Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 61(4): 1861-1872, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37798599

ABSTRACT

High-voltage-gated calcium channels have pivot role in the cellular and molecular mechanisms of various neurological disorders, including epilepsy. Similar to other calcium channels, P/Q-type calcium channels (Cav2.1) are also responsible for vesicle release at synaptic terminals. Up to date, there are very limited reports showing the mechanisms of Cav2.1 in epileptogenesis. In the present study, we investigated the anticonvulsive and neuroprotective effects of ω-agatoxin IVA, a specific Cav2.1 blocker, in a chemical kindling model of epileptogenesis. Righting reflex and inclined plane tests were used to assess motor coordination. Electroencephalography was recorded for electrophysiological monitoring of seizure activity in freely moving rats. Immunohistochemical analyses were performed for brain-derived neurotrophic factor (BDNF) and cleaved caspase-3 expressions in the prefrontal cortex, striatum, hippocampus, and thalamic nucleus. ω-Agatoxin IVA injected into the right lateral ventricle significantly prolonged the onset of seizures in a dose-dependent manner. In addition, repeated intraperitoneal administrations of ω-agatoxin IVA significantly suppressed the development of kindling and epileptic discharges without altering motor coordination. In addition, ω-agatoxin IVA significantly increased BDNF expressions, and decreased cleaved caspase-3 expressions in the brain when compared to PTZ + saline group. Our current study emphasizes the significance of the inhibition of P/Q type calcium channels by ω-agatoxin IVA, which suppresses the development of epileptogenesis and provides a new potential pathway for epilepsy treatment.


Subject(s)
Calcium Channel Blockers , Epilepsy , Rats , Animals , Calcium Channel Blockers/pharmacology , omega-Agatoxin IVA , Brain-Derived Neurotrophic Factor/metabolism , Caspase 3/metabolism , omega-Conotoxin GVIA/metabolism , omega-Conotoxin GVIA/pharmacology , Calcium Channels, N-Type/metabolism , Brain/metabolism , Epilepsy/metabolism , Seizures/metabolism , Calcium/metabolism
2.
Neurotoxicology ; 97: 1-11, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37146888

ABSTRACT

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons and sustained neuroinflammation due to microglial activation. Adipose tissue-derived mesenchymal stem cells (AD-MSCs) secrete neuroprotective factors to prevent neuronal damage. Furthermore, Zn regulates stem cell proliferation and differentiation and has immunomodulatory functions. Our in vivo study aimed to investigate whether Zn affects the activities of AD-MSCs in the MPTP-induced mouse model. Male C57BL/6 mice were randomly divided into six groups (n = 6): Control, Zn, PD, PD+Zn, PD+ (AD-MSC), PD+ (AD-MSC)+Zn. MPTP toxin (20 mg/kg) was dissolved in saline and intraperitoneally injected into experimental groups for two days with 12 h intervals. On the 3rd day, AD-MSCs were given to the right lateral ventricle of the PD+ (AD-MSC) and PD+ (AD-MSC)+Zn groups by stereotaxic surgery. Then, ZnSO4H2O was administered intraperitoneally for 4 days at 2 mg/kg. Seven days post MPTP injection, the motor activities of the mouse were evaluated. Then immunohistochemical analyzes were performed in SNpc. Our results showed that motor activity was lower in Group PD. AD-MSC and Zn administration have improved this impairment. MPTP caused a decrease in TH and BDNF expressions in dopaminergic neurons in Group PD. However, TH and BDNF expressions were more intense in the other groups. MCP-1, TGF-ß, and IL-10 expressions increased in administered groups compared to the Group PD. The present study indicates that Zn's individual and combined administration with AD-MSCs reduces neuronal damage in the MPTP-induced mouse model. In addition, anti-inflammatory responses that emerge with Zn and AD-MSCs may have a neuroprotective effect.


Subject(s)
Mesenchymal Stem Cells , Neuroprotective Agents , Parkinson Disease , Male , Animals , Mice , Parkinson Disease/therapy , Parkinson Disease/metabolism , Zinc/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Mice, Inbred C57BL , Dopaminergic Neurons , Mesenchymal Stem Cells/metabolism , Disease Models, Animal , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism
3.
Int J Mol Sci ; 23(18)2022 Sep 06.
Article in English | MEDLINE | ID: mdl-36142156

ABSTRACT

Breast cancer is the second most common cancer in women. The roles of the SIRT and FoxO proteins in tumor progression are known, but their roles in metastasis have not yet been clearly elucidated. In our study, we investigated the roles of SIRT and FoxO proteins their downstream pathways, proteins p21 and p53, in tumor progression and metastasis. We evaluated these proteins in vitro using metastatic 4TLM and 67NR cell lines, as well as their expression levels in tumor-bearing mice. In addition, the regulatory role of SIRT and FoxO proteins in different transduction cascades was examined by IPA core analysis, and clinicopathological evidence was investigated in the TCGA database. In primary tumors, the expression levels of SIRT1, p21, p53, E2F1 and FoxO proteins were higher in 67NR groups. In metastatic tissues, the expression levels of SIRT1, E2F1 and FoxO proteins were found to be enhanced, whereas the levels of p53 and p21 expression were noted to be reduced. IPA analysis also provided empirical evidence of the mechanistic involvement of SIRT and FoxO proteins in tumor progression and metastasis. In conclusion, SIRT1 was found to co-operate with FoxO proteins and to play a critical role in metastasis. Additional research is required to determine why overexpression of SIRT1 in metastatic tissues has oncogenic effects.


Subject(s)
Breast Neoplasms , Sirtuin 1 , Animals , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Female , Humans , Mice , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Neuropharmacology ; 208: 108977, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35092748

ABSTRACT

Parkinson's disease (PD) is characterized by motor and non-motor symptoms associated with dopaminergic and non-dopaminergic injury. Vortioxetine is a multimodal serotonergic antidepressant with potential procognitive effects. This study aimed to explore the effects of vortioxetine on motor functions, spatial learning and memory, and depression-like behavior in the rotenone-induced rat model of PD. Male Sprague-Dawley rats were daily administered with the rotenone (2 mg kg-1, s.c.) and/or vortioxetine (10 mg kg-1, s.c.) for 28 days. Motor functions (rotarod, catalepsy, open-field), depression-like behaviors (sucrose preference test), anxiety (elevated plus maze), and spatial learning and memory abilities (novel object recognition and Morris water maze) were evaluated in behavioral tests. Then immunohistochemical, neurochemical, and biochemical analysis on specific brain areas were performed. Vortioxetine treatment markedly reduced rotenone-induced neurodegeneration, improved motor and cognitive dysfunction, decreased depression-like behaviors without affecting anxiety-like parameters. Vortioxetine also restored the impaired inflammatory response and affected neurotransmitter levels in brain tissues. Interestingly, vortioxetine was thought to trigger a sort of dysfunction in basal ganglia as evidenced by increased Toll-like receptor-2 (TLR-2) and decreased TH immunoreactivity only in substantia nigra tissue of PD rats compared to the control group. The present study indicates that vortioxetine has beneficial effects on motor dysfunction as well as cognitive impairment associated with neurodegeneration in the rotenone-induced PD model. Possible mechanisms underlying these beneficial effects cover TLR-2 inhibition and neurochemical restoration of vortioxetine.


Subject(s)
Cognitive Dysfunction , Parkinson Disease , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Disease Models, Animal , Male , Neuroinflammatory Diseases , Rats , Rats, Sprague-Dawley , Rotenone/toxicity , Toll-Like Receptor 2 , Vortioxetine
5.
Neurotoxicology ; 88: 168-177, 2022 01.
Article in English | MEDLINE | ID: mdl-34808223

ABSTRACT

Neuroinflammation has an essential role in various neurodegenerative diseases including Parkinson's disease (PD). Microglial activation as a result of neuroinflammation exacerbates the pathological consequences of the disease. The toxic effect of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) causes alpha-synuclein (α-synuclein) accumulation, which leads to dopaminergic neuron death in the MPTP-induced mouse model. Toll-like receptor 4 (TLR4) stimulates release of cytokine through NF-kB by activating glial cells, thus resulting in the death of dopaminergic neurons. Melatonin has the ability to cross the blood-brain barrier and protect neurons through anti-inflammatory properties. We hypothesized that melatonin could suppress TLR4-mediated neuroinflammation, decrease cytokine release due to the inflammatory response, and reduce dopaminergic neuron loss in the MPTP-induced mouse model. In the MPTP-induced mouse model, we aimed to assess the neuroinflammatory responses caused by TLR4 activation as well as the effect of melatonin on these responses. Three-month-old male C57BL/6 mice were randomly divided into five groups; Control (Group-C), Sham (Group-S), Melatonin-treated (Group-M), MPTP-injected (Group-P), and MPTP + melatonin-injected (Group-P + M). MPTP toxin (20 mg/kg) was dissolved in saline and intraperitoneally (i.p.) injected to mice for two days with 12 h intervals. The total dose per mouse was 80 mg/kg. Melatonin was administered (20 mg/kg) intraperitoneally to Group-M and Group-P + M twice a day for five days. Eight days after starting the experiment, the motor activities of mice were evaluated by locomotor activity tests. The effects on dopamine neurons in the SNPc was determined by tyrosine hydroxylase (TH) immunohistochemistry. TLR4, α-synuclein, and p65 expression was evaluated by immunostaining as well. The amount of TNF-alpha in the total brain was evaluated by western blot analysis. In our results seen that locomotor activity was lower in Group-P compared to Group-C. However, melatonin administration was improved this impairment. MPTPcaused decrease in TH immuno-expression in dopaminergic neurons in Group-P. TLR4 (p < 0.001), α-synuclein (p < 0.001), and p65 (p < 0.01) immuno-expressions were also decreased in Group-P+M compared to Group-P (using MPTP). TNF-α expression was lower in Group-C, Group-S, Group-M, and Group-P+M, when compared to Group-P (p < 0.0001) due to the absence of inflammatory response. In conclusion, our study revealed that melatonin administration reduced α-synuclein aggregation and TLR4-mediated inflammatory response in the MPTP-induced mouse model.


Subject(s)
Melatonin/metabolism , Neuroinflammatory Diseases/chemically induced , Parkinsonian Disorders/chemically induced , Toll-Like Receptor 4/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/metabolism , Parkinsonian Disorders/metabolism , Tropolone/analogs & derivatives , Tropolone/pharmacology
6.
Daru ; 29(1): 85-99, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33469802

ABSTRACT

PURPOSE: This study investigated whether thymoquinone (TQ) could alleviate central nervous system (CNS) and cardiovascular toxicity of prilocaine, a commonly used local anesthetic. METHODS: Rats were randomized to the following groups: control, prilocaine treated, TQ treated and prilocaine + TQ treated. Electroencephalography and electrocardiography electrodes were placed and trachea was intubated. Mechanical ventilation was initiated, right femoral artery was cannulated for continuous blood pressure measurements and blood-gas sampling while the left femoral vein was cannulated for prilocaine infusion. Markers of myocardial injury, reactive oxygen/nitrogen species (ROS/RNS) generation and total antioxidant capacity (TAC) were assayed by standard kits. Aquaporin-4 (AQP4), nuclear factor(NF)κB-p65 and -p50 subunit in brain tissue were evaluated by histological scoring. RESULTS: Blood pH and partial oxygen pressure, was significantly decreased after prilocaine infusion. The decrease in blood pH was alleviated in the prilocaine + TQ treated group. Prilocaine produced seizure activity, cardiac arrhythmia and asystole at significantly lower doses compared to prilocaine + TQ treated rats. Thymoquinone administration attenuated levels of myocardial injury induced by prilocaine. Prilocaine treatment caused increased ROS/RNS formation and decreased TAC in heart and brain tissue. Thymoquinone increased heart and brain TAC and decreased ROS/RNS formation in prilocaine treated rats. AQP4, NFκB-p65 and NFκB-p50 expressions were increased in cerebellum, cerebral cortex, choroid plexus and thalamic nucleus in prilocaine treated rats. Thymoquinone, decreased the expression of AQP4, NFκB-p65 and NFκB-p50 in brain tissue in prilocaine + TQ treated rats. CONCLUSION: Results indicate that TQ could ameliorate prilocaine-induced CNS and cardiovascular toxicity.


Subject(s)
Anticonvulsants/therapeutic use , Benzoquinones/therapeutic use , Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Epilepsy/drug therapy , Neuroprotective Agents/therapeutic use , Prilocaine , Animals , Anticonvulsants/pharmacology , Aquaporin 4/metabolism , Benzoquinones/pharmacology , Blood Pressure/drug effects , Brain/drug effects , Brain/metabolism , Cardiotonic Agents/pharmacology , Cardiotoxicity/metabolism , Cardiotoxicity/physiopathology , Epilepsy/chemically induced , Epilepsy/metabolism , Epilepsy/physiopathology , Heart/drug effects , Heart/physiology , Heart Rate/drug effects , Male , Myocardium/metabolism , NF-kappa B p50 Subunit/metabolism , Neuroprotective Agents/pharmacology , Rats, Wistar , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor RelA/metabolism
7.
Pharmacol Rep ; 72(5): 1397-1406, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32207092

ABSTRACT

BACKGROUND: Metformin, a widely prescribed antidiabetic drug, has been suggested to have a neuroprotective effect on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity in mice. In this study, we investigated the neuroprotective potential of metformin against rotenone-induced dopaminergic neuron damage and its underlying mechanisms. METHODS: C57BL/6 mice were given saline or rotenone (2.5 mg/kg/day, ip) injection for 10 days. Metformin treatment (300 mg/kg/day, ip) was started concurrently with rotenone administration and continued for 10 days. The neuroprotective effect of metformin on rotenone-induced dopaminergic toxicity was assessed by tyrosine hydroxylase (TH), cleaved caspase-3 and α-synuclein immunohistochemistry in substantia nigra (SN). SN tissues were extracted for biochemical analysis. Malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) protein levels were measured by spectrophotometric assay. RESULTS: We found that metformin treatment attenuated the rotenone-induced loss of TH+ neurons in the SN. Additionally, metformin significantly decreased the rotenone-induced increase of cleaved caspase-3 and α-synuclein accumulation in the SN; however, there was no difference in motor behaviours between the experimental groups. Meanwhile, the levels of MDA and 4-HNE in SN were significantly reduced in the rotenone-metformin group compared to the rotenone group. CONCLUSIONS: Results showed that metformin treatment attenuated dopaminergic neuron loss in SN induced by rotenone by decreasing lipid peroxidation.


Subject(s)
Dopaminergic Neurons/drug effects , Lipid Peroxidation/drug effects , Metformin/pharmacology , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Rotenone/pharmacology , Animals , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Male , Mice , Mice, Inbred C57BL , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...