Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 452
Filter
1.
Acta Neurobiol Exp (Wars) ; 84(3): 266-274, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39392022

ABSTRACT

Evidence is provided that the glycosylated flavonoid vitexin (apigenin­8­C­beta­D­glucopyranoside) attenuates pentylenetetrazole (PTZ)­induced acute tonic­clonic seizures in rats. However, the effects of chronic and systemic vitexin in PTZ­kindled rats remain unknown. The aim of this work was to investigate the effect of long­term treatment with vitexin in the PTZ­kindling model of epilepsy. Male Wistar rats received intraperitoneal injections of PTZ at a subconvulsive dose of 35 mg/kg every other day for 29 days. Either saline containing dimethyl sulfoxide - DMSO 1%  (vehicle), diazepam (2 mg/kg; positive control) or vitexin (2.5 mg/kg) was administered intraperitoneally 30 min before each PTZ injection. The behavioral reactions were recorded by 30 min immediately after each PTZ injection. Furthermore, on the 31st day, that is, 48 h after the latter dose of PTZ, the animals were euthanized and renal and hepatic biochemical markers were evaluated in blood serum. Chronic treatment with either diazepam or vitexin attenuated the seizures provoked by PTZ injections. Neither diazepam nor vitexin caused changes in renal levels of creatinine and urea and in hepatic levels of aspartate aminotransferase and alanine aminotransferase. Our findings suggest that chronic administration of vitexin attenuates the progression of PTZ­induced kindling without causing side effects on kidneys and liver.


Subject(s)
Anticonvulsants , Apigenin , Diazepam , Kindling, Neurologic , Pentylenetetrazole , Rats, Wistar , Seizures , Animals , Male , Apigenin/pharmacology , Anticonvulsants/pharmacology , Kindling, Neurologic/drug effects , Diazepam/pharmacology , Seizures/drug therapy , Seizures/chemically induced , Disease Models, Animal , Rats , Time Factors , Convulsants/toxicity , Epilepsy/drug therapy , Epilepsy/chemically induced
2.
Sci Rep ; 14(1): 22695, 2024 09 30.
Article in English | MEDLINE | ID: mdl-39349563

ABSTRACT

Epilepsy, frequently comorbid with anxiety, is a prevalent neurological disorder. Available drugs often have side effects that hinder adherence, creating a need for new treatments. Potassium channel activators have emerged as promising candidates for treating both epilepsy and anxiety. This study aimed to evaluate the potential anticonvulsant and anxiolytic effects of pinacidil, an ATP-sensitive potassium channel activator used as antihypertensive, in rats. Our results indicate that pinacidil at 10 mg/kg (i.p.) fully protected animals from seizures induced by pentylenetetrazol (PTZ) and provided 85.7%, 100% and 100% protection against pilocarpine-induced seizures at 2.5, 5 and 10 mg/kg (i.p.), respectively. Although the 2.5 and 5 mg/kg (i.p) doses did not significantly protect the animals from PTZ-induced seizures, they did significantly increase the latency to the first seizure. Pinacidil also demonstrated mild anxiolytic activity, particularly at 10 mg/kg (i.p), evidenced by increased time spent in the open or illuminated areas of the Elevated Plus Maze (EPM) and Light-Dark Box (LDB) and increased exploratory activity in the Open Filed, EPM and LDB. Pinacidil did not affect locomotor performance, supporting its genuine anticonvulsant effects. This study holds significant medical and pharmaceutical value by characterizing pinacidil's anticonvulsant and anxiolytic effects and highlighting its potential for therapeutic repositioning.


Subject(s)
Anti-Anxiety Agents , Anticonvulsants , Disease Models, Animal , Pentylenetetrazole , Pinacidil , Seizures , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Male , Seizures/drug therapy , Seizures/chemically induced , Mice , Rats , Pinacidil/pharmacology , Drug Repositioning , Anxiety/drug therapy , Pilocarpine , Behavior, Animal/drug effects , Rats, Wistar
3.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337323

ABSTRACT

This work provides insight into carbamazepine polymorphs (Forms I, II, III, IV, and V), with reports on the cytoprotective, exploratory, motor, CNS-depressant, and anticonvulsant properties of carbamazepine (CBZ), carbamazepine formulation (CBZ-F), topiramate (TOP), oxcarbazepine (OXC), and diazepam (DZP) in mice. Structural analysis highlighted the significant difference in molecular conformations, which directly influence the physicochemical properties; and density functional theory description provided indications about CBZ reactivity and stability. In addition to neuron viability assessment in vitro, animals were treated orally with vehicle 10 mL/kg, as well as CBZ, CBZ-F, TOP, OXC, and DZP at the dose of 5 mg/kg and exposed to open-field, rotarod, barbiturate sleep induction and pentylenetetrazol (PTZ 70 mg/kg)-induced seizure. The involvement of GABAergic mechanisms in the activity of these drugs was evaluated with the intraperitoneal pretreatment of flumazenil (2 mg/kg). The CBZ, CBZ-F, and TOP mildly preserved neuronal viability. The CBZ-F and the reference AEDs potentiated barbiturate sleep, altered motor activities, and attenuated PTZ-induced convulsion. However, flumazenil pretreatment blocked these effects. Additional preclinical assessments could further establish the promising utility of CBZ-F in clinical settings while expanding the scope of AED formulations and designs.


Subject(s)
Anticonvulsants , Carbamazepine , Carbamazepine/pharmacology , Carbamazepine/analogs & derivatives , Animals , Mice , Anticonvulsants/pharmacology , Seizures/drug therapy , Seizures/chemically induced , Neurons/drug effects , Neurons/metabolism , Oxcarbazepine/pharmacology , Diazepam/pharmacology , Male , Pentylenetetrazole , Cell Survival/drug effects , Topiramate/pharmacology , Barbiturates/pharmacology
4.
Int J Mol Sci ; 25(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273262

ABSTRACT

Epilepsy is a disorder characterized by a predisposition to generate seizures. Levetiracetam (LEV) is an antiseizure drug that has demonstrated oxidant-antioxidant effects during the early stages of epilepsy in several animal models. However, the effect of LEV on oxidant-antioxidant activity during long-term epilepsy has not been studied. Therefore, the objective of the present study was to determine the effects of LEV on the concentrations of five antioxidant enzymes and on the levels of four oxidant stress markers in the hippocampus of rats with temporal lobe epilepsy at 5.7 months after status epilepticus (SE). The results revealed that superoxide dismutase (SOD) activity was significantly greater in the epileptic group (EPI) than in the control (CTRL), CTRL + LEV and EPI + LEV groups. No significant differences were found among the groups' oxidant markers. However, the ratios of SOD/hydrogen peroxide (H2O2), SOD/glutathione peroxidase (GPx) and SOD/GPx + catalase (CAT) were greater in the EPI group than in the CTRL and EPI + LEV groups. Additionally, there was a positive correlation between SOD activity and GPx activity in the EPI + LEV group. LEV-mediated modulation of the antioxidant system appears to be time dependent; at 5.7 months after SE, the role of LEV may be as a stabilizer of the redox state.


Subject(s)
Antioxidants , Catalase , Epilepsy, Temporal Lobe , Glutathione Peroxidase , Levetiracetam , Oxidative Stress , Superoxide Dismutase , Animals , Levetiracetam/pharmacology , Levetiracetam/therapeutic use , Rats , Antioxidants/metabolism , Antioxidants/pharmacology , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/metabolism , Male , Superoxide Dismutase/metabolism , Oxidative Stress/drug effects , Glutathione Peroxidase/metabolism , Catalase/metabolism , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Oxidants/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Disease Models, Animal , Hydrogen Peroxide/metabolism , Rats, Wistar
5.
Toxins (Basel) ; 16(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39195752

ABSTRACT

Epilepsy, a neurological disorder characterized by excessive neuronal activity and synchronized electrical discharges, ranks among the most prevalent global neurological conditions. Despite common use, antiepileptic drugs often result in adverse effects and lack effectiveness in controlling seizures in temporal lobe epilepsy (TLE) patients. Recent research explored the potential of occidentalin-1202, a peptide inspired by Polybia occidentalis venom, in safeguarding Wistar rats from chemically induced seizures. The present study evaluated the new analog from occidentalin-1202 named NOR-1202 using acute and chronic pilocarpine-induced models and an acute kainic acid (KA) male mice model. NOR-1202 was administered through the intracerebroventricular (i.c.v.), subcutaneous, or intraperitoneal routes, with stereotaxic procedures for the i.c.v. injection. In the acute pilocarpine-induced model, NOR-1202 (i.c.v.) protected against generalized seizures and mortality but lacked systemic antiepileptic activity. In the KA model, it did not prevent generalized seizures but improved survival. In the chronic TLE model, NOR-1202's ED50 did not differ significantly from the epileptic or healthy groups regarding time spent in spontaneous recurrent seizures during the five-day treatment. However, the NOR-1202 group exhibited more seizures than the healthy group on the second day of treatment. In summary, NOR-1202 exhibits antiepileptic effects against chemoconvulsant-induced seizures, but no effect was observed when administered systemically.


Subject(s)
Anticonvulsants , Disease Models, Animal , Seizures , Animals , Male , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Mice , Seizures/drug therapy , Seizures/chemically induced , Pilocarpine , Kainic Acid/analogs & derivatives , Peptides/pharmacology , Peptides/therapeutic use , Peptides/administration & dosage , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/chemically induced
6.
Exp Mol Pathol ; 139: 104921, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39096892

ABSTRACT

Seizures are neurological disorders triggered by an imbalance in the activity of excitatory and inhibitory neurotransmitters in the brain. When triggered chronically, this imbalance can lead to epilepsy. Critically, many of the affected individuals are refractory to treatment. Given this, anti-inflammatory drugs, in particular glucocorticoids, have been considered as a potential antiepileptogenic therapy. Glucocorticoids are currently used in the treatment of refractory patients, although there have been contradictory results in terms of their use in association with antiepileptic drugs, which reinforces the need for a more thorough investigation of their effects. In this context, the present study evaluated the effects of dexamethasone (DEX, 0.6 mg/kg) on the electroencephalographic (EEG) and histopathological parameters of male Wistar rats submitted to acute seizure induced by pentylenetetrazol (PTZ). The EEG monitoring revealed that DEX reduced the total brainwave power, in comparison with PTZ, in 12 h after the convulsive episode, exerting this effect in up to 36 h (p < 0.05 for all comparisons). An increase in the accommodation of the oscillations of the delta, alpha, and gamma frequencies was also observed from the first 12 h onwards, with the accommodation of the theta frequency occurring after 36 h, and that of the beta frequency 24 h after the seizure. The histopathological analyses showed that the CA3 region and hilum of the hippocampus suffered cell loss after the PTZ-induced seizure (control vs. PTZ, p < 0.05), although DEX was not able to protect these regions against cell death (PTZ vs. DEX + PTZ, p > 0.05). While DEX did not reverse the cell damage caused by PTZ, the data indicate that DEX has beneficial properties in the EEG analysis, which makes it a promising candidate for the attenuation of the epileptiform wave patterns that can precipitate refractory seizures.


Subject(s)
Brain Waves , Dexamethasone , Electroencephalography , Pentylenetetrazole , Rats, Wistar , Seizures , Animals , Dexamethasone/pharmacology , Dexamethasone/adverse effects , Pentylenetetrazole/toxicity , Seizures/drug therapy , Seizures/chemically induced , Seizures/physiopathology , Male , Rats , Brain Waves/drug effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Anticonvulsants/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology
7.
Expert Opin Drug Discov ; 19(8): 975-990, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38963148

ABSTRACT

INTRODUCTION: Despite the availability of around 30 antiseizure medications, 1/3 of patients with epilepsy fail to become seizure-free upon pharmacological treatment. Available medications provide adequate symptomatic control in two-thirds of patients, but disease-modifying drugs are still scarce. Recently, though, new paradigms have been explored. AREAS COVERED: Three areas are reviewed in which a high degree of innovation in the search for novel antiseizure and antiepileptogenic medications has been implemented: development of novel screening approaches, search for novel therapeutic targets, and adoption of new drug discovery paradigms aligned with a systems pharmacology perspective. EXPERT OPINION: In the past, worldwide leaders in epilepsy have reiteratively stated that the lack of progress in the field may be explained by the recurrent use of the same molecular targets and screening procedures to identify novel medications. This landscape has changed recently, as reflected by the new Epilepsy Therapy Screening Program and the introduction of many in vitro and in vivo models that could possibly improve our chances of identifying first-in-class medications that may control drug-resistant epilepsy or modify the course of disease. Other milestones include the study of new molecular targets for disease-modifying drugs and exploration of a systems pharmacology perspective to design new drugs.


Subject(s)
Anticonvulsants , Drug Discovery , Epilepsy , Humans , Anticonvulsants/pharmacology , Drug Discovery/methods , Epilepsy/drug therapy , Animals , Drug Development/methods , Molecular Targeted Therapy , Network Pharmacology , Drug Resistant Epilepsy/drug therapy
8.
Curr Neuropharmacol ; 22(13): 2168-2190, 2024.
Article in English | MEDLINE | ID: mdl-38847378

ABSTRACT

Epilepsy is a neurological disease with no defined cause, characterized by recurrent epileptic seizures. These occur due to the dysregulation of excitatory and inhibitory neurotransmitters in the central nervous system (CNS). Psychopharmaceuticals have undesirable side effects; many patients require more than one pharmacotherapy to control crises. With this in mind, this work emphasizes the discovery of new substances from natural products that can combat epileptic seizures. Using in silico techniques, this review aims to evaluate the antiepileptic and multi-target activity of phenylpropanoid derivatives. Initially, ligand-based virtual screening models (LBVS) were performed with 468 phenylpropanoid compounds to predict biological activities. The LBVS were developed for the targets alpha- amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), voltage-gated calcium channel Ttype (CaV), gamma-aminobutyric acid A (GABAA), gamma-aminobutyric acid transporter type 1 (GAT-1), voltage-gated potassium channel of the Q family (KCNQ), voltage-gated sodium channel (NaV), and N-methyl D-aspartate (NMDA). The compounds that had good results in the LBVS were analyzed for the absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters, and later, the best molecules were evaluated in the molecular docking consensus. The TR430 compound showed the best results in pharmacokinetic parameters; its oral absorption was 99.03%, it did not violate any Lipinski rule, it showed good bioavailability, and no cytotoxicity was observed either from the molecule or from the metabolites in the evaluated parameters. TR430 was able to bind with GABAA (activation) and AMPA (inhibition) targets and demonstrated good binding energy and significant interactions with both targets. The studied compound showed to be a promising molecule with a possible multi-target activity in both fundamental pharmacological targets for the treatment of epilepsy.


Subject(s)
Anticonvulsants , Epilepsy , Humans , Epilepsy/drug therapy , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Anticonvulsants/chemistry , Molecular Docking Simulation
9.
ChemMedChem ; 19(15): e202400135, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38687623

ABSTRACT

Tetrahydrolinalool (THL) is an acyclic monoterpene alcohol, produced during linalol metabolism and also a constituent of essential oils. As described in the literature, many monoterpenes present anticonvulsant properties, and thus we became interested in evaluating the anticonvulsant activity of Tetrahydrolinalool using in mice model as well as in silico approaches. Our results demonstrated that THL increased latency to seizure onset and also reduced the mortality, in picrotoxin induced seizure tests. The results may be related to GABAergic regulation, which was also suggested in seizure testing induced by 3-mercapto-propionic acid. In the strychnine-induced seizure testing, none of the groups pretreated with THL modulated the parameters indicative of anticonvulsant effect. The electrophysiological results revealed that THL treatment reduces seizures induced by pentylenetetrazole. The in silico molecular docking studies showed that the interaction between THL and a GABAA receptor model formed a stable complex, in comparison to the crystaligraphic structure of diazepam, a structurally related ligand. In conclusion, all the evidences showed that THL presents effective anticonvulsant activity related to the GABAergic pathway, being a candidate for treatment of epileptic syndromes.


Subject(s)
Acyclic Monoterpenes , Anticonvulsants , Molecular Docking Simulation , Monoterpenes , Pentylenetetrazole , Seizures , Anticonvulsants/pharmacology , Anticonvulsants/chemistry , Anticonvulsants/chemical synthesis , Animals , Mice , Seizures/drug therapy , Monoterpenes/pharmacology , Monoterpenes/chemistry , Monoterpenes/chemical synthesis , Acyclic Monoterpenes/pharmacology , Acyclic Monoterpenes/chemistry , Acyclic Monoterpenes/chemical synthesis , Male , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Structure-Activity Relationship , Behavior, Animal/drug effects , Picrotoxin/pharmacology
10.
Naunyn Schmiedebergs Arch Pharmacol ; 397(9): 6989-6999, 2024 09.
Article in English | MEDLINE | ID: mdl-38625552

ABSTRACT

Epilepsy is a condition marked by sudden, self-sustained, and recurring brain events, showcasing unique electro-clinical and neuropathological phenomena that can alter the structure and functioning of the brain, resulting in diverse manifestations. Antiepileptic drugs (AEDs) can be very effective in 30% of patients in controlling seizures. Several factors contribute to this: drug resistance, individual variability, side effects, complexity of epilepsy, incomplete understanding, comorbidities, drug interactions, and no adherence to treatment. Therefore, research into new AEDs is important for several reasons such as improved efficacy, reduced side effects, expanded treatment options, treatment for drug-resistant epilepsy, improved safety profiles, targeted therapies, and innovation and progress. Animal models serve as crucial biological tools for comprehending neuronal damage and aiding in the discovery of more effective new AEDs. The utilization of antioxidant agents that act on the central nervous system may serve as a supplementary approach in the secondary prevention of epilepsy, both in laboratory animals and potentially in humans. Chlorogenic acid (CGA) is a significant compound, widely prevalent in numerous medicinal and food plants, exhibiting an extensive spectrum of biological activities such as neuroprotection, antioxidant, anti-inflammatory, and analgesic effects, among others. In this research, we assessed the neuroprotective effects of commercially available CGA in Wistar rats submitted to lithium-pilocarpine-induced status epilepticus (SE) model. After 72-h induction of SE, rats received thiopental and were treated for three consecutive days (1st, 2nd, and 3rd doses). Next, brains were collected and studied histologically for viable cells in the hippocampus with staining for cresyl-violet (Nissl staining) and for degenerating cells with Fluoro-Jade C (FJC) staining. Moreover, to evaluate oxidative stress, the presence of malondialdehyde (MDA) and superoxide dismutase (SOD) was quantified. Rats administered with CGA (30 mg/kg) demonstrated a significant decrease of 59% in the number of hippocampal cell loss in the CA3, and of 48% in the hilus layers after SE. A significant reduction of 75% in the cell loss in the CA3, shown by FJC+ staining, was also observed with the administration of CGA (30 mg/kg). Furthermore, significant decreases of 49% in MDA production and 72% in the activity of SOD were seen, when compared to animals subjected to SE that received vehicle. This study introduces a novel finding: the administration of CGA at a dosage of 30 mg/kg effectively reduced oxidative stress induced by lithium-pilocarpine, with its effects lasting until the peak of neural damage 72 h following the onset of SE. Overall, the research and development of new AEDs are essential for advancing epilepsy treatment, improving patient outcomes, and ultimately enhancing the quality of life for individuals living with epilepsy.


Subject(s)
Chlorogenic Acid , Neuroprotective Agents , Oxidative Stress , Pilocarpine , Rats, Wistar , Status Epilepticus , Animals , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Pilocarpine/toxicity , Oxidative Stress/drug effects , Chlorogenic Acid/pharmacology , Chlorogenic Acid/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Male , Rats , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Disease Models, Animal , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Lithium/pharmacology
11.
Neurol Res ; 46(8): 717-726, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38679045

ABSTRACT

Introduction: The close relationship between inflammatory processes and epileptic seizures is already known, although the exact pathophysiological mechanism is unclear. In this study, the anticonvulsant capacity of piroxicam, an anti-inflammatory drug, was evaluated. A rat pentylenetetrazole kindling model was used.Methods: Male Wistar rats, 8-9 weeks old, received piroxicam (0.15 and 0.30 mg/kg), diazepam (2 mg/kg) or saline for 14 days, and PTZ, on alternate days. Intraperitoneal was chosen as the route of administration. The intensity of epileptic seizures was assessed using a modified Racine scale. The open field test and object recognition analysis were performed at the beginning of the study to ensure the safety of the drugs used. At the end of the protocol, the animals were euthanized to measure the levels of inflammatory (TNF-a and IL-6) and anti-inflammatory (IL-10) cytokines in the cortex, hippocampus, and serum.Results:There were no changes in the open field test and object recognition analysis. Piroxicam was found to decrease Racine scale scores at both concentrations. The reported values for IL-6 levels remained steady in all structures, whereas the TNF-alpha level in the cortex was higher in animals treated with piroxicam than in the saline and diazepam subjects. Finally, animals treated with the anti-inflammatory drug presented reduced IL-10 levels in the cortex and hippocampus.onclusions: Using inflammation as a guiding principle, the anticonvulsant effect of PIRO could be associated with the hippocampal circuits, since this structure showed no increase in inflammatory cytokines.


Subject(s)
Anticonvulsants , Disease Models, Animal , Kindling, Neurologic , Piroxicam , Rats, Wistar , Animals , Piroxicam/pharmacology , Male , Kindling, Neurologic/drug effects , Anticonvulsants/pharmacology , Rats , Pentylenetetrazole , Seizures/drug therapy , Cytokines/metabolism , Diazepam/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Dose-Response Relationship, Drug , Epilepsy/drug therapy
12.
Acta Neurobiol Exp (Wars) ; 84(1): 51-58, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38587324

ABSTRACT

Levetiracetam (LEV) is a drug commonly used as an anticonvulsant. However, recent evidence points to a possible role as an antioxidant. We previously demonstrated the antioxidant properties of LEV by significantly increasing catalase and superoxide dismutase activities and decreasing the hydrogen peroxide (H2O2) levels in the hippocampus of rats with temporal lobe epilepsy (TLE) showing scavenging properties against the hydroxyl radical. The aim of the present work was to evaluate, the effect of LEV on DNA oxidation, by determining 8­hydroxy­2­deoxyguanosine (8­OHdG) levels, and glutathione content, through reduced (GSH) and oxidized (GSSG) glutathione levels, in the hippocampus of rats with TLE. Male Wistar rats were assigned to the control (CTRL), CTRL+LEV, epileptic (EPI) and EPI+LEV groups. TLE was induced using the lithium­pilocarpine model. Thirteen weeks after TLE induction, LEV was administered for one week through osmotic pumps implanted subcutaneously. The determination of 8­OHdG, GSH and GSSG levels were measured using spectrophotometric methods. We showed that LEV alone significantly increased 8­OHdG and GSSG levels in the hippocampus of control rats compared to those in epileptic condition. No significant differences in GSH levels were observed. LEV could induce changes in the hippocampus increasing DNA oxidation and GSSG levels under nonepileptic condition but not protecting against the mitochondrial dysfunction observed in TLE probably by mechanisms related to changes in chromatin structure, neuroinflammation and alterations in redox components.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Piracetam , Male , Rats , Animals , Levetiracetam/adverse effects , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/drug therapy , Piracetam/adverse effects , Antioxidants/therapeutic use , Glutathione Disulfide/adverse effects , Hydrogen Peroxide/adverse effects , Rats, Wistar , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Glutathione/metabolism , Oxidation-Reduction
13.
Int J Mol Sci ; 25(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38338984

ABSTRACT

Temporal lobe epilepsy (TLE) is one of the most common forms of focal epilepsy. Levetiracetam (LEV) is an antiepileptic drug whose mechanism of action at the genetic level has not been fully described. Therefore, the aim of the present work was to evaluate the relevant gene expression changes in the dentate gyrus (DG) of LEV-treated rats with pilocarpine-induced TLE. Whole-transcriptome microarrays were used to obtain the differential genetic profiles of control (CTRL), epileptic (EPI), and EPI rats treated for one week with LEV (EPI + LEV). Quantitative RT-qPCR was used to evaluate the RNA levels of the genes of interest. According to the results of the EPI vs. CTRL analysis, 685 genes were differentially expressed, 355 of which were underexpressed and 330 of which were overexpressed. According to the analysis of the EPI + LEV vs. EPI groups, 675 genes were differentially expressed, 477 of which were downregulated and 198 of which were upregulated. A total of 94 genes whose expression was altered by epilepsy and modified by LEV were identified. The RT-qPCR confirmed that LEV treatment reversed the increased expression of Hgf mRNA and decreased the expression of the Efcab1, Adam8, Slc24a1, and Serpinb1a genes in the DG. These results indicate that LEV could be involved in nonclassical mechanisms involved in Ca2+ homeostasis and the regulation of the mTOR pathway through Efcab1, Hgf, SLC24a1, Adam8, and Serpinb1a, contributing to reduced hyperexcitability in TLE patients.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Piracetam , Humans , Rats , Animals , Levetiracetam/pharmacology , Levetiracetam/therapeutic use , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/genetics , Transcriptome , Piracetam/pharmacology , Piracetam/therapeutic use , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Dentate Gyrus
14.
J Neurochem ; 168(6): 1045-1059, 2024 06.
Article in English | MEDLINE | ID: mdl-38291613

ABSTRACT

Glial cells play relevant roles in neuroinflammation caused by epilepsy. Elevated hemichannel (HC) activity formed by connexins (Cxs) or pannexin1 (Panx1) largely explains brain dysfunctions commonly caused by neuroinflammation. Glia express HCs formed by Cxs 43, 30, or 26, while glia and neurons both express HCs formed by Panx1. Cx43 HCs allow for the influx of Ca2+, which promotes glial reactivity, enabling the release of the gliotransmitters that contribute to neuronal over-stimulation. Valproate (VPA), an antiseizure medication, has pleiotropic actions on neuronal molecular targets, and their action on glial cell HCs remains elusive. We used HeLa cells transfected with Cx43, Cx30, Cx26, or Panx1 to determine the effect of VPA on HC activity in the brain. VPA slightly increased HC activity under basal conditions, but significantly enhanced it in cells pre-exposed to conditions that promoted HC activity. Furthermore, VPA increased ATP release through Cx43 HCs. The increased HC activity caused by VPA was resistant to washout, being consistent with in silico studies, which predicted the binding site for VPA and Cx43, as well as for Panx1 HCs on the intracellular side, suggesting that VPA first enters through HCs, after which their activity increases.


Subject(s)
Anticonvulsants , Connexins , Valproic Acid , Valproic Acid/pharmacology , Humans , Anticonvulsants/pharmacology , Connexins/metabolism , HeLa Cells , Brain/metabolism , Brain/drug effects , Connexin 43/metabolism , Nerve Tissue Proteins/metabolism , Adenosine Triphosphate/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Animals , Epilepsy/metabolism , Epilepsy/drug therapy , Epilepsy/chemically induced
15.
Toxicon ; 237: 107538, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38030096

ABSTRACT

Epilepsy affects around 50 million people worldwide and 30% of patients have difficulty controlling the disease. The search for substances that can fill the existing gaps in the treatment of epilepsy is of great importance. Arthropod venoms are promising sources for this purpose due to the presence of small peptides that modulate the activity of ion channels and neuron receptors. The aim of this study was to investigate dinoponeratoxins from the Dinoponera quadriceps ant venom (M-PONTX-Dq3a, M-PONTX-Dq3b and M-PONTX-Dq3c) as potential anticonvulsants. We evaluated them in a seizure model induced by pentylenetetrazole (PTZ) in male swiss mice. Interestingly, intraperitoneal treatment with each peptide increased the time until the first seizure and the percentage of survival, with M-PONTX-Dq3b showing the best results. M-PONTX-Dq3a was discarded due to the appearance of some signs of toxicity with the increase in malondialdehyde (MDA) levels in the striatum. Both, M-PONTX-Dq3b and M-PONTX-Dq3c decreased iNOS and TNF-α in the hippocampus. Notably, M-PONTX-Dq3c treatment decreased the levels of MDA and nitrite in the cortex and hippocampus. Our results indicate that, M-PONTX-Dq3b and M-PONTX-Dq3c have anticonvulsant activity and exhibit anti-inflammatory effects in epilepsy, offering new perspectives for biopharmaceutical development.


Subject(s)
Ants , Epilepsy , Humans , Mice , Animals , Male , Antimicrobial Peptides , Pentylenetetrazole/toxicity , Venoms/toxicity , Seizures/chemically induced , Seizures/drug therapy , Seizures/prevention & control , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Peptides/chemistry
16.
Neurologia (Engl Ed) ; 39(1): 1-9, 2024.
Article in English | MEDLINE | ID: mdl-38161069

ABSTRACT

INTRODUCTION: The growth hormone (GH) has been reported as a crucial neuronal survival factor in the hippocampus against insults of diverse nature. Status epilepticus (SE) is a prolonged seizure that produces extensive neuronal cell death. The goal of this study was to evaluate the effect of intracerebroventricular administration of GH on seizure severity and SE-induced hippocampal neurodegeneration. METHODOLOGY: Adult male rats were implanted with a guide cannula in the left ventricle and different amounts of GH (70, 120 or 220ng/3µl) were microinjected for 5 days; artificial cerebrospinal fluid was used as the vehicle. Seizures were induced by the lithium-pilocarpine model (3mEq/kg LiCl and 30mg/kg pilocarpine hydrochloride) one day after the last GH administration. Neuronal injury was assessed by Fluoro-Jade B (F-JB) staining. RESULTS: Rats injected with 120ng of GH did not had SE after 30mg/kg pilocarpine, they required a higher number of pilocarpine injections to develop SE than the rats pretreated with the vehicle, 70ng or 220ng GH. Prefrontal and parietal cortex EEG recordings confirmed that latency to generalized seizures and SE was also significantly higher in the 120ng group when compared with all the experimental groups. FJ-B positive cells were detected in the hippocampus after SE in all rats, and no significant differences in the number of F-JB cells in the CA1 area and the hilus was observed between experimental groups. CONCLUSION: Our results indicate that, although GH has an anticonvulsive effect in the lithium-pilocarpine model of SE, it does not exert hippocampal neuroprotection after SE.


Subject(s)
Anticonvulsants , Growth Hormone , Neuroprotective Agents , Status Epilepticus , Animals , Male , Rats , Anticonvulsants/pharmacology , Growth Hormone/pharmacology , Lithium/adverse effects , Neuroprotective Agents/pharmacology , Pilocarpine/adverse effects , Seizures/drug therapy , Status Epilepticus/drug therapy , Status Epilepticus/chemically induced
17.
J Chem Inf Model ; 63(22): 7083-7096, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37917937

ABSTRACT

Epilepsy is a neurological disorder characterized by recurrent seizures that arise from abnormal electrical activity in the brain. Voltage-gated sodium channels (NaVs), responsible for the initiation and propagation of action potentials in neurons, play a critical role in the pathogenesis of epilepsy. This study sought to discover potential anticonvulsant compounds that interact with NaVs, specifically, the brain subtype hNaV1.2. A ligand-based QSAR model and a docking model were constructed, validated, and applied in a parallel virtual screening over the DrugBank database. Montelukast, Novobiocin, and Cinnarizine were selected for in vitro testing, using the patch-clamp technique, and all of them proved to inhibit hNaV1.2 channels heterologously expressed in HEK293 cells. Two hits were evaluated in the GASH/Sal model of audiogenic seizures and demonstrated promising activity, reducing the severity of sound-induced seizures at the doses tested. The combination of ligand- and structure-based models presents a valuable approach for identifying potential NaV inhibitors. These findings may provide a basis for further research into the development of new antiseizure drugs for the treatment of epilepsy.


Subject(s)
Anticonvulsants , Epilepsy , Humans , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , HEK293 Cells , Ligands , Epilepsy/drug therapy , Seizures/drug therapy , NAV1.7 Voltage-Gated Sodium Channel
18.
Cell Mol Neurobiol ; 43(8): 4231-4244, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37742326

ABSTRACT

Status epilepticus (SE) is described as continuous and self-sustaining seizures, which triggers hippocampal neurodegeneration, inflammation, and gliosis. N-formyl peptide receptor (FPR) has been associated with inflammatory process. N-formyl-methionyl-leucyl-phenylalanine (fMLP) peptide plays an anti-inflammatory role, mediated by the activation of G-protein-coupled FPR. Here, we evaluated the influence of fMLP peptides on the behavior of limbic seizures, memory consolidation, and hippocampal neurodegeneration process. Male Wistar rats (Rattus norvegicus) received microinjections of pilocarpine in hippocampus (H-PILO, 1.2 mg/µL, 1 µL) followed by fMLP (1 mg/mL, 1 µL) or vehicle (VEH, saline 0.9%, 1 µL). During the 90 min of SE, epileptic seizures were analyzed according to the Racine's Scale. After 24 h of SE, memory impairment was assessed by the inhibitory avoidance test and the neurodegeneration process was evaluated in hippocampal areas. There was no change in latency and number of wet dog shake (WDS) after administration of fMLP. However, our results showed that the intrahippocampal infusion of fMLP reduced the severity of seizures, as well as the number of limbic seizures. In addition, fMLP infusion protected memory dysfunction followed by SE. Finally, the intrahippocampal administration of fMLP attenuated the process of neurodegeneration in both hippocampi. Taken together, our data suggest a new insight into the functional role of fMLP peptides, with important implications for their potential use as a therapeutic agent for the treatment of brain disorders, such as epilepsy. Schematic drawing on the neuroprotective and anticonvulsant role of fMLP during status epilepticus. Initially, a cannula was implanted in hippocampus and pilocarpine/saline was administered into the hippocampus followed by fMLP/saline (A-C). fMLP reduced seizure severity and neuronal death in the hippocampus, as well as protecting against memory deficit (D).


Subject(s)
Epilepsy , Status Epilepticus , Rats , Male , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/therapeutic use , Pilocarpine/therapeutic use , Rats, Wistar , Status Epilepticus/drug therapy , Status Epilepticus/complications , Seizures/drug therapy , Epilepsy/drug therapy , Peptides/therapeutic use
19.
Arch Oral Biol ; 155: 105805, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37741048

ABSTRACT

OBJECTIVE: To investigate the effects of the anticonvulsant valproic acid (VPA) on salivary glands in male rat using biochemical, functional, histomorphometric, and redox state parameters. MATERIALS AND METHODS: Twenty-four male Wistar rats were randomly distributed into three groups (n = 8 per group): Control (0.9% saline solution), VPA100 (100 mg/kg), and VPA400 (400 mg/kg). After 21 consecutive days of treatment with by intragastric gavage. Pilocarpine-induced saliva was collected to determine salivary flow rate, pH, buffering capacity, and biochemical composition. Analyses of histomorphometric parameters and redox balance markers were performed on the parotid and submandibular glands. RESULTS: Salivary flow rate, pH, buffering capacity, total protein, potassium, sodium, and chloride were similar between groups. However, phosphate and calcium were reduced in VPA400, while amylase was increased in both VPA100 and VPA400. We did not detect significant differences in the areas of acini, ducts, and connective tissue in the salivary glands between the groups. There were no significant changes in the redox status of the submandibular glands. In turn, in the parotid glands we detected reduced total oxidizing capacity and lipid peroxidation, measured as thiobarbituric acid reactive substances (TBARs) and higher uric acid concentration in both the VPA100 and VPA400 groups, and increased superoxide dismutase (SOD) in the VPA400 group. CONCLUSION: Chronic treatment with VPA modified the salivary biochemical composition and caused disruption in the redox state of the parotid gland in rats.


Subject(s)
Anticonvulsants , Valproic Acid , Rats , Male , Animals , Anticonvulsants/pharmacology , Valproic Acid/pharmacology , Valproic Acid/analysis , Valproic Acid/metabolism , Rats, Wistar , Salivary Glands/metabolism , Saliva/chemistry , Parotid Gland/metabolism , Submandibular Gland/metabolism , Oxidation-Reduction
20.
PLoS One ; 18(6): e0287681, 2023.
Article in English | MEDLINE | ID: mdl-37390086

ABSTRACT

The Clibadium spp. is a shrub of occurrence in the Amazon, popularly known as Cunambi. The compounds in the leaves demonstrate ichthyotoxic properties, and its major substance, cunaniol, is a powerful central nervous system stimulant with proconvulsant activity. Few current studies relate behavioral changes to the electrophysiological profile of fish poisoning. This study aimed to describe the behavioral, electromyographic, electroencephalographic, electrocardiographic, and seizure control characteristics of anticonvulsant drugs in Colossoma macropomum submitted to cunaniol intoxication during bathing containing 0.3 µg/L cunaniol. The behavioral test showed rapid evolution presenting excitability and spasms, which were confirmed by the analysis of Electroencephalogram (EEG), Electromyogram (EMG), and changes in cardiac function detected in the ECG. Cunaniol-induced excitability control was evaluated using three anticonvulsant agents: Phenytoin, Phenobarbital, and Diazepam. While phenytoin was not effective in seizure control, diazepam proved to be the most efficient. These results demonstrate the susceptibility of Colossoma macropomum to cunaniol poisoning, given that the central nervous system and electrocardiographic changes were considered severe.


Subject(s)
Characiformes , Electrocorticography , Animals , Electroencephalography , Anticonvulsants/pharmacology , Diazepam
SELECTION OF CITATIONS
SEARCH DETAIL