase immunoreactivity was reduced in substantia nigra of reserpine-treated animals. Results point to a possible late effect of SD, aggravating the deficits in mice submitted to the reserpine progressive model of PD.


Disease Models, Animal , Parkinsonian Disorders , Reserpine , Sleep Deprivation , Animals , Male , Reserpine/pharmacology , Sleep Deprivation/complications , Mice , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , Catalepsy/chemically induced , Oxidative Stress/physiology , Oxidative Stress/drug effects , Tyrosine 3-Monooxygenase/metabolism , Motor Activity/physiology , Motor Activity/drug effects , Recognition, Psychology/physiology , Recognition, Psychology/drug effects , Anhedonia/physiology , Anhedonia/drug effects
6.
Brain Res ; 1834: 148893, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38554797

Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. The dopamine D3 receptor (D3R) plays a significant role in the pathogenesis and treatment of PD. Activation of receptor tyrosine kinases (RTKs) inhibits signaling mediated by G protein-coupled receptor (GPCR). Epidermal growth factor receptors (EGFRs) and dopamine D3 receptors in the brain are directly associated with PD, both in terms of its development and potential treatment. Therefore, we investigated the impact of modulating the EGFR, a member of the RTKs family, and the dopamine D3R, a member of the GPCR family. In the present study, 100 mg/kg of lapatinib (LAP) was administered to rotenone-intoxicated rats for three weeks. Our findings indicate that LAP effectively alleviated motor impairment, improved histopathological abnormalities, and restored dopaminergic neurons in the substantia nigra. This restoration was achieved through the upregulation of dopamine D3R and increase of tyrosine hydroxylase (TH) expression, as well as boosting dopamine levels. Furthermore, LAP inhibited the activity of p-EGFR, GRK2, and SCR. Additionally, LAP exhibited antioxidant properties by inhibiting the 4-hydroxynonenal (4-HNE) and PLCγ/PKCßII pathway, while enhancing the antioxidant defense mechanism by increasing GSH-GPX4 pathway. The current study offers insights into the potential repositioning of LAP as a disease-modifying drug for PD. This could be achieved by modulating the dopaminergic system and curbing oxidative stress.


Dopaminergic Neurons , ErbB Receptors , Lapatinib , Parkinsonian Disorders , Receptors, Dopamine D3 , Rotenone , Animals , Male , Rats , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Lapatinib/pharmacology , Oxidative Stress/drug effects , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/chemically induced , Receptors, Dopamine D3/metabolism , Receptors, Dopamine D3/antagonists & inhibitors , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
7.
Biomed Khim ; 70(1): 25-32, 2024 Feb.
Article En | MEDLINE | ID: mdl-38450678

Isatin (indoldione-2,3) is an endogenous biological regulator found in the brain, peripheral tissues, and biological fluids of humans and animals. Its biological activity is realized via isatin-binding proteins, many of which were identified during proteomic profiling of the brain of mice and rats. A number of these proteins are related to the development of neurodegenerative diseases. Previously, using a model of experimental Parkinsonism induced by a seven-day course of rotenone injections, we have observed behavioral disturbances, as well as changes in the profile and relative content of brain isatin-binding proteins. In this study, we have investigated behavioral responses and the relative content of brain isatin-binding proteins in rats with rotenone-induced Parkinsonism 5 days after the last administration of this neurotoxin. Despite the elimination of rotenone, animals exhibited motor and coordination impairments. Proteomic profiling of isatin-binding proteins revealed changes in the relative content of 120 proteins (the relative content of 83 proteins increased and that of 37 proteins decreased). Comparison of isatin-binding proteins characterized by the changes in the relative content observed in the brain right after the last injection of rotenone (n=16) and 5 days later (n=11) revealed only two common proteins (glyceraldehyde-3-phosphate dehydrogenase and subunit B of V-type proton ATPase). However, most of these proteins are associated with neurodegeneration, including Parkinson's and Alzheimer's diseases.


Isatin , Parkinsonian Disorders , Humans , Animals , Rats , Carrier Proteins , Isatin/pharmacology , Rotenone/pharmacology , Proteomics , Brain , Parkinsonian Disorders/chemically induced
8.
Neurotoxicology ; 101: 117-127, 2024 Mar.
Article En | MEDLINE | ID: mdl-38423185

The study investigated the protective effects of Hesperetin (HSP) and Hesperidin (HSD) on 1 methyl, 4 phenyl, 1,2,3,6 tetrahydropyridine hydrochloride (MPTP)-induced Parkinsonism in Drosophila melanogaster (D. melanogaster). After a lifespan study to select exposure time and concentrations, flies were co-exposed to MPTP (0.4 mg/g diet), Hesperetin (0.2 and 0.4 mg/g diet), and Hesperidin (0.1 and 0.4 mg/g) for 7 days. In addition to in vivo parameters, we assayed some markers of oxidative stress and antioxidant status (lipid peroxidation, protein carbonylation, thiol content, hydrogen peroxide, and nitrate/nitrite levels, mRNA expression of Keap-1 (Kelch-like ECH associated protein 1), /Nrf2 (Nuclear factor erythroid 2 related factor 2), catalase, and glutathione-S-transferase (GST) activities), and cholinergic (acetyl cholinesterase activity (AChE) and dopaminergic signaling content and the mRNA expression of tyrosine hydroxylase (TH), monoamine oxidase (MAO-like) activity). In addition to increasing the lifespan of flies, we found that both flavonoids counteracted the adverse effects of MPTP on survival, offspring emergence, and climbing ability of flies. Both flavonoids also reduced the oxidative damage on lipids and proteins and reestablished the basal levels of pro-oxidant species and activities of antioxidant enzymes in MPTP-exposed flies. These responses were accompanied by the normalization of the mRNA expression of Keap1/Nrf2 disrupted in flies exposed to MPTP. MPTP exposure also elicited changes in mRNA expression and content of TH as well as in MAO and AChE activity, which were reversed by HST and HSD. By efficiently hindering the oxidative stress in MPTP-exposed flies, our findings support the promising role of Hesperetin and Hesperidin as adjuvant therapy to manage Parkinsonism induced by chemicals such as MPTP.


Hesperidin , Parkinson Disease , Parkinsonian Disorders , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Drosophila melanogaster , Hesperidin/pharmacology , Hesperidin/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Flavonoids/pharmacology , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/prevention & control , Phenotype , Monoamine Oxidase/metabolism , RNA, Messenger/metabolism
9.
Eur J Neurosci ; 59(6): 1169-1176, 2024 Mar.
Article En | MEDLINE | ID: mdl-37515363

Nelotanserin is a serotonin 2A and 2C (5-HT2A/2C) inverse agonist that was previously tested in the clinic for rapid-eye movement sleep behaviour disorder and psychosis in patients with Parkinson's disease (PD) dementia. Its effect on L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia has however not been investigated. As 5-HT2A antagonism/inverse agonism is a validated approach to alleviate dyskinesia, we undertook the current study to evaluate the anti-dyskinetic potential of nelotanserin in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned marmoset. Parkinsonism was induced in six common marmosets (Callithrix jacchus, three females and three males) that were then chronically treated with L-DOPA to induce dyskinesia. On experimental days, they were administered L-DOPA in combination with vehicle or nelotanserin (0.1, 0.3 and 1 mg/kg) subcutaneously, in a randomised fashion. Dyskinesia and parkinsonism were rated post hoc by a blinded observer. In comparison to vehicle, the addition of nelotanserin 0.3 and 1 mg/kg to L-DOPA diminished peak dose dyskinesia by 47% (P < 0.05) and 69% (P < 0.001). Nelotanserin 0.3 and 1 mg/kg also reduced the severity of global dyskinesia, by 40% (P < 0.01) and 55% (P < 0.001), when compared to vehicle. Nelotanserin 0.1 mg/kg did not alleviate peak dose or global dyskinesia severity. Nelotanserin had no impact on the anti-parkinsonian action of L-DOPA. Our results highlight that nelotanserin may represent an efficacious anti-dyskinetic drug and provide incremental evidence of the potential benefit of 5-HT2A/2C antagonism/inverse agonism for drug-induced dyskinesia in PD.


Dyskinesia, Drug-Induced , Parkinsonian Disorders , Phenylurea Compounds , Pyrazoles , Animals , Female , Male , Antiparkinson Agents/adverse effects , Callithrix , Drug Inverse Agonism , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/etiology , Levodopa/adverse effects , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/chemically induced , Serotonin
10.
Mol Neurobiol ; 61(2): 953-970, 2024 Feb.
Article En | MEDLINE | ID: mdl-37674036

Cypermethrin impairs mitochondrial function, induces redox imbalance, and leads to Parkinsonism in experimental animals. Knockdown of deglycase-1 (DJ-1) gene, which encodes a redox-sensitive antioxidant protein, aggravates cypermethrin-mediated α-synuclein overexpression and oxidative alteration of proteins. DJ-1 is also reported to be essential for maintaining stability of nuclear factor erythroid 2-related factor 2 (Nrf2), shielding cells against oxidative insult. Leucine-rich repeat kinase 2 (LRRK2), another protein associated with Parkinson's disease, is also involved in regulating mitochondrial function. However, underlying molecular mechanisms remain elusive. The study intended to explore an interaction of DJ-1, LRRK2, and Nrf2 in the regulation of mitochondrial function in cypermethrin-induced Parkinsonism. Small interfering RNA-mediated knockdown of DJ-1 and LRRK2 gene and pharmacological activation of Nrf2 were performed in rats and/or human neuroblastoma cells with or without cypermethrin. Indexes of oxidative stress, mitochondrial impairment, and Parkinsonism along with α-synuclein expression, post-translational modification, and aggregation were measured. DJ-1 gene knockdown exacerbated cypermethrin-induced increase in oxidative stress and intrinsic apoptosis and reduction in expression of mitochondrial antioxidant proteins via inhibiting nuclear translocation of Nrf2. Additionally, cypermethrin-induced oxidative stress, mitochondrial impairment, and α-synuclein expression and aggregation were found to be suppressed by LRRK2 gene knockdown, by promoting Nrf2 nuclear translocation and expression of mitochondrial antioxidant proteins. Furthermore, Nrf2 activator, sulforaphane, ameliorated cypermethrin-induced mitochondrial impairment and oxidative stress and provided protection against dopaminergic neuronal death. The findings indicate that DJ-1 and LRRK2 independently alter Nrf2-mediated changes and a complex interplay among DJ-1, LRRK2, and Nrf2 exists in the regulation of mitochondrial function in cypermethrin-induced Parkinsonism.


Antioxidants , Parkinsonian Disorders , Pyrethrins , Animals , Humans , Rats , alpha-Synuclein/metabolism , Antioxidants/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism
11.
J Chem Neuroanat ; 135: 102366, 2024 Jan.
Article En | MEDLINE | ID: mdl-38040269

BACKGROUND: Earlier reports suggest that vitamin D3 (Vit D3) supplementation attenuates Parkinsonism in drug-induced motor deficits. Moreover, the function of Vit D3 may be optimized by co-administration with vitamin A (Vit A). In line with the synergistic interplay between vitamins, we hypothesized that the efficacy of Vit D3 to attenuate Parkinsonism in a haloperidol-induced mouse model of motor deficits would be more potent when concomitantly administered with Vit A. METHODS: Thirty-six (36) adult male mice were randomly divided into six groups of six animals each: the control group, the PD model (haloperidol-treated only group) (-D2), and four other groups treated with haloperidol together with either one or two of the following vitamin supplementations: Vit D3, Vit A, Vit D3 +VA, or bromocriptine a known PD drug respectively. Motor functions were assessed using a battery of neurobehavioral tests in experimental animals, after which brain tissues were harvested and processed for biochemical and histomorphological analysis. RESULTS: We recorded a significant decline in motor activity in the PD mice model treated with haloperidol alone compared to other experimental groups that received vitamin supplementations. The significant decrease in motor activity observed in the PD mice model corresponded with marked neurodegenerative features in the cytoarchitecture of the pyramidal cells in the striatum and primary motor cortex (M1). Furthermore, the haloperidol-induced PD mice model treated with Vit D3 +Vit A showed significant improvement in motor activity and attenuation of oxidative stress levels and neurodegenerative features compared to other groups treated with Vit A, Vit D3 and bromocriptine alone. CONCLUSION: Altogether, our findings suggest that concomitant administration of both Vit D3 and Vit A prevents the development of Parkinsonism features in the haloperidol mouse model of motor deficit. Thus, supplementation with Vit D3 +Vit A may be a viable option for slowing the onset and progression of motor deficits.


Cholecalciferol , Parkinsonian Disorders , Male , Mice , Animals , Cholecalciferol/pharmacology , Cholecalciferol/therapeutic use , Haloperidol/pharmacology , Bromocriptine , Vitamins/pharmacology , Vitamins/therapeutic use , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Dietary Supplements
12.
Asian J Psychiatr ; 91: 103857, 2024 Jan.
Article En | MEDLINE | ID: mdl-38128353

INTRODUCTION: Given the similar efficacies across antipsychotic medications for schizophrenia, understanding their safety profiles, particularly concerning receptor-binding differences, is crucial for optimal drug selection, especially for patients with first episode schizophrenia. We aimed to compare the safety outcomes of second-generation antipsychotics. METHODS: We conducted a retrospective cohort study with new user active comparator design using a nationwide claims database in South Korea. Participants were drug-naïve adult patients with first-episode schizophrenia. Three representative drugs with different pharmacologic profiles were compared: risperidone, olanzapine, and aripiprazole. Propensity scores were used to match the study groups, and the Cox proportional hazard model was used to calculate hazard ratios. Sensitivity analyses were performed in various epidemiological settings. Seventeen safety outcomes, including neuropsychiatric, cardiometabolic and gastrointestinal events, were assessed, with upper-respiratory-tract infection as a negative control outcome. RESULTS: A total of 1044, 2078, and 3634 participants were matched for olanzapine vs. risperidone, olanzapine vs. aripiprazole, and risperidone vs. aripiprazole comparisons, respectively. For parkinsonism, there was a significant difference in outcomes between the risperidone and aripiprazole groups (HR 1.80 [95% CI 1.13-2.91]), with consistent sensitivity analysis results. There were no significant differences in other neuropsychiatry outcomes or in the risk of cardiometabolic and gastrointestinal outcomes between any of the comparative group pairs. CONCLUSIONS: The risk of drug-induced parkinsonism was significantly higher with risperidone than with aripiprazole. Although olanzapine is known for its metabolic risk, there were no significant differences in risk between the other pairs.


Antipsychotic Agents , Cardiovascular Diseases , Parkinsonian Disorders , Quinolones , Schizophrenia , Adult , Humans , Antipsychotic Agents/adverse effects , Schizophrenia/drug therapy , Olanzapine/adverse effects , Aripiprazole/adverse effects , Risperidone/adverse effects , Cohort Studies , Retrospective Studies , Benzodiazepines/adverse effects , Piperazines , Republic of Korea/epidemiology , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Cardiovascular Diseases/chemically induced
13.
J Chem Neuroanat ; 136: 102385, 2024 03.
Article En | MEDLINE | ID: mdl-38160784

Parkinson's Disease (PD) is an age-dependent, incessant, dynamic neurodegenerative illness. In animal models, the administration of the dopaminergic D2 antagonist Haloperidol (HP) affects the nigrostriatal pathway, inducing catalepsy, a state of immobility like PD, bradykinesia, and akinesia. The present study investigated the neural effects of Icariin (ICA), a flavonoid derived from Herba Epimedii, against HP-induced PD in rats compared to a standard drug levodopa (L-DOPA). Twenty-four adult male rats were divided into 4 groups: the control group treated with vehicle, the 2nd group treated with HP intraperitoneally, the 3rd group treated with the same dose of HP+L-DOPA orally, and the 4th one, treated with the same dose of HP+ICA orally. All the groups were treated for fourteen consecutive days. Two days before the last dose, locomotor activity was assessed in open field and rotarod tasks. At the end of the experiment, the malondialdehyde, nitric oxide (NO), iron, glycogen synthase kinase-3beta (GSK-3ß), and tyrosine hydroxylase (TH) contents, glutathione S-transferase, catalase, superoxide dismutase, activities were estimated in the midbrain. Also, cortex and midbrain monoamine contents (norepinephrine, dopamine, and serotonin) were determined. Moreover, the midbrain histopathology was detected in all treated groups. The results suggested that the neuroleptic effect of HP was completely improved by ICA. This improvement occurred by decreasing the neurotoxicity via lowering midbrain lipid peroxidation, NO, GSK-3ß contents, increasing antioxidant biomarkers, TH, and recovering the treated groups' cortex and midbrain monoamines contents. In conclusion, this study suggests that ICA is a suitable treatment for Parkinson's induced by HP.


Flavonoids , Parkinson Disease , Parkinsonian Disorders , Rats , Male , Animals , Dopamine/metabolism , Glycogen Synthase Kinase 3 beta , Levodopa/therapeutic use , Haloperidol/adverse effects , Tyrosine 3-Monooxygenase/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Disease Models, Animal
14.
Behav Brain Res ; 460: 114815, 2024 Mar 05.
Article En | MEDLINE | ID: mdl-38122905

Transcranial direct current stimulation (tDCS) is increasingly being used for Parkinson's disease (PD); however, the evaluation of its clinical impact remains complex owing to the heterogeneity of patients and treatments. Therefore, we used a unilateral 6-hydroxydopamine-induced PD rat model to investigate whether anodal tDCS of the primary motor cortex (M1) alleviates PD motor deficits. Before tDCS treatment, unilateral PD rats preferentially used the forelimb ipsilateral to the lesion in the exploratory cylinder test and showed reduced locomotor activity in the open field test. In addition, PD-related clumsy forelimb movements during treadmill walking were detected using deep learning-based video analysis (DeepLabCut). When the 5-day tDCS treatment began, the forelimb-use asymmetry was ameliorated gradually, and locomotor activity increased to pre-lesion levels. tDCS treatment also normalized unnatural forelimb movement during walking and restored a balanced gait. However, these therapeutic effects were rapidly lost or gradually disappeared when the tDCS treatment was terminated. Histological analysis at the end of the experiment revealed that the animals had moderately advanced PD, with 40-50% of dopamine neurons and fibers preserved on the injured side compared with those on the intact side. Although it remains a challenge to elucidate the neural mechanisms of the transient improvement in motor function induced by tDCS, the results of this study provide evidence that tDCS of the M1 produces positive behavioral outcomes in PD animals and provides the basis for further clinical research examining the application of tDCS in patients with PD.


Parkinson Disease , Parkinsonian Disorders , Transcranial Direct Current Stimulation , Humans , Rats , Animals , Transcranial Direct Current Stimulation/methods , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/therapy , Locomotion
15.
Biomed Khim ; 69(5): 290-299, 2023 Nov.
Article En | MEDLINE | ID: mdl-37937431

Effects of the endogenous neuroprotector isatin and the pharmacological drug afobazole (exhibiting neuroprotective properties) on behavioral reactions and quantitative changes in the brain proteomic profile have been investigated in rats with experimental rotenone Parkinsonism. A single dose of isatin (100 mg/kg subcutaneously on the last day of a 7-day course of rotenone administration) improved the motor activity of rats with rotenone-induced Parkinsonism in the open field test (horizontal movements) and the rotating rod test. Afobazole (10 mg/kg intraperitoneally, daily during the 7-day course of rotenone administration) reduced the manifestations of rigidity and postural instability. Proteomic analysis, performed using brain samples obtained the day after the last administration of rotenone and neuroprotectors, revealed similar quantitative changes in the brain of rats with rotenone Parkinsonism. An increase in the relative content of 65 proteins and a decrease in the relative content of 21 proteins were detected. The most pronounced changes - an almost ninety-fold increase in the alpha-synuclein content - were found in the brains of rats treated with isatin. In animals of the experimental groups treated with "Rotenone + Isatin", as well as "Rotenone + Afobazole", the increase in the relative content of this protein in the brain was almost 60 and 50 times higher than the control values. Taking into consideration the known data on the physiological role of alpha-synuclein, an increase in the content of this protein in the brain upon administration of neuroprotectors to animals with rotenone Parkinsonism may represent a compensatory reaction, at least in the early stages of this disease and the beginning of its treatment.


Isatin , Neuroprotective Agents , Parkinsonian Disorders , Rats , Animals , Rotenone/adverse effects , Rotenone/metabolism , Neuroprotective Agents/therapeutic use , Isatin/pharmacology , Isatin/metabolism , Octoxynol/adverse effects , Octoxynol/metabolism , alpha-Synuclein , Proteomics , Brain , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism
16.
Neurotoxicology ; 99: 226-243, 2023 Dec.
Article En | MEDLINE | ID: mdl-37926220

Exposure to pesticides is associated with an increased risk of developing Parkinson's disease (PD). Currently, rodent-based risk assessment studies cannot adequately capture neurodegenerative effects of pesticides due to a lack of human-relevant endpoints targeted at neurodegeneration. Thus, there is a need for improvement of the risk assessment guidelines. Specifically, a mechanistic assessment strategy, based on human physiology and (patho)biology is needed, which can be applied in next generation risk assessment. The Adverse Outcome Pathway (AOP) framework is particularly well-suited to provide the mechanistic basis for such a strategy. Here, we conducted a semi-systematic review in Embase and MEDLINE, focused on neurodegeneration and pesticides, to develop an AOP network for parkinsonian motor symptoms. Articles were labelled and included/excluded using the online platform Sysrev. Only primary articles, written in English, focused on effects of pesticides or PD model compounds in models for the brain were included. A total of 66 articles, out of the 1700 screened, was included. PD symptoms are caused by loss of function and ultimately death of dopaminergic neurons in the substantia nigra (SN). Our literature review highlights that a unique feature of these cells that increases their vulnerability is their reliance on continuous low-level influx of calcium. As such, excess intracellular calcium was identified as a central early Key Event (KE). This KE can lead to death of dopaminergic neurons of the SN, and eventually parkinsonian motor symptoms, via four distinct pathways: 1) activation of calpains, 2) endoplasmic reticulum stress, 3) impairment of protein degradation, and 4) oxidative damage. Several receptors have been identified that may serve as molecular initiating events (MIEs) to trigger one or more of these pathways. The proposed AOP network provides the biological basis that can be used to develop a mechanistic testing strategy that captures neurodegenerative effects of pesticides.


Adverse Outcome Pathways , Parkinson Disease , Parkinsonian Disorders , Pesticides , Humans , Parkinson Disease/metabolism , Calcium/metabolism , Parkinsonian Disorders/chemically induced , Dopaminergic Neurons , Pesticides/adverse effects , Substantia Nigra
17.
Plant Foods Hum Nutr ; 78(4): 654-661, 2023 Dec.
Article En | MEDLINE | ID: mdl-37796415

Parkinson's disease (PD) and other age-related neurodegenerative ailments have a strong link to oxidative stress. Bioflavonoid naringenin has antioxidant properties. The effects of pre- and post-naringenin supplementation on a rotenone-induced PD model were examined in this work. Naringenin (50 mg/kg, p.o.) was administered to rats for two weeks before the administration of rotenone in the pre-treatment phase. In contrast, rotenone (1.5 mg/kg, s.c.) was administered for eight days before naringenin (50 mg/kg, p.o.) was supplemented for two weeks in the post-treatment phase. During behavioral investigation, the motor and non-motor signs of PD were observed. Additionally, estimation of neurochemical and biochemical parameters was also carried out. Compared to controls, rotenone treatment substantially increased oxidative stress, altered neurotransmitters, and caused motor and non-motor impairments. Rotenone-induced motor and non-motor impairments were considerably reduced by naringenin supplementation. The supplementation also increased antioxidant enzyme activities and restored the changes in neurotransmitter levels. The findings of this work strongly imply that daily consumption of flavonoids such as naringenin may have a therapeutic potential to combat PD.


Neuroprotective Agents , Parkinsonian Disorders , Rats , Animals , Rotenone/toxicity , Antioxidants/pharmacology , Functional Food , Disease Models, Animal , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Oxidative Stress , Neuroprotective Agents/adverse effects
18.
Food Chem Toxicol ; 181: 114069, 2023 Nov.
Article En | MEDLINE | ID: mdl-37820786

Parkinson's disease (PD) is characterized by motor impairments and progressive dopaminergic neuronal death in the substantia nigra (SN). Recently, the involvement of other regulated cell death (RCD) machineries has been highlighted in PD. Necroptosis is controlled by p-RIPK1, p-RIPK3, and p-MLKL and negatively regulated by caspase-8. Ferroptosis is characterized by iron overload and accumulation of reactive oxygen species. Interestingly, the molecular chaperone complex HSP90/CDC37 has been reported to directly regulate necroptosis, ferroptosis, and some PD-associated proteins. We investigated the potential anti-necroptotic and anti-ferroptotic effects of the anti-cancer drug pazopanib, uncovering the HSP90/CDC37 complex as a master RCD modulator in rotenone-induced Parkinsonism in rats. Oral administration of 15 mg/kg pazopanib to rotenone-intoxicated rats for three weeks improved motor deficits, debilitated histopathological changes, and increased striatal dopaminergic levels. Pazopanib suppressed LRRK2 and c-Abl. Pazopanib displayed an anti-necroptotic effect through inhibition of the p-RIPK1/p-RIPK3/p-MLKL pathway and activation of caspase-8. Moreover, pazopanib inhibited the ferroptotic p-VEGFR2-PKCßII-PLC-γ-ACSL-4 pathway, iron, 4-HNE, and PTGS2 while increasing GPX-4 and GSH levels. Taken together, the current research sheds light on the repositioning of pazopanib targeting HSP90/CDC37 and its multiple RCD mechanisms, which would offer a new perspective for therapeutic strategies in PD.


Ferroptosis , Parkinson Disease , Parkinsonian Disorders , Rats , Animals , Rotenone/toxicity , Caspase 8/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinson Disease/metabolism , Dopamine/metabolism , Molecular Chaperones/metabolism , HSP90 Heat-Shock Proteins/metabolism
19.
Cell Rep Med ; 4(10): 101208, 2023 10 17.
Article En | MEDLINE | ID: mdl-37774703

Dyskinesia is involuntary movement caused by long-term medication with dopamine-related agents: the dopamine agonist 3,4-dihydroxy-L-phenylalanine (L-DOPA) to treat Parkinson's disease (L-DOPA-induced dyskinesia [LID]) or dopamine antagonists to treat schizophrenia (tardive dyskinesia [TD]). However, it remains unknown why distinct types of medications for distinct neuropsychiatric disorders induce similar involuntary movements. Here, we search for a shared structural footprint using magnetic resonance imaging-based macroscopic screening and super-resolution microscopy-based microscopic identification. We identify the enlarged axon terminals of striatal medium spiny neurons in LID and TD model mice. Striatal overexpression of the vesicular gamma-aminobutyric acid transporter (VGAT) is necessary and sufficient for modeling these structural changes; VGAT levels gate the functional and behavioral alterations in dyskinesia models. Our findings indicate that lowered type 2 dopamine receptor signaling with repetitive dopamine fluctuations is a common cause of VGAT overexpression and late-onset dyskinesia formation and that reducing dopamine fluctuation rescues dyskinesia pathology via VGAT downregulation.


Dyskinesia, Drug-Induced , Parkinsonian Disorders , Mice , Animals , Dopamine Agonists/adverse effects , Levodopa/adverse effects , Dopamine , Antiparkinson Agents/adverse effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Oxidopamine/adverse effects , gamma-Aminobutyric Acid/adverse effects
20.
Pharmacol Biochem Behav ; 231: 173637, 2023 10.
Article En | MEDLINE | ID: mdl-37714223

Group II metabotropic glutamate receptors (mGlu2/3 receptors) have been regarded as promising candidates for the treatment of L-DOPA-induced dyskinesia (LID); however, confirmation is still lacking. As the hub of the basal ganglia circuit, the striatum plays a critical role in action control. Supersensitive responsiveness of glutamatergic corticostriatal input may be the key mechanism for the development of LID. In this study, we first examined the potency of LY354740 (12 mg/kg, i.p.) in modulating glutamate and dopamine release in lesioned striatum of stable LID rats. Then, we injected LY354740 (20nmoL or 40nmoL in 4 µL of sterile 0.9 % saline) directly into the lesioned striatum to verify its ability to reduce or attenuate L-DOPA-induced abnormal involuntary movements. In experiment conducted in established LID rats, after continuous injection for 4 days, we found that LY354740 significantly reduced the expression of dyskinesia. In another experiment conducted in parkinsonism rat models, we found that LY354740 attenuated the development of LID with an inverted-U dose-response curve. The role of LY354740 in modulating striatal expressions of LID-related molecular changes was also assessed after these behavioral experiments. We found that LY354740 significantly inhibited abnormal expressions of p-Fyn/p-NMDA/p-ERK1/2/p-HistoneH3/ΔFosB, which is in line with its ability to alleviate abnormal involuntary movements in both LID expression and induction phase. Our study indicates that activation of striatal mGlu2/3 receptors can attenuate the development of dyskinesia in parkinsonism rats and provide some functional improvements in LID rats by inhibiting LID-related molecular changes.


Dyskinesia, Drug-Induced , Parkinsonian Disorders , Rats , Animals , Levodopa/adverse effects , Rats, Sprague-Dawley , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/metabolism , Corpus Striatum/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Oxidopamine , Antiparkinson Agents/adverse effects , Disease Models, Animal
...