Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 827
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Neurochem Res ; 49(7): 1677-1686, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38451434

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease and the most common movement disorder. Although PD etiology is not fully understood, alpha (α)-synuclein is a key protein involved in PD pathology. MicroRNAs (miRNA), small gene regulatory RNAs that control gene expression, have been identified as biomarkers and potential therapeutic targets for brain diseases, including PD. In particular, miR-124 is downregulated in the plasma and brain samples of PD patients. Recently we showed that the brain delivery of miR-124 counteracts 6-hydroxydopamine-induced motor deficits. However, its role in α-synuclein pathology has never been addressed. Here we used paraquat (PQ)-induced rat PD model to evaluate the role of miR-124-3p in α-synuclein accumulation and dopaminergic neuroprotection. Our results showed that an intranigral administration of miR-124-3p reduced the expression and aggregation of α-synuclein in the substantia nigra (SN) of rats exposed to PQ. NADPH oxidases (NOX), responsible for reactive oxygen species generation, have been considered major players in the development of α-synuclein pathology. Accordingly, miR-124-3p decreased protein expression levels of NOX1 and its activator, small GTPase Rac1, in the SN of PQ-lesioned rats. Moreover, miR-124-3p was able to counteract the reduced levels of pituitary homeobox 3 (PITX3), a protein required for the dopaminergic phenotype, induced by PQ in the SN. This is the first study showing that miR-124-3p decreases PQ-induced α-synuclein levels and the associated NOX1/Rac1 signaling pathway, and impacts PITX3 protein levels, supporting the potential of miR-124-3p as a disease-modifying agent for PD and related α-synucleinopathies.


Subject(s)
MicroRNAs , Paraquat , alpha-Synuclein , Animals , MicroRNAs/metabolism , alpha-Synuclein/metabolism , Paraquat/toxicity , Male , Rats , Rats, Wistar , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Substantia Nigra/drug effects , Disease Models, Animal , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Rats, Sprague-Dawley
2.
Exp Brain Res ; 240(6): 1713-1725, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35384454

ABSTRACT

In Parkinson's disease (PD) state, with progressive loss of dopaminergic neurons in the substantia nigra, the striatal dopamine (DA) and glutamate (Glu) levels change, resulting in dysfunction of basal ganglia motor regulation. The PD patient presents motor dysfunction such as resting tremor, bradykinesia, and muscular rigidity. To investigate the mechanism of aerobic exercise to improve PD-related motor dysfunction, in the current study, 6-hydroxydopamine (6-OHDA) was used to induce the PD mice model, and the motor function of PD mice was comprehensively evaluated by open-field test, rotarod test, and gait test. The co-expression of prodynorphin (PDYN) and proenkephalin (PENK) with extracellular signal-regulated kinase (Erk1/2) and phosphorylation Erk1/2 (p-Erk1/2) were detected by double-labeling immunofluorescence. The results showed that a 4-week aerobic exercise intervention could effectively improve the motor dysfunction of PD mice. Moreover, it was found that the expressions of Erk1/2 and p-Erk1/2 in the dorsal striatum (Str) of PD mice were significantly increased, and the number of positive cells co-expressed by Erk1/2, p-Erk1/2, and PENK was significantly higher than PDYN. The above phenomenon was reversed by a 4-week aerobic exercise intervention. Therefore, this study suggests that the mechanism by which aerobic exercise improves PD-related motor dysfunction may be related to that the aerobic exercise intervention alleviates the activity of extracellular signal-regulated kinase/mitogen-activated protein kinases (Erk/MAPK) signaling pathway in striatal medium spiny neurons expressing D2-like receptors (D2-MSNs) of PD mice by regulating the striatal DA and Glu signaling.


Subject(s)
Parkinson Disease, Secondary , Physical Conditioning, Animal , Signal Transduction , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/pharmacology , Dopaminergic Neurons/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Oxidopamine , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/therapy
3.
Metab Brain Dis ; 37(5): 1435-1450, 2022 06.
Article in English | MEDLINE | ID: mdl-35488941

ABSTRACT

Wuzi Yanzong Pill (WYP) was found to play a protective role on nerve cells and neurological diseases, however the molecular mechanism is unclear. To understand the molecular mechanisms that underly the neuroprotective effect of WYP on dopaminergic neurons in Parkinson's disease (PD). PD mouse model was induced by the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Gait and hanging tests were used to assess motor behavioral function. Immunofluorescence assay was used to determine TH-positive neurons in substantia nigra (SN). Apoptosis, dopamine and neurotrophic factors as well as expression of PI3K/Akt pathway were detected by TUNEL staining, ELISA and western blotting, respectively. First, it was observed that WYP intervention improved abnormal motor function in MPTP-induced PD model, alleviated the loss of TH+ neurons in SN, and increased dopamine content in brain, revealing a potential protective effect. Second, network pharmacology was used to analyze the possible targets and pathways of WYP action in the treatment of PD. A total of 126 active components related to PD were screened in WYP, and the related core targets included ALB, GAPDH, Akt1, TP53, IL6 and TNF. Particularly, the effect of WYP on PD may be medicate through PI3K/Akt signaling pathway and apoptotic regulation. The WYP treated PD mice had higher expression of p-PI3K, p-Akt and Bcl-2 but lower expression of Bax and cleaved caspase-3 than the non-WYP treated PD mice. Secretion of brain-derived neurotrophic factor (BDNF) and cerebral dopamine neurotrophic factor (CDNF) were also increased in the treated mice. WYP may inhibit apoptosis and increase the secretion of neurotrophic factor via activating PI3K/ Akt signaling pathway, thus protecting the loss of dopamine neurons in MPTP-induced PD mice.


Subject(s)
Neuroprotective Agents , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons , Drugs, Chinese Herbal/therapeutic use , Mice , Mice, Inbred C57BL , Nerve Growth Factors/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Substantia Nigra
4.
J Neurosci ; 40(50): 9772-9783, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33188066

ABSTRACT

Neuron subtype dysfunction is a key contributor to neurologic disease circuits, but identifying associated gene regulatory pathways is complicated by the molecular complexity of the brain. For example, parvalbumin-expressing (PV+) neurons in the external globus pallidus (GPe) are critically involved in the motor deficits of dopamine-depleted mouse models of Parkinson's disease, where cell type-specific optogenetic stimulation of PV+ neurons over other neuron populations rescues locomotion. Despite the distinct roles these cell types play in the neural circuit, the molecular correlates remain unknown because of the difficulty of isolating rare neuron subtypes. To address this issue, we developed a new viral affinity purification strategy, Cre-Specific Nuclear Anchored Independent Labeling, to isolate Cre recombinase-expressing (Cre+) nuclei from the adult mouse brain. Applying this technology, we performed targeted assessments of the cell type-specific transcriptomic and epigenetic effects of dopamine depletion on PV+ and PV- cells within three brain regions of male and female mice: GPe, striatum, and cortex. We found GPe PV+ neuron-specific gene expression changes that suggested increased hypoxia-inducible factor 2α signaling. Consistent with transcriptomic data, regions of open chromatin affected by dopamine depletion within GPe PV+ neurons were enriched for hypoxia-inducible factor family binding motifs. The gene expression and epigenomic experiments performed on PV+ neurons isolated by Cre-Specific Nuclear Anchored Independent Labeling identified a transcriptional regulatory network mediated by the neuroprotective factor Hif2a as underlying neural circuit differences in response to dopamine depletion.SIGNIFICANCE STATEMENT Cre-Specific Nuclear Anchored Independent Labeling is an enhanced, virus-based approach to isolate nuclei of a specific cell type for transcriptome and epigenome interrogation that decreases dependency on transgenic animals. Applying this technology to GPe parvalbumin-expressing neurons in a mouse model of Parkinson's disease, we discovered evidence for an upregulation of the oxygen homeostasis maintaining pathway involving Hypoxia-inducible factor 2α. These results provide new insight into how neuron subtypes outside the substantia nigra pars compacta may be compensating at a molecular level for differences in the motor production neural circuit during the progression of Parkinson's disease. Furthermore, they emphasize the utility of cell type-specific technologies, such as Cre-Specific Nuclear Anchored Independent Labeling, for isolated assessment of specific neuron subtypes in complex systems.


Subject(s)
Globus Pallidus/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Parkinson Disease, Secondary/metabolism , Animals , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Mice , Mice, Transgenic , Oxidopamine , Parkinson Disease, Secondary/chemically induced
5.
J Neuroinflammation ; 18(1): 175, 2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34376193

ABSTRACT

BACKGROUND: Diabetes has been recognized as a risk factor contributing to the incidence and progression of Parkinson's disease (PD). Although several hypotheses suggest a number of different mechanisms underlying the aggravation of PD caused by diabetes, less attention has been paid to the fact that diabetes and PD share pathological microvascular alterations in the brain. The characteristics of the interaction of diabetes in combination with PD at the vascular interface are currently not known. METHODS: We combined a high-fat diet (HFD) model of diabetes mellitus type 2 (DMT2) with the 6-OHDA lesion model of PD in male mice. We analyzed the association between insulin resistance and the achieved degree of dopaminergic nigrostriatal pathology. We further assessed the impact of the interaction of the two pathologies on motor deficits using a battery of behavioral tests and on microglial activation using immunohistochemistry. Vascular pathology was investigated histologically by analyzing vessel density and branching points, pericyte density, blood-brain barrier leakage, and the interaction between microvessels and microglia in the striatum. RESULTS: Different degrees of PD lesion were obtained resulting in moderate and severe dopaminergic cell loss. Even though the HFD paradigm did not affect the degree of nigrostriatal lesion in the acute toxin-induced PD model used, we observed a partial aggravation of the motor performance of parkinsonian mice by the diet. Importantly, the combination of a moderate PD pathology and HFD resulted in a significant pericyte depletion, an absence of an angiogenic response, and a significant reduction in microglia/vascular interaction pointing to an aggravation of vascular pathology. CONCLUSION: This study provides the first evidence for an interaction of DMT2 and PD at the brain microvasculature involving changes in the interaction of microglia with microvessels. These pathological changes may contribute to the pathological mechanisms underlying the accelerated progression of PD when associated with diabetes.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Dopaminergic Neurons/metabolism , Microglia/pathology , Parkinson Disease, Secondary/pathology , Pericytes/pathology , Amphetamine/pharmacology , Animals , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Insulin Resistance/physiology , Male , Mice , Microglia/drug effects , Microglia/metabolism , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Pericytes/drug effects , Pericytes/metabolism
6.
Cell Tissue Res ; 386(2): 249-260, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34370080

ABSTRACT

Patients with Parkinson's disease (PD) have a higher incidence rate of duodenal ulcers. The mucus barrier provides the first line of defense for duodenal mucosal protection. However, it is unknown whether duodenal mucus secretion is affected in PD. In the present study, we used the rats microinjected 6-hydroxydopamine (6-OHDA) into the bilateral substantia nigra to investigate duodenal mucus secretion and potential therapeutic targets in duodenal ulcer in PD. Alcian blue-periodic acid-Schiff, transmission electron microscopy, immunofluorescence, duodenal mucosal incubation, and enzyme-linked immunosorbent assays were used. The 6-OHDA rats exhibited mucin accumulation and retention in duodenal goblet cells. Mucin granules were unable to fuse with the apical membranes of goblet cells, and the exocytosis ratio of goblet cells was significantly reduced. Moreover, decreased acetylcholine and increased muscarinic receptor 2 (M2R) levels were detected in the duodenal mucosa of 6-OHDA rats. Bilateral vagotomy rats were also characterized by defective duodenal mucus secretion and decreased acetylcholine with increased M2R levels in the duodenal mucosa. Application of the cholinomimetic drug carbachol or blocking M2R with methoctramine significantly promoted mucus secretion by goblet cells and increased MUC2 content in duodenal mucosa-incubated solutions from 6-OHDA and vagotomy rats. We conclude that the reduced acetylcholine and increased M2R contribute to the impaired duodenal mucus secretion of 6-OHDA rats. The study provides new insights into the mechanism of duodenal mucus secretion and potential therapeutic targets for the treatment of duodenal ulcers in PD patients.


Subject(s)
Acetylcholine/metabolism , Intestinal Mucosa/metabolism , Mucus/metabolism , Parkinson Disease, Secondary/metabolism , Receptor, Muscarinic M2/metabolism , Animals , Duodenum/metabolism , Male , Oxidopamine , Rats , Rats, Sprague-Dawley
7.
Neurochem Res ; 46(7): 1859-1868, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33900518

ABSTRACT

MicroRNA-93 (miR-93) is an oncogene that promotes tumor growth and angiogenesis. However, its role in Parkinson's disease (PD) remains unknown. This study aimed at investigating the role of miR-93 in PD and the molecular mechanisms involved. 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced PD mouse model and lipopolysaccharide (LPS)-exposed BV2 cells were constructed. Real-time quantitative PCR was used to detect the mRNA expression of miR-93, iNOS, IL-6, IL-10, TNF-α and TGF-ß1. Bioinformatics analysis and luciferase reporter assay were used to predict and confirm the interaction between miR-93 and STAT3. Flow cytometry was used to detect cell apoptosis. Western blotting was used to detect the protein expression of STAT3. Immunohistochemistry was used to analyze the Iba1-positive and TH positive cells. It was found that the expression of miR-93 was down-regulated in LPS-exposed BV2 cells. Overexpression of miR-93 inhibited the expression of iNOS, IL-6 and TNF-α, while enhanced the expression of TGF-ß1 and IL-10. The expression of transcriptional activator 3 (STAT3) was found to be up-regulated in LPS-exposed BV2 cells. Knockdown of STAT3 inhibited the expression of iNOS, IL-6 and TNF-α, while enhanced the expression of TGF-ß1 and IL-10. Moreover, STAT3 was found to be a direct target of miR-93, and miR-93 overexpression inhibited the expression of STAT3. Furthermore, both miR-93 overexpression and STAT3 knockdown reduced LPS-induced BV2 cell apoptosis, whereas STAT3 overexpression eliminated the inhibitory effect of miR-93 on LPS-induced BV2 cell apoptosis. In addition, miR-93 overexpression inhibited MPTP-induced STAT3 expression, microglial activation and inflammatory reaction and reduced the loss of tyrosine hydroxylase in the substantia nigra of mice. In conclusion, we demonstrate that miR-93 may be involved in PD by regulating the expression of STAT3.


Subject(s)
Dopaminergic Neurons/metabolism , MicroRNAs/metabolism , Parkinson Disease, Secondary/metabolism , STAT3 Transcription Factor/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Apoptosis/physiology , Cells, Cultured , Gene Knockdown Techniques , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides , Male , Mice, Inbred C57BL , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , STAT3 Transcription Factor/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology
8.
Neurochem Res ; 46(2): 299-308, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33179210

ABSTRACT

Parkinson's disease (PD) is a severe neurodegenerative disease characterized by selective loss of dopaminergic neurons, which reduces quality of life of patients and poses a heavy burden to the society. The pathological mechanism of PD remains unclear, and increasing efforts are aimed to solve this problem. MiRNAs are a kind of small noncoding RNA regulating target gene expression. Previous studies have shown that dysregulation of miRNAs is involved in the development of PD. In the present study, we determined that miR-421 and MEF2D are increased and decreased, respectively, in a cellular model of PD. The data on the mechanism of action indicate that miR-421 directly binds to MEF2D mRNA and negatively regulates MEF2D expression. An increase in miR-421 disrupted the Bcl2/Bax system. Functional assays indicated that enhanced miR-421 promotes cell death by negative modulation of MEF2D expression. Inhibition of miR-421 or restoration of MEF2D protected neurons from neurotoxicity in cellular and animal models of PD. Our study is the first to demonstrate that miR-421 is decreased in PD models and to determine a novel putative mechanism of PD pathogenesis.


Subject(s)
Cell Death/physiology , MicroRNAs/metabolism , Neurotoxicity Syndromes/metabolism , Parkinson Disease, Secondary/metabolism , Animals , Binding Sites , Cell Line , Dopaminergic Neurons/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice, Inbred C57BL , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolism
9.
Neurochem Res ; 46(6): 1514-1539, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33719004

ABSTRACT

Gut microbial dysbiosis and alteration of gut microbiota composition in Parkinson's disease (PD) have been increasingly reported, no recognized therapies are available to halt or slow progression of PD and more evidence is still needed to illustrate its causative impact on gut microbiota and PD and mechanisms for targeted mitigation. Epidemiological evidence supported an association between milk intake and a higher incidence of Parkinson's disease (PD), questions have been raised about prospective associations between dietary factors and the incidence of PD. Here, we investigated the significance of casein in the development of PD. The mice were given casein (6.75 g/kg i.g.) for 21 days after MPTP (25 mg/kg i.p. × 5 days) treatment, the motor function, dopaminergic neurons, inflammation, gut microbiota and fecal metabolites were observed. The experimental results revealed that the mice with casein gavage after MPTP treatment showed a persisted dyskinesia, the content of dopamine in striatum and the expression of TH in midbrain and ileum were decreased, the expression of Iba-1, CD4, IL-22 in midbrain and ileum increased continuously with persisted intestinal histopathology and intestinal barrier injury. Decreased intestinal bile secretion in addition with abnormal digestion and metabolism of carbohydrate, lipids and proteins were found, whereas these pathological status for the MPTP mice without casein intake had recovered after 24 days, no significant differences were observed with regard to only treated with casein. Our study demonstrates that intestinal pathologic injury, intestinal dysbacteriosis and metabolism changes promoted by casein in MPTP mice ultimately exacerbated the lesions to dopaminergic neurons.


Subject(s)
Caseins/pharmacology , Dysbiosis/metabolism , Inflammation/metabolism , Parkinson Disease, Secondary/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Caseins/administration & dosage , Colon/drug effects , Colon/metabolism , Colon/pathology , Dopaminergic Neurons/drug effects , Dysbiosis/chemically induced , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Ileum/drug effects , Ileum/enzymology , Ileum/metabolism , Ileum/pathology , Inflammation/etiology , Intestinal Mucosa/drug effects , Male , Metabolome/drug effects , Mice, Inbred C57BL , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/complications , Pars Compacta/drug effects , Pars Compacta/enzymology , Pars Compacta/metabolism , Pars Compacta/pathology , Tight Junctions/metabolism , Tyrosine 3-Monooxygenase/metabolism
10.
J Biochem Mol Toxicol ; 35(4): e22720, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33491302

ABSTRACT

Parkinson's disease (PD) is the most common neurodegenerative movement disorder with obscure etiology and no disease-modifying therapy to date. Hence, novel, safe, and low cost-effective approaches employing medicinal plants are currently receiving increased attention. A growing body of evidence has revealed that cinnamon, being widely used as a spice of unique flavor and aroma, may exert neuroprotective effects in several neurodegenerative diseases, including PD. In vitro evidence has indicated that the essential oils of Cinnamomum species, mainly cinnamaldehyde and sodium benzoate may protect against oxidative stress-induced cell death, reactive oxygen species generation, and autophagy dysregulation, thus acting in a potentially neuroprotective manner. In vivo evidence has demonstrated that oral administration of cinnamon powder and sodium benzoate may protect against dopaminergic cell death, striatal neurotransmitter dysregulation, and motor deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse models of PD. The underlying mechanisms of its action include autophagy regulation, antioxidant effects, upregulation of Parkin, DJ-1, glial cell line-derived neurotrophic factor, as well as modulation of the TLR/NF-κB pathway and inhibition of the excessive proinflammatory responses. In addition, in vitro and in vivo studies have shown that cinnamon extracts may affect the oligomerization process and aggregation of α-synuclein. Herein, we discuss recent evidence on the novel therapeutic opportunities of this phytochemical against PD, indicating additional mechanistic aspects that should be explored, and potential obstacles/limitations that need to be overcome, for its inclusion in experimental PD therapeutics.


Subject(s)
Acrolein/analogs & derivatives , Cinnamomum zeylanicum/chemistry , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Parkinson Disease, Secondary/drug therapy , Acrolein/chemistry , Acrolein/therapeutic use , Animals , Humans , Mice , Neuroprotective Agents/chemistry , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology
11.
J Biochem Mol Toxicol ; 35(10): e22873, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34342104

ABSTRACT

Bisphenol A (BPA) is noted for its adversative effects by inducing oxidative stress, carcinogenicity, neurotoxicity, inflammation, etc. However, the likely act of BPA in inducing neurodegenerative phenotypes remains elusive in the available literature. Hence, the present study was conducted to decipher the neurodegenerative potential of BPA in inducing Parkinson's disease like phenotypes in zebrafish. Zebrafish were subjected to chronic waterborne exposure to BPA for 56 days. Locomotor activities and neurobehavioral response were assessed by the NTDT (novel tank diving test), OFT (open field test), and LDPT (light-dark preference test). The oxidative stress markers and histopathological observation for pyknosis and chromatin condensation were carried out. Immunohistochemistry for activated caspase-3 and targeted proteins expression study was performed. The basic findings reveal that chronic BPA exposure significantly induces locomotor dysfunction through a significant decline in mean velocity and total distance traveled. As a measure of pyknosis and chromatin condensation, pyknotic and Hoechst positive neurons in telencephalon and diencephalon significantly increased by BPA exposure. A higher concentration of BPA adversely affects the neurobehavioral response, antioxidant status, and neuromorphology in zebrafish. Parkinson-relevant targeted protein expression viz. alpha-synuclein and LRRK2, were significantly upregulated, whereas tyrosine hydroxylase, NeuN, and Nurr1 were significantly downregulated in the zebrafish brain. As an indicator of cell death by apoptosis, the expression of activated caspase-3 was significantly increased in the BPA-exposed zebrafish brain. These basic results of the current study indicate that chronic waterborne exposure to BPA induces neuropathological manifestation leading to the development of motor dysfunction and Parkinsonism-like neurodegenerative phenotypes in zebrafish.


Subject(s)
Behavior, Animal/drug effects , Benzhydryl Compounds/adverse effects , Brain/metabolism , Caspase 3/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Oxidative Stress/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Phenols/adverse effects , Signal Transduction/drug effects , Water Pollutants, Chemical/adverse effects , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Brain/drug effects , Female , Locomotion/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Open Field Test/drug effects , Parkinson Disease, Secondary/psychology , Phenotype
12.
Metab Brain Dis ; 36(5): 927-937, 2021 06.
Article in English | MEDLINE | ID: mdl-33656625

ABSTRACT

Parkinson's disease (PD) is a common and severe neurodegenerative disorder associated with a selective loss of dopaminergic neurons in substantia nigra pars compacta. The crucial role of oxidative stress and inflammation in PD onset and progression is evident. It has been proven that garlic extract (GE) protects the cells from oxidative stress, inflammation, mitochondrial dysfunction and apoptosis. That is, we aimed to investigate if GE reveals protective features on the preclinical model of PD. The study has been designed to evaluate both preventive (GE administered before 6-OHDA injection) and therapeutic (GE administered after 6-OHDA injection) effects of GE on the animal model. Forty male Wistar rats were divided into 4 groups including control, lesion, treatment I (received GE before 6-OHDA injection) and treatment II (received GE both before and after 6-OHDA injection). At the end of treatment, hanging, rotarod, open field and passive avoidance tests as well as immunohistochemistry were performed to evaluate the neuroprotective effects of garlic against PD. Our immunohistochemistry analysis revealed that the tyrosine hydroxylase positive cells (TH+) in GE treated groups were significantly higher (p˂0.001) than the lesion group. The motor deficiency significantly improved in hanging, rotarod, open-field and apomorphine-induced rotational tests. We observed an attenuation in memory impairment induced by PD on GE treated group. Therefore, we found that GE protects dopaminergic neurons in 6-OHDA-induced neurotoxicity and ameliorates movement disorders and behavioral deficits.


Subject(s)
Dopaminergic Neurons/drug effects , Garlic , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/drug therapy , Plant Extracts/pharmacology , Substantia Nigra/drug effects , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Male , Neuroprotective Agents/therapeutic use , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Plant Extracts/therapeutic use , Rats , Rats, Wistar , Substantia Nigra/metabolism
13.
Metab Brain Dis ; 36(1): 153-167, 2021 01.
Article in English | MEDLINE | ID: mdl-33057922

ABSTRACT

Hesperidin is a flavonoid glycoside that is frequently found in citrus fruits. Our group have demonstrated that hesperidin has neuroprotective effect in 6-hydroxydopamine (6-OHDA) model of Parkinson's disease (PD), mainly by antioxidant mechanisms. Although the pathophysiology of PD remains uncertain, a large body of evidence has demonstrated that mitochondrial dysfunction and apoptosis play a critical role in dopaminergic nigrostriatal degeneration. However, the ability of hesperidin in modulating these mechanisms has not yet been investigated. In the present study, we examined the potential of a 28-day hesperidin treatment (50 mg/kg/day, p.o.) in preventing behavioral alterations induced by 6-OHDA injection via regulating mitochondrial dysfunction, apoptosis and dopaminergic neurons in the substantia nigra pars compacta (SNpc) in C57BL/6 mice. Our results demonstrated that hesperidin treatment improved motor, olfactory and spatial memory impairments elicited by 6-OHDA injection. Moreover, hesperidin treatment attenuated the loss of dopaminergic neurons (TH+ cells) in the SNpc and the depletion of dopamine (DA) and its metabolities 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA) in the striatum of 6-OHDA-lesioned mice. Hesperidin also protected against the inhibition of mitochondrial respiratory chain complex-I, -IV and V, the decrease of Na + -K + -ATPase activity and the increase of caspase-3 and -9 activity in the striatum. Taken together, our findings indicate that hesperidin mitigates the degeneration of dopaminergic neurons in the SNpc by preventing mitochondrial dysfunction and modulating apoptotic pathways in the striatum of 6-OHDA-treated mice, thus improving behavioral alterations. These results provide new insights on neuroprotective mechanisms of hesperidin in a relevant preclinical model of PD.


Subject(s)
Apoptosis/drug effects , Behavior, Animal/drug effects , Dopaminergic Neurons/drug effects , Hesperidin/pharmacology , Mitochondria/drug effects , Parkinson Disease, Secondary/pathology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Discrimination Learning/drug effects , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Male , Maze Learning/drug effects , Mice , Mitochondria/metabolism , Mitochondria/pathology , Motor Activity/drug effects , NADH Dehydrogenase/metabolism , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Tyrosine 3-Monooxygenase/metabolism
14.
Neural Plast ; 2021: 1763533, 2021.
Article in English | MEDLINE | ID: mdl-34987572

ABSTRACT

Repetitive transcranial magnetic stimulation (rTMS) is a popular noninvasive technique for modulating motor cortical plasticity and has therapeutic potential for the treatment of Parkinson's disease (PD). However, the therapeutic benefits and related mechanisms of rTMS in PD are still uncertain. Accordingly, preclinical animal research is helpful for enabling translational research to explore an effective therapeutic strategy and for better understanding the underlying mechanisms. Therefore, the current study was designed to identify the therapeutic effects of rTMS on hemiparkinsonian rats. A hemiparkinsonian rat model, induced by unilateral injection of 6-hydroxydopamine (6-OHDA), was applied to evaluate the therapeutic potential of rTMS in motor functions and neuroprotective effect of dopaminergic neurons. Following early and long-term rTMS intervention with an intermittent theta burst stimulation (iTBS) paradigm (starting 24 h post-6-OHDA lesion, 1 session/day, 7 days/week, for a total of 4 weeks) in awake hemiparkinsonian rats, the effects of rTMS on the performance in detailed functional behavioral tests, including video-based gait analysis, the bar test for akinesia, apomorphine-induced rotational analysis, and tests of the degeneration level of dopaminergic neurons, were identified. We found that four weeks of rTMS intervention significantly reduced the aggravation of PD-related symptoms post-6-OHDA lesion. Immunohistochemically, the results showed that tyrosine hydroxylase- (TH-) positive neurons in the substantia nigra pars compacta (SNpc) and fibers in the striatum were significantly preserved in the rTMS treatment group. These findings suggest that early and long-term rTMS with the iTBS paradigm exerts neuroprotective effects and mitigates motor impairments in a hemiparkinsonian rat model. These results further highlight the potential therapeutic effects of rTMS and confirm that long-term rTMS treatment might have clinical relevance and usefulness as an additional treatment approach in individuals with PD.


Subject(s)
Gait/physiology , Motor Cortex/physiopathology , Motor Skills/physiology , Neuroprotection/physiology , Parkinson Disease, Secondary/therapy , Transcranial Magnetic Stimulation/methods , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Male , Motor Cortex/metabolism , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/physiopathology , Rats , Rats, Wistar , Tyrosine 3-Monooxygenase/metabolism
15.
Mol Cell Biochem ; 465(1-2): 89-102, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31820278

ABSTRACT

Parkinson's disease (PD) is the second common age-related neurodegenerative disease. It is characterized by control loss of voluntary movements control, resting tremor, postural instability, bradykinesia, and rigidity. The aim of the present work is to evaluate curcumin, niacin, dopaminergic and non-dopaminergic drugs in mice model of Parkinson's disease through behavioral, biochemical, genetic and histopathological observations. Mice treated with rotenone rerecorded significant increase in adenosine A2A receptor (A2AR) gene expression, α synuclein, acetylcholinesterase (AchE), malondialdehyde (MDA), angiotensin-II (Ang-II), c-reactive protein (CRP), interleukin-6 (IL-6), caspase-3 (Cas-3) and DNA fragmentation levels as compared with the control group. While, significant decrease in dopamine (DA), norepinephrine (NE), serotonin (5-HT), superoxide dismutase (SOD), reduced glutathione (GSH), ATP, succinate and lactate dehydrogenases (SDH &LDH) levels were detected. Treatment with curcumin, niacin, adenosine A2AR antagonist; ZM241385 and their combination enhanced the animals' behavior and restored all the selected parameters with variable degrees of improvement. The brain histopathological features of hippocampal and substantia nigra regions confirmed our results. In conclusion, the combination of curcumin, niacin and ZM241385 recorded the most potent treatment effect in Parkinsonism mice followed by ZM241385, as a single treatment. ZM241385 succeeded to antagonize adenosine A2A receptor by diminishing its gene expression and ameliorating all biochemical parameters under investigation. The newly investigated agent; ZM241385 has almost the same pattern of improvement as the classical drug; Sinemet®. This could shed the light to the need of detailed studies on ZM241385 for its possible role as a promising treatment against PD. Additionally, food supplements such as curcumin and niacin were effective in Parkinson's disease eradication.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Curcumin/pharmacology , Niacin/pharmacology , Parkinson Disease, Secondary , Receptor, Adenosine A2A/metabolism , Rotenone/administration & dosage , Animals , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Rotenone/pharmacology , Substantia Nigra/metabolism , Substantia Nigra/pathology
16.
Purinergic Signal ; 16(3): 379-387, 2020 09.
Article in English | MEDLINE | ID: mdl-32725400

ABSTRACT

Parkinson's disease (PD) signs and symptoms regularly include tremor. Interestingly, the nucleoside guanosine (GUO) has already proven to be effective in reducing reserpine-induced tremulous jaw movements (TJMs) in rodent models, thus becoming a promising antiparkinsonian drug. Here, we aimed at revealing the mechanism behind GUO antiparkinsonian efficacy by assessing the role of adenosine A1 and A2A receptors (A1R and A2AR) on GUO-mediated anti-tremor effects in the reserpinized mouse model of PD. Reserpinized mice showed elevated reactive oxygen species (ROS) production and cellular membrane damage in striatal slices assessed ex vivo and GUO treatment reversed ROS production. Interestingly, while the simultaneous administration of sub-effective doses of GUO (5 mg/kg) and SCH58261 (0.01 mg/kg), an A2AR antagonist, precluded reserpine-induced TJMs, these were ineffective on reverting ROS production in ex vivo experiments. Importantly, GUO was able to reduce TJM and ROS production in reserpinized mouse lacking the A2AR, thus suggesting an A2AR-independent mechanism of GUO-mediated effects. Conversely, the administration of DPCPX (0.75 mg/kg), an A1R antagonist, completely abolished both GUO-mediated anti-tremor effects and blockade of ROS production. Overall, these results indicated that GUO anti-tremor and antioxidant effects in reserpinized mice were A1R dependent but A2AR independent, thus suggesting a differential participation of adenosine receptors in GUO-mediated effects.


Subject(s)
Guanosine/therapeutic use , Parkinson Disease, Secondary/metabolism , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Tremor/metabolism , Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Guanosine/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Reactive Oxygen Species/metabolism , Tremor/chemically induced , Tremor/drug therapy , Xanthines/pharmacology
17.
Cell Biochem Funct ; 38(8): 1025-1035, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32474958

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease which results in damage in neuronal cells. Insulin-like growth factor (IGF)-1 was previously reported to play a role of neuroprotection in some diseases. Nitric oxide (NO) can also regulate neuronal cells. However, the mechanisms underlying IGF-1 and NO in PD still need to be elucidated. In present study, we explored the interaction between IGF-1 and inducible Nitric-Oxide Synthase (iNOS) in PD progression. We firstly constructed PD models by methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or MPP+ treatment. Then RT-qPCR revealed that IGF-1 expression was downregulated while iNOS expression was upregulated in MPTP model. Moreover, IGF-1 elevation or iNOS depletion enhanced cell viability and blocked cell apoptosis. Rescue assay disclosed iNOS overexpression reversed the effect on viability and apoptosis mediated by IGF-1 upregulation. Furthermore, IGF-1 was identified to positively regulate miR-302b-5p which could target iNOS. MiR-302b-5p could abolish the inhibitory function IGF-1 exerted on cell apoptosis and iNOS could counteract miR-302b-5p upregulation-triggered inhibition on cell apoptosis as well. Besides, we observed the deficiency of miR-302b-5p improved the lesioned neurobehavior of MPTP-treated mice. To sum up, present study proved that miR-302b-5p enhanced the neuroprotective effect of IGF-1 in MPTP-induced PD by regulating iNOS, recommending a novel therapeutic target for PD treatment. SIGNIFICANCE OF THE STUDY: In this study, we mainly explored that IGF-1 was decreased while iNOS was boosted in MPTP-induced PD mice model; IGF-1 suppressed while iNOS promoted MPP+ -induced toxicity and apoptosis in SH-SY5Y cells; miR-302b-5p ehanhced the neuroprotective effect of IGF-1 via targeting Inos; deficiency of miR-302b-5p improved the lesioned neurobehavior of MPTP-treated mice.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , Insulin-Like Growth Factor I/metabolism , MicroRNAs/metabolism , Neuroprotective Agents/metabolism , Nitric Oxide Synthase Type II/metabolism , Parkinson Disease, Secondary/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Line, Tumor , Humans , Insulin-Like Growth Factor I/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Nitric Oxide Synthase Type II/genetics , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/genetics
18.
Metab Brain Dis ; 35(7): 1189-1200, 2020 10.
Article in English | MEDLINE | ID: mdl-32529399

ABSTRACT

Pistachio contains polyphenolic compounds including flavonoids and anthocyanins which have antioxidant and antiinflammatory activity. Present study was aimed to evaluate the protective effects of pistachio on neurobehavioral and neurochemical changes in rats with Parkinson's disease (PD). Animal model of PD was induced by the injection of rotenone (1.5 mg/kg/day, s.c.) for 8 days. Pistachio (800 mg/kg/day, p.o.) was given for two weeks in both pre- and post-treatment. At the end of treatment brain was dissected out and striatum was isolated for biochemical and neurochemical analysis. Memory was assessed by Morris water maze (MWM) and novel object recognition (NOR) test while open field test (OFT), Kondziela inverted screen test (KIST), pole test (PT), beam walking test (BWT), inclined plane test (IPT) and footprint (FP) test were used to observe motor behavior. Rotenone administration significantly (p < 0.01) impaired the memory but pistachio in both pre- and post-treatment groups significantly (p < 0.01) improved memory performance. Rotenone-induced motor deficits were significantly attenuated in both pre- and post-pistachio treatment. Increased oxidative stress and decreased DA and 5-HT levels induced by rotenone were also significantly attenuated by pistachio supplementation. Furthermore, raised apolipoprotein E (APoE) levels in rotenone injected rats were also normalized following treatment with pistachio. Present findings show that pistachio possesses neuroprotective effects and improves memory and motor deficits via increasing DA levels and improving oxidative status in brain.


Subject(s)
Apolipoproteins E/metabolism , Corpus Striatum/drug effects , Motor Skills/drug effects , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/drug therapy , Pistacia , Plant Extracts/therapeutic use , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Plant Extracts/pharmacology , Rats , Rotenone
19.
Metab Brain Dis ; 35(3): 517-525, 2020 03.
Article in English | MEDLINE | ID: mdl-31834548

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by loss of dopaminergic neurons in substantia nigra region and the presence of α-synuclein aggregates in the striatum and surrounding areas of brain. Evidences suggest that neuroinflammation plays a role in the progression of PD. We examined the neuro-protective effects of Bacopa monnieri (BM) in regulating neuroinflammation. Administration of BM suppressed the level of pro-inflammatory cytokines, decreased the levels of α-synuclein, and reduced reactive oxygen species (ROS) generation in PD animal model. Pre-treatment of BM showed more prominent results as compare to co- and post-treatment. Results suggest that Bacopa can limit inflammation in the different areas of brain, thus, offers a promising source of novel therapeutics for the treatment of many CNS disorders.


Subject(s)
Bacopa , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Parkinson Disease, Secondary/drug therapy , Plant Extracts/therapeutic use , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chemokine CCL4/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Plant Extracts/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Rotenone , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tumor Necrosis Factor-alpha/metabolism , alpha-Synuclein/metabolism
20.
Metab Brain Dis ; 35(3): 539-548, 2020 03.
Article in English | MEDLINE | ID: mdl-32016817

ABSTRACT

Loss of dopaminergic neurons following Parkinson's disease (PD) diminishes quality of life in patients. The present study was carried out to investigate the protective effects of simultaneous inhibition of dipeptidyl peptidase-4 (DPP-4) and P2X7 purinoceptors in a PD model and explore possible mechanisms. The 6-hydroxydopamine (6-OHDA) was used as a tool to establish PD model in male Wister rats. The expressions of SIRT1, SIRT3, mTOR, PGC-1α, PTEN, P53 and DNA fragmentation were evaluated by ELISA assay. Behavioral impairments were determined using apomorphine-induced rotational and narrow beam tests. Dopamine synthesis and TH-positive neurons were detected by tyrosine hydroxylase (TH) immunohistochemistry. Neuronal density was determined by Nissl staining. OHDA-lesioned rats exhibited behavioral impairments that reversed by BBG, lin and lin + BBG. We found significant reduced levels of SIRT1, SIRT3, PGC-1α and mTOR in both mid brain and striatum from OHDA-lesioned rats that reversed by BBG, lin and lin + BBG. Likewise, significant increased levels of PTEN and P53 were found in both mid brain and striatum from OHDA-lesioned rats that was reversed by BBG, lin and lin + BBG. TH-positive neurons and neuronal density were markedly reduced OHDA-lesioned rats that reversed by BBG, lin and lin + BBG. Collectively, our results showed protective effects of simultaneous inhibition of DPP-4 and P2X7 purinoceptors in a rat model of PD can be linked to targeting SIRT1/SIRT3, PTEN-mTOR pathways. Moreover, our findings demonstrated that simultaneous inhibition of DPP-4 and P2X7 purinoceptors might have stronger effect on mitochondrial biogenesis compared to only one.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dopamine/biosynthesis , Dopaminergic Neurons/drug effects , Parkinson Disease, Secondary/drug therapy , Purinergic P2X Receptor Antagonists/therapeutic use , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , DNA Fragmentation/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Drug Therapy, Combination , Motor Activity/drug effects , Oxidopamine , PTEN Phosphohydrolase/metabolism , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Rats , Rats, Wistar , Sirtuin 1/metabolism , Sirtuin 3/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , TOR Serine-Threonine Kinases/metabolism , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL