1
|
Davletshin AI, Matveeva AA, Poletaeva II, Evgen'ev MB, Garbuz DG. The role of molecular chaperones in the mechanisms of epileptogenesis. Cell Stress Chaperones 2023; 28:599-619. [PMID: 37755620 PMCID: PMC10746656 DOI: 10.1007/s12192-023-01378-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Epilepsy is a group of neurological diseases which requires significant economic costs for the treatment and care of patients. The central point of epileptogenesis stems from the failure of synaptic signal transmission mechanisms, leading to excessive synchronous excitation of neurons and characteristic epileptic electroencephalogram activity, in typical cases being manifested as seizures and loss of consciousness. The causes of epilepsy are extremely diverse, which is one of the reasons for the complexity of selecting a treatment regimen for each individual case and the high frequency of pharmacoresistant cases. Therefore, the search for new drugs and methods of epilepsy treatment requires an advanced study of the molecular mechanisms of epileptogenesis. In this regard, the investigation of molecular chaperones as potential mediators of epileptogenesis seems promising because the chaperones are involved in the processing and regulation of the activity of many key proteins directly responsible for the generation of abnormal neuronal excitation in epilepsy. In this review, we try to systematize current data on the role of molecular chaperones in epileptogenesis and discuss the prospects for the use of chemical modulators of various chaperone groups' activity as promising antiepileptic drugs.
Collapse
Affiliation(s)
| | - Anna A Matveeva
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
- Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Moscow Region, Russia
| | - Inga I Poletaeva
- Biology Department, Lomonosov Moscow State University, 119991, Moscow, Russia
| | | | - David G Garbuz
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
| |
Collapse
|
2
|
Javaid S, Alqahtani F, Ashraf W, Anjum SMM, Rasool MF, Ahmad T, Alasmari F, Alasmari AF, Alqarni SA, Imran I. Tiagabine suppresses pentylenetetrazole-induced seizures in mice and improves behavioral and cognitive parameters by modulating BDNF/TrkB expression and neuroinflammatory markers. Biomed Pharmacother 2023; 160:114406. [PMID: 36791567 DOI: 10.1016/j.biopha.2023.114406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Tiagabine (Tia), a new-generation antiseizure drug that mimics the GABAergic signaling by inhibiting GABA transporter type-1, is the least studied molecule in chronic epilepsy models with comorbid neurobehavioral and neuroinflammatory parameters. Therefore, the current study investigated the effects of Tia in a real-time manner on electroencephalographic (EEG) activity, behavioral manifestations and mRNA expression in pentylenetetrazole (PTZ)-kindled mice. Male BALB/c mice were treated with tiagabine (0.5, 1 and 2 mg/kg) for 21 days with simultaneous PTZ (40 mg/kg) injection every other day for a total of 11 injections and monitored for seizure progression with synchronized validation through EEG recordings from cortical electrodes. The post-kindling protection from anxiety and memory deficit was verified by a battery of behavioral experiments. Isolated brains were evaluated for oxidative alterations and real-time changes in mRNA expression for BDNF/TrkB, GAT-1 and GAT-3 as well as neuroinflammatory markers. Experimental results revealed that Tia at the dose of 2 mg/kg maximally inhibited the development of full bloom seizure and reduced epileptic spike discharges from the cortex. Furthermore, Tia dose-dependently exerted the anxiolytic effects and protected from PTZ-evoked cognitive impairment. Tia reduced lipid peroxidation and increased superoxide dismutase and glutathione levels in the brain via augmentation of GABAergic modulation. PTZ-induced upregulated BDNF/TrkB signaling and pro-inflammatory cytokines were mitigated by Tia with upregulation of GAT-1 and GAT-3 transporters in whole brains. In conclusion, the observed effects of Tia might have resulted from reduced oxidative stress, BDNF/TrkB modulation and mitigated neuroinflammatory markers expression leading to reduced epileptogenesis and improved epilepsy-related neuropsychiatric effects.
Collapse
Affiliation(s)
- Sana Javaid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan; Department of Pharmacy, The Women University, Multan 60000, Pakistan
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Waseem Ashraf
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Syed Muhammad Muneeb Anjum
- The Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore 75270, Pakistan
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Tanveer Ahmad
- Institut pour l'Avancée des Biosciences, Centre de Recherche UGA / INSERM U1209 / CNRS 5309, Université Grenoble Alpes, France
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh Abdullah Alqarni
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan.
| |
Collapse
|
3
|
Rodent Models of Audiogenic Epilepsy: Genetic Aspects, Advantages, Current Problems and Perspectives. Biomedicines 2022; 10:biomedicines10112934. [PMID: 36428502 PMCID: PMC9687921 DOI: 10.3390/biomedicines10112934] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Animal models of epilepsy are of great importance in epileptology. They are used to study the mechanisms of epileptogenesis, and search for new genes and regulatory pathways involved in the development of epilepsy as well as screening new antiepileptic drugs. Today, many methods of modeling epilepsy in animals are used, including electroconvulsive, pharmacological in intact animals, and genetic, with the predisposition for spontaneous or refractory epileptic seizures. Due to the simplicity of manipulation and universality, genetic models of audiogenic epilepsy in rodents stand out among this diversity. We tried to combine data on the genetics of audiogenic epilepsy in rodents, the relevance of various models of audiogenic epilepsy to certain epileptic syndromes in humans, and the advantages of using of rodent strains predisposed to audiogenic epilepsy in current epileptology.
Collapse
|
4
|
Bresnahan R, Martin‐McGill KJ, Hutton JL, Marson AG. Tiagabine add-on therapy for drug-resistant focal epilepsy. Cochrane Database Syst Rev 2019; 10:CD001908. [PMID: 31608990 PMCID: PMC6953346 DOI: 10.1002/14651858.cd001908.pub4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Epilepsy is a common neurological condition that affects up to 1% of the population. Nearly 30% of people with epilepsy are resistant to currently available antiepileptic drugs (AEDs) and require treatment with multiple antiepileptic drugs in combination. Tiagabine is one of the newer AEDs that can be used as an adjunct (add-on) to standard AEDs. OBJECTIVES To evaluate the efficacy and tolerability of tiagabine when used as an add-on treatment for people with drug-resistant focal seizures. SEARCH METHODS This is an updated Cochrane review, last published in 2014. For the latest update, we searched the following databases on 22 January 2019: Cochrane Register of Studies (CRS Web), which includes the Cochrane Epilepsy Group's Specialized Register and the Cochrane Central Register of Controlled Trials, MEDLINE (Ovid, 1946 to January 21, 2019), ClinicalTrials.gov, and the WHO International Clinical Trials Registry Platform. We imposed no language restrictions. We also contacted the manufacturers of tiagabine and experts in the field to identify any ongoing or unpublished studies. SELECTION CRITERIA We included randomised placebo-controlled add-on trials conducted in people of any age with focal epilepsy. The studies could be double-, single-, or unblinded and of parallel or cross-over design. They had to have a minimum treatment period of eight weeks. We also included trials using an active drug control group. DATA COLLECTION AND ANALYSIS Two review authors independently assessed trials for inclusion and extracted data according to the standard methodological procedures expected by the Cochrane Collaboration for this review update. We resolved disagreements by discussion. Outcomes investigated included 50% or greater reduction in seizure frequency, treatment withdrawal, adverse effects, effects on cognition and quality of life. The primary analyses were performed by intention-to-treat. We calculated worst-case and best-case analyses for seizure outcomes. We evaluated dose response using regression models. Two review authors assessed risk of bias in each study using the Cochrane 'Risk of bias' tool. MAIN RESULTS No further studies were added since the previous update in 2014. The review included six trials (four parallel-group and two cross-over group trials) consisting of 948 participants. For the main comparison, tiagabine versus placebo, all participants were aged between 12 and 77 years and the study treatment periods ranged from 12 to 22 weeks. The overall risk ratio (RR) with 95% confidence intervals (CIs) for a 50% or greater reduction in seizure frequency (tiagabine versus placebo) was 3.16 (95% CI 1.97 to 5.07; 3 trials; 769 participants; high-certainty evidence). Because of differences in response rates among trials, regression models were unable to provide reliable estimates of response to individual doses. The RR for treatment withdrawal (tiagabine versus placebo) was 1.81 (95% CI 1.25 to 2.62; 3 trials, 769 participants; moderate-certainty evidence). Dizziness and tremor were significantly associated with tiagabine therapy. For cognitive and quality-of-life outcomes, the limited available data suggested no significant effects on cognition, mood, or adjustment. One trial comparing tiagabine with an active drug control group (tiagabine versus topiramate) found no significant differences between the two add-on drugs for a 50% or greater reduction in seizure frequency (RR 0.54, 95% CI 0.19 to 1.58; 1 trial; 41 participants) or for treatment withdrawal (RR 1.43, 95% CI 0.74 to 2.74; one trial; 41 participants). We judged two of the six included studies to have low risk of bias, three studies to have an unclear risk of bias, and one study to have a high risk of bias. Methods for randomisation sequence generation were the least reported trial design factor and generated the most concerns regarding risk of bias. We rated the overall certainty of the evidence as largely moderate to high using the GRADE approach. We rated the evidence for two of the adverse effect outcomes, nausea and tremor, as low certainty. AUTHORS' CONCLUSIONS Tiagabine reduced seizure frequency but was associated with some adverse effects when used as an add-on treatment in people with drug-resistant focal epilepsy. The findings of the current review are mainly applicable to adults and adolescents, and may not necessarily be applicable to children as none of the trials included participants aged under 12 years. We found no significant differences between tiagabine and topiramate as add-on drugs; however, evidence was provided by a single trial and was therefore limited.
Collapse
Affiliation(s)
- Rebecca Bresnahan
- Institute of Translational Medicine, University of LiverpoolDepartment of Molecular and Clinical PharmacologyLower LaneLiverpoolUKL9 7LJ
| | - Kirsty J Martin‐McGill
- Institute of Translational Medicine, University of LiverpoolDepartment of Molecular and Clinical PharmacologyLower LaneLiverpoolUKL9 7LJ
- University of ChesterDepartment of Clinical Sciences and NutritionFaculty of Medicine, Dentistry and Life SciencesChesterUK
| | - Jane L Hutton
- University of WarwickDepartment of StatisticsCoventryUKCV4 7AL
| | - Anthony G Marson
- Institute of Translational Medicine, University of LiverpoolDepartment of Molecular and Clinical PharmacologyLower LaneLiverpoolUKL9 7LJ
- The Walton Centre NHS Foundation TrustLiverpoolUK
- Liverpool Health PartnersLiverpoolUK
| | | |
Collapse
|
5
|
Valdés-Cruz A, Villasana-Salazar B, Williams B, Martínez-Vargas D, Magdaleno-Madrigal VM, Almazán-Alvarado S, Besio WG. Transcranial focal electrical stimulation via concentric ring electrodes in freely moving cats: Antiepileptogenic and postictal effects. Exp Neurol 2019; 320:113012. [DOI: 10.1016/j.expneurol.2019.113012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/26/2019] [Accepted: 07/09/2019] [Indexed: 01/13/2023]
|
6
|
Metcalf CS, Huff J, Thomson KE, Johnson K, Edwards SF, Wilcox KS. Evaluation of antiseizure drug efficacy and tolerability in the rat lamotrigine-resistant amygdala kindling model. Epilepsia Open 2019; 4:452-463. [PMID: 31440726 PMCID: PMC6698678 DOI: 10.1002/epi4.12354] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/09/2019] [Accepted: 07/21/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE The lamotrigine-resistant amygdala kindling model uses repeated administration of a low dose of lamotrigine during the kindling process to produce resistance to lamotrigine, which also extends to some other antiseizure drugs (ASDs). This model of pharmacoresistant epilepsy has been incorporated into the testing scheme utilized by the Epilepsy Therapy Screening Program (ETSP). Although some ASDs have been evaluated in this model, a comprehensive evaluation of ASD prototypes has not been reported. METHODS Following depth electrode implantation and recovery, rats were exposed to lamotrigine (5 mg/kg, i.p.) prior to each stimulation during the kindling development process (~3 weeks). A test dose of lamotrigine was used to confirm that fully kindled rats were lamotrigine-resistant. Efficacy (unambiguous protection against electrically elicited convulsive seizures) was defined as a Racine score < 3 in the absence of overt compound-induced side effects. Various ASDs, comprising several mechanistic classes, were administered to fully kindled, lamotrigine-resistant rats. Where possible, multiple doses of each drug were administered in order to obtain median effective dose (ED50) values. RESULTS Five sodium channel blockers tested (eslicarbazepine, lacosamide, lamotrigine, phenytoin, and rufinamide) were either not efficacious or effective only at doses that were not well-tolerated in this model. In contrast, compounds targeting either GABA receptors (clobazam, clonazepam, phenobarbital) or GABA-uptake proteins (tiagabine) produced dose-dependent efficacy against convulsive seizures. Compounds acting to modulate Ca2+ channels show differential activity: Ethosuximide was not effective, whereas gabapentin was moderately efficacious. Ezogabine and valproate were also highly effective, whereas topiramate and levetiracetam were not effective at the doses tested. SIGNIFICANCE These results strengthen the conclusion that the lamotrigine-resistant amygdala kindling model demonstrates pharmacoresistance to certain ASDs, including, but not limited to, sodium channel blockers, and supports the utility of the model for helping to identify compounds with potential efficacy against pharmacoresistant seizures.
Collapse
Affiliation(s)
- Cameron S. Metcalf
- Anticonvulsant Drug Development Program, Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUTUSA
| | - Jennifer Huff
- Anticonvulsant Drug Development Program, Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUTUSA
| | - Kyle E. Thomson
- Anticonvulsant Drug Development Program, Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUTUSA
| | - Kristina Johnson
- Anticonvulsant Drug Development Program, Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUTUSA
| | - Sharon F. Edwards
- Anticonvulsant Drug Development Program, Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUTUSA
| | - Karen S. Wilcox
- Anticonvulsant Drug Development Program, Department of Pharmacology and ToxicologyUniversity of UtahSalt Lake CityUTUSA
| |
Collapse
|
7
|
Koene LMC, van Grondelle SE, Proietti Onori M, Wallaard I, Kooijman NHRM, van Oort A, Schreiber J, Elgersma Y. Effects of antiepileptic drugs in a new TSC/mTOR-dependent epilepsy mouse model. Ann Clin Transl Neurol 2019; 6:1273-1291. [PMID: 31353861 PMCID: PMC6649373 DOI: 10.1002/acn3.50829] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE An epilepsy mouse model for Tuberous Sclerosis Complex (TSC) was developed and validated to investigate the mechanisms underlying epileptogenesis. Furthermore, the possible antiepileptogenic properties of commonly used antiepileptic drugs (AEDs) and new compounds were assessed. METHODS Tsc1 deletion was induced in CAMK2A-expressing neurons of adult mice. The antiepileptogenic properties of commonly used AEDs and inhibitors of the mTOR pathways were assessed by EEG recordings and by molecular read outs. RESULTS Mice developed epilepsy in a narrow time window (10 ± 2 days) upon Tsc1 gene deletion. Seizure frequency but not duration increased over time. Seizures were lethal within 18 days, were unpredictable, and did not correlate to seizure onset, length or frequency, reminiscent of sudden unexpected death in epilepsy (SUDEP). Tsc1 gene deletion resulted in a strong activation of the mTORC1 pathway, and both epileptogenesis and lethality could be entirely prevented by RHEB1 gene deletion or rapamycin treatment. However, other inhibitors of the mTOR pathway such as AZD8055 and PF4708671 were ineffective. Except for ketogenic diet, none of commonly used AEDs showed an effect on mTORC1 activity. Vigabatrin and ketogenic diet treatment were able to significantly delay seizure onset. In contrast, survival was shortened by lamotrigine. INTERPRETATION This novel Tsc1 mouse model is highly suitable to assess the efficacy of antiepileptic and -epileptogenic drugs to treat mTORC1-dependent epilepsy. Additionally, it allows us to study the mechanisms underlying mTORC1-mediated epileptogenesis and SUDEP. We found that early treatment with vigabatrin was not able to prevent epilepsy, but significantly delayed seizure onset.
Collapse
Affiliation(s)
- Linda M. C. Koene
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| | - Saskia E. van Grondelle
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| | - Martina Proietti Onori
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| | - Ilse Wallaard
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| | - Nathalie H. R. M. Kooijman
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| | - Annabel van Oort
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| | - Jadwiga Schreiber
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| | - Ype Elgersma
- Department of Neuroscience and ENCORE Expertise Center for Neurodevelopmental DisordersErasmus MC University Medical CenterRotterdam3015 CNThe Netherlands
| |
Collapse
|
8
|
Liberato JL, Godoy LD, Cunha AOS, Mortari MR, de Oliveira Beleboni R, Fontana ACK, Lopes NP, Dos Santos WF. Parawixin2 Protects Hippocampal Cells in Experimental Temporal Lobe Epilepsy. Toxins (Basel) 2018; 10:toxins10120486. [PMID: 30469496 PMCID: PMC6316435 DOI: 10.3390/toxins10120486] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/27/2018] [Accepted: 11/13/2018] [Indexed: 12/20/2022] Open
Abstract
Epilepsy is considered as one of the major disabling neuropathologies. Almost one third of adult patients with temporal lobe epilepsy (TLE) do not respond to current antiepileptic drugs (AEDs). Additionally, most AEDs do not have neuroprotective effects against the inherent neurodegenerative process underlying the hippocampal sclerosis on TLE. Dysfunctions in the GABAergic neurotransmission may contribute not only to the onset of epileptic activity but also constitute an important system for therapeutic approaches. Therefore, molecules that enhance GABA inhibitory effects could open novel avenues for the understanding of epileptic plasticity and for drug development. Parawixin2, a compound isolated from Parawixia bistriata spider venom, inhibits both GABA and glycine uptake and has an anticonvulsant effect against a wide range of chemoconvulsants. The neuroprotective potential of Parawixin2 was analyzed in a model of TLE induced by a long-lasting Status Epilepticus (SE), and its efficiency was compared to well-known neuroprotective drugs, such as riluzole and nipecotic acid. Neuroprotection was assessed through histological markers for cell density (Nissl), astrocytic reactivity (GFAP) and cell death labeling (TUNEL), which were performed 24 h and 72 h after SE. Parawixin2 treatment resulted in neuroprotective effects in a dose dependent manner at 24 h and 72 h after SE, as well as reduced reactive astrocytes and apoptotic cell death. Based on these findings, Parawixin2 has a great potential to be used as a tool for neuroscience research and as a probe to the development of novel GABAergic neuroprotective agents.
Collapse
Affiliation(s)
- José Luiz Liberato
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
- Neuroscience Behavioral Institute (INEC), Av. do Café, 2450, Ribeirão Preto, 14050-220 São Paulo, Brazil.
| | - Lívea Dornela Godoy
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
- Neuroscience Behavioral Institute (INEC), Av. do Café, 2450, Ribeirão Preto, 14050-220 São Paulo, Brazil.
| | - Alexandra Olimpio Siqueira Cunha
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
| | - Marcia Renata Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, DF 70910-900 Brasília, Brazil.
| | - Rene de Oliveira Beleboni
- Department of Biotechnology/School of Medicine, University of Ribeirão Preto, Av. Costábile Romano, 2201, Ribeirão Preto, 14096-900 São Paulo, Brazil.
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA 19102, USA.
| | - Norberto Peporine Lopes
- NPPNS, Department of Physics and Chemistry, College of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil, Av. do Cafe s/n, Ribeirão Preto, 14040-903 São Paulo, Brazil.
| | - Wagner Ferreira Dos Santos
- Neurobiology and Venoms Laboratory (LNP), Department of Biology, College of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, 14040-901 São Paulo, Brazil.
- Neuroscience Behavioral Institute (INEC), Av. do Café, 2450, Ribeirão Preto, 14050-220 São Paulo, Brazil.
| |
Collapse
|
9
|
Silverman RB. Design and Mechanism of GABA Aminotransferase Inactivators. Treatments for Epilepsies and Addictions. Chem Rev 2018; 118:4037-4070. [PMID: 29569907 PMCID: PMC8459698 DOI: 10.1021/acs.chemrev.8b00009] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
When the brain concentration of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) diminishes below a threshold level, the excess neuronal excitation can lead to convulsions. This imbalance in neurotransmission can be corrected by inhibition of the enzyme γ-aminobutyric acid aminotransferase (GABA-AT), which catalyzes the conversion of GABA to the excitatory neurotransmitter l-glutamic acid. It also has been found that raising GABA levels can antagonize the rapid elevation and release of dopamine in the nucleus accumbens, which is responsible for the reward response in addiction. Therefore, the design of new inhibitors of GABA-AT, which increases brain GABA levels, is an important approach to new treatments for epilepsy and addiction. This review summarizes findings over the last 40 or so years of mechanism-based inactivators (unreactive compounds that require the target enzyme to catalyze their conversion to the inactivating species, which inactivate the enzyme prior to their release) of GABA-AT with emphasis on their catalytic mechanisms of inactivation, presented according to organic chemical mechanism, with minimal pharmacology, except where important for activity in epilepsy and addiction. Patents, abstracts, and conference proceedings are not covered in this review. The inactivation mechanisms described here can be applied to the inactivations of a wide variety of unrelated enzymes.
Collapse
Affiliation(s)
- Richard B. Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, Evanston, Illinois, 60208-3113, United States
| |
Collapse
|
10
|
Godoy LD, Liberato JL, Celani MVB, Gobbo-Neto L, Lopes NP, Dos Santos WF. Disease Modifying Effects of the Spider Toxin Parawixin2 in the Experimental Epilepsy Model. Toxins (Basel) 2017; 9:toxins9090262. [PMID: 28841161 PMCID: PMC5618195 DOI: 10.3390/toxins9090262] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/27/2017] [Accepted: 08/18/2017] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Temporal lobe epilepsy (TLE) is the most common type of epilepsy in adults. It is also the one with the highest percentage of drug-resistance to the current available anti-epileptic drugs (AED). Additionaly, most antiepileptic drugs are only able to control seizures in epileptogenesis, but do not decrease the hippocampal neurodegenerative process. TLE patients have a reduced population of interneuronal cells, which express Parvalbumin (PV) proteins. This reduction is directly linked to seizure frequency and severity in the chronic period of epilepsy. There is therefore a need to seek new therapies with a disease-modifying profile, and with efficient antiepileptic and neuroprotective properties. Parawixin2, a compound isolated from the venom of the spider Parawixia bistriata, has been shown to inhibit GABA transporters (GAT) and to have acute anticonvulsant effects in rats. (2) Methods: In this work, we studied the effects of Parawixin2 and Tiagabine (an FDA- approved GAT inhibitor), and compared these effects in a TLE model. Rats were subjected to lithium-pilocarpine TLE model and the main features were evaluated over a chronic period including: (a) spontaneous recurrent seizures (SRS), (b) neuronal loss, and (c) PV cell density in different regions of the hippocampus (CA1, CA3, DG and Hilus). (3) Results: Parawixin2 treatment reduced SRS frequency whereas Tiagabine did not. We also found a significant reduction in neuronal loss in CA3 and in the hilus regions of the hippocampus, in animals treated with Parawixin2. Noteworthy, Parawixin2 significantly reversed PV cell loss observed particularly in DG layers. (4) Conclusions: Parawixin2 exerts a promising neuroprotective and anti-epileptic effect and has potential as a novel agent in drug design.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Laboratório de Neurobiologia e Peçonhas (LNP), Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes, 3900, CEP 14040-901 Ribeirão Preto, São Paulo, Brazil.
- Instituto de Neurociências e Comportamento (INEC), Av. do Café, 2450, CEP 14050-220 Ribeirão Preto, São Paulo, Brazil.
| | - José Luiz Liberato
- Laboratório de Neurobiologia e Peçonhas (LNP), Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes, 3900, CEP 14040-901 Ribeirão Preto, São Paulo, Brazil.
- Instituto de Neurociências e Comportamento (INEC), Av. do Café, 2450, CEP 14050-220 Ribeirão Preto, São Paulo, Brazil.
| | - Marcus Vinícius Batista Celani
- Laboratório de Neurobiologia e Peçonhas (LNP), Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes, 3900, CEP 14040-901 Ribeirão Preto, São Paulo, Brazil.
| | - Leonardo Gobbo-Neto
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos (NPPNS), Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Cafe s/n, CEP 14040-903 Ribeirão Preto, São Paulo, Brazil.
| | - Norberto Peporine Lopes
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos (NPPNS), Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Cafe s/n, CEP 14040-903 Ribeirão Preto, São Paulo, Brazil.
| | - Wagner Ferreira Dos Santos
- Laboratório de Neurobiologia e Peçonhas (LNP), Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes, 3900, CEP 14040-901 Ribeirão Preto, São Paulo, Brazil.
- Instituto de Neurociências e Comportamento (INEC), Av. do Café, 2450, CEP 14050-220 Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
11
|
Eskandari S, Willford SL, Anderson CM. Revised Ion/Substrate Coupling Stoichiometry of GABA Transporters. ADVANCES IN NEUROBIOLOGY 2017; 16:85-116. [PMID: 28828607 DOI: 10.1007/978-3-319-55769-4_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The purpose of this review is to highlight recent evidence in support of a 3 Na+: 1 Cl-: 1 GABA coupling stoichiometry for plasma membrane GABA transporters (SLC6A1 , SLC6A11 , SLC6A12 , SLC6A13 ) and how the revised stoichiometry impacts our understanding of the contribution of GABA transporters to GABA homeostasis in synaptic and extrasynaptic regions in the brain under physiological and pathophysiological states. Recently, our laboratory probed the GABA transporter stoichiometry by analyzing the results of six independent measurements, which included the shifts in the thermodynamic transporter reversal potential caused by changes in the extracellular Na+, Cl-, and GABA concentrations, as well as the ratio of charge flux to substrate flux for Na+, Cl-, and GABA under voltage-clamp conditions. The shifts in the transporter reversal potential for a tenfold change in the external concentration of Na+, Cl-, and GABA were 84 ± 4, 30 ± 1, and 29 ± 1 mV, respectively. Charge flux to substrate flux ratios were 0.7 ± 0.1 charges/Na+, 2.0 ± 0.2 charges/Cl-, and 2.1 ± 0.1 charges/GABA. We then compared these experimental results with the predictions of 150 different transporter stoichiometry models, which included 1-5 Na+, 0-5 Cl-, and 1-5 GABA per transport cycle. Only the 3 Na+: 1 Cl-: 1 GABA stoichiometry model correctly predicts the results of all six experimental measurements. Using the revised 3 Na+: 1 Cl-: 1 GABA stoichiometry, we propose that the GABA transporters mediate GABA uptake under most physiological conditions. Transporter-mediated GABA release likely takes place under pathophysiological or extreme physiological conditions.
Collapse
Affiliation(s)
- Sepehr Eskandari
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA.
| | - Samantha L Willford
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA
| | - Cynthia M Anderson
- Biological Sciences Department, California State Polytechnic University, Pomona, CA, 91768, USA
| |
Collapse
|
12
|
Mula M. The Clinical Spectrum of Bipolar Symptoms in Epilepsy: A Critical Reappraisal. Postgrad Med 2015; 122:17-23. [DOI: 10.3810/pgm.2010.07.2171] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
13
|
Hanada T, Ido K, Kosasa T. Effect of perampanel, a novel AMPA antagonist, on benzodiazepine-resistant status epilepticus in a lithium-pilocarpine rat model. Pharmacol Res Perspect 2014; 2:e00063. [PMID: 25505607 PMCID: PMC4186423 DOI: 10.1002/prp2.63] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/17/2014] [Accepted: 06/24/2014] [Indexed: 12/15/2022] Open
Abstract
This study assessed the efficacy of diazepam, and the alpha-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor antagonists perampanel and GYKI52466 in a lithium-pilocarpine status epilepticus (SE) model. SE was induced in rats using lithium chloride, scopolamine methyl bromide, and pilocarpine. Diazepam 10, 20, or 40 mg kg−1, or perampanel 1, 2.5, 5, or 8 mg kg−1 were administered intravenously at 10 or 30 min after seizure onset, and GYKI52466 50 mg kg−1, or combinations of diazepam 2.5–5 mg kg−1 and perampanel 0.5–1 mg kg−1, were administered intravenously at 30 min after seizure onset. Diazepam 20 mg kg−1 terminated seizures (based on electroencephalography and assessment of behavioral seizures) in 2/6 rats at 10 min and 0/6 rats at 30 min (ED50: 10 min, 30 mg kg−1; 30 min, not determined). Perampanel 8 mg kg−1 terminated seizures in 6/6 rats at both 10 and 30 min (ED50: 10 min 1.7 mg kg−1; 30 min, 5.1 mg kg−1). GYKI52466 50 mg kg−1 terminated seizures in 2/4 rats at 30 min. Co-administration of diazepam 5 mg kg−1 and perampanel 1 mg kg−1 terminated seizures in 9/9 rats at 30 min. In conclusion, perampanel and GYKI52466 provided efficacy in a lithium-pilocarpine SE model at 30 min after seizure onset, when SE was refractory to diazepam, supporting the therapeutic potential of AMPA receptor antagonists for refractory SE. The perampanel dose required to terminate seizures was reduced by combination with diazepam, suggesting synergy.
Collapse
Affiliation(s)
- Takahisa Hanada
- Global Biopharmacology, Neuroscience & General Medicine Product Creation System, Eisai Co., Ltd Tsukuba, Ibaraki, Japan ; Center for Tsukuba Advanced Research Alliance, Graduate School of Life and Environmental Sciences, University of Tsukuba Tsukuba, Ibaraki, Japan
| | - Katsutoshi Ido
- Global Biopharmacology, Neuroscience & General Medicine Product Creation System, Eisai Co., Ltd Tsukuba, Ibaraki, Japan
| | - Takashi Kosasa
- Global Biopharmacology, Neuroscience & General Medicine Product Creation System, Eisai Co., Ltd Tsukuba, Ibaraki, Japan
| |
Collapse
|
14
|
|
15
|
Shahrokhi A, Hassanzadeh G, Vousooghi N, Joghataei MT, Eftekhari S, Zarrindast MR. The effect of tiagabine on physical development and neurological reflexes and their relationship with the γ-aminobutyric acid switch in the rat cerebral cortex during developmental stages. Behav Pharmacol 2013; 24:561-8. [PMID: 26057770 DOI: 10.1097/fbp.0b013e328365422f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In the present study, we focused on γ-aminobutyric acid (GABA) signaling through the γ-aminobutyric acid transporter (GAT) in the developing rat cerebral cortex. Tiagabine was used as a GAT inhibitor. The offspring received injections from birth until postnatal day 21 intraperitoneally. Physical development and neurological reflexes were assessed daily. Tiagabine did not influence body weight, the onset and completion of incisor eruption, or the time to appearance of cliff avoidance. However, the onset and completion of eye opening, ear unfolding, and fur growth occurred earlier in treated pups. Further, the slanted board test and righting reflex showed accelerated development (i.e. decreased time to criterion) when compared with the control group. To determine whether the obtained effects are related to the GABA switch, we examined the protein and mRNA expression of the K(+)-Cl(-) cotransporter KCC2 using western blotting and RT-PCR, respectively. Downregulation of KCC2 mRNA and protein levels was observed when GAT was inhibited. The results may indicate a role of GAT in the neurobehavioral changes that accompany the developmental switch in GABA function.
Collapse
Affiliation(s)
- Amene Shahrokhi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
16
|
Lalonde R, Strazielle C. Brain regions and genes affecting myoclonus in animals. Neurosci Res 2012; 74:69-79. [PMID: 22824643 DOI: 10.1016/j.neures.2012.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 06/02/2012] [Accepted: 07/12/2012] [Indexed: 01/26/2023]
Abstract
Myoclonus is defined as large-amplitude rhythmic movements. Brain regions underlying myoclonic jerks include brainstem, cerebellum, and cortex. Gamma-aminobutyric acid (GABA) appears to be the main neurotransmitter involved in myoclonus, possibly interacting with biogenic amines, opiates, acetylcholine, and glycine. Myoclonic jumping is a specific subtype seen in rodents, comprising rearing and hopping continuously against a wall. Myoclonic jumping can be seen in normal mouse strains, possibly as a result of simply being put inside a cage. Like other types, it is also triggered by changes in GABA, 5HT, and dopamine neurotransmission. Implicated brain regions include hippocampus and dorsal striatum, possibly with respect to D(1) dopamine, NMDA, and δ opioid receptors. There is reason to suspect that myoclonic jumping is underreported due to insufficient observations into mouse cages.
Collapse
Affiliation(s)
- R Lalonde
- Université de Rouen, UFR des Sciences Humaines et Sociales, Laboratoire de Psychologie et Neurosciences: Intégration COgnitive du NEurone à la Société (ICONES), 76821 Mont Saint-Aignan Cedex, France.
| | | |
Collapse
|
17
|
Porter RJ, Dhir A, Macdonald RL, Rogawski MA. Mechanisms of action of antiseizure drugs. HANDBOOK OF CLINICAL NEUROLOGY 2012; 108:663-681. [PMID: 22939059 DOI: 10.1016/b978-0-444-52899-5.00021-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Roger J Porter
- Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
18
|
Faust MR, Höfner G, Pabel J, Wanner KT. Azetidine derivatives as novel γ-aminobutyric acid uptake inhibitors: Synthesis, biological evaluation, and structure–activity relationship. Eur J Med Chem 2010; 45:2453-66. [DOI: 10.1016/j.ejmech.2010.02.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 02/08/2010] [Accepted: 02/10/2010] [Indexed: 11/29/2022]
|
19
|
Fujiwara A, Watanabe Y, Takechi K, Ishikawa T, Kaida Y, Akagi M, Kamei C. The usefulness of olfactory bulb kindling as a model for evaluation of antiepileptics. Epilepsia 2010; 51:445-53. [DOI: 10.1111/j.1528-1167.2009.02378.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Watanabe Y, Kaida Y, Takechi K, Kamei C. Anticonvulsant Effect of (RS)-1-Aminoindan-1,5-dicarboxylic Acid on Pentetrazol-Induced Kindled Seizures in Mice. Biol Pharm Bull 2010; 33:647-52. [DOI: 10.1248/bpb.33.647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yusuke Watanabe
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Yuko Kaida
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Kenshi Takechi
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Chiaki Kamei
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| |
Collapse
|
21
|
Takechi K, Fujiwara A, Watanabe Y, Kamei C. Participation of GABA-ergic system in epileptogenic activity induced by teicoplanin in mice. Epilepsy Res 2009; 84:127-34. [DOI: 10.1016/j.eplepsyres.2009.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 12/25/2008] [Accepted: 01/19/2009] [Indexed: 10/21/2022]
|
22
|
Borowicz KK, Zadrozniak M, Luszczki JJ, Czuczwar SJ. Interactions between tiagabine and conventional antiepileptic drugs in the rat model of complex partial seizures. J Neural Transm (Vienna) 2008; 115:661-7. [DOI: 10.1007/s00702-007-0006-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Accepted: 12/04/2007] [Indexed: 11/27/2022]
|
23
|
Abstract
Post-traumatic stress disorder (PTSD) is a disruptive, chronic, and relatively common disorder that is often difficult to treat. Many patients with PTSD are unresponsive, have only moderate or marginal responses, or have troubling side effects to first-line serotonin reuptake inhibitor treatment. Studies suggest that antiepileptic drugs (AEDs) may be an effective treatment alternative or adjunctive treatment for the symptoms of PTSD. Recent results from case reports and open and controlled studies on the efficacy and tolerability of AEDs in PTSD are reviewed here, and their methodological limitations are discussed when relevant. AEDs shown to be effective in double-blind, placebo-controlled trials of PTSD include lamotrigine, topiramate, and tiagabine. Other AEDs that appear promising in open-label trials of PTSD include carbamazepine, valproate, gabapentin, vigabatrin, phenytoin, and levetiracetam. Stress-activated limbic kindling may be involved in the pathogenesis of PTSD. The possibility that AEDs may be effective in the treatment of PTSD due to their antikindling effect is discussed, and suggestions for future research are made.
Collapse
Affiliation(s)
- Heather A Berlin
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
24
|
Li X, Yang Q, Hu Y. Regulation of the expression of GABAA receptor subunits by an antiepileptic drug QYS. Neurosci Lett 2005; 392:145-9. [PMID: 16214289 DOI: 10.1016/j.neulet.2005.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2005] [Revised: 09/04/2005] [Accepted: 09/06/2005] [Indexed: 11/23/2022]
Abstract
It has been reported that the antiepileptic drug qingyangshenylycosides (QYS) modulated the function of GABAergic system. However, little is known about the effects of QYS on the gene expression of GABA receptors in the central nervous system (CNS). In the present study, we examined the effects of QYS on the expression of GABAA receptor subunits in different regions of the mouse brain. The results showed that treatment of QYS significantly increased the expressions of Gabra1, Gabra2 and Gabr4 and decreased the expression of Gabrg2 in inferior colliculus. Moreover, Gabrb2 expression was up-regulated and Gabra5 was down-regulated in hippocampus, while the expressions of Gabra1 and Gabrb2 were induced in cortex after QYS treatment. These data indicated that QYS had different effects on the expression of GABAA receptor subunits in different brain regions. These results may help to reveal the molecular mechanism of anticonvulsant action of QYS.
Collapse
Affiliation(s)
- Xianchun Li
- Key Lab of Brain Functional Genomics, MOE & STCSM, Shanghai Institute of Brain Functional Genomics, East China Normal University, Shanghai
| | | | | |
Collapse
|
25
|
Fueta Y, Kunugita N, Schwarz W. Antiepileptic action induced by a combination of vigabatrin and tiagabine. Neuroscience 2005; 132:335-45. [PMID: 15802187 DOI: 10.1016/j.neuroscience.2004.12.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2004] [Indexed: 11/23/2022]
Abstract
Vigabatrin, an inhibitor of GABA breakdown by GABA transaminase and of GABA transporter isoform 1 (GAT1), and tiagabine, a highly specific inhibitor of GAT1, have successfully been applied in the treatment of epilepsy. We investigated the effects of individual and combined application of these drugs on GAT1 expressed in Xenopus oocytes, and examined the effects on epileptiform discharges in the CA3 area of brain slices of genetically epileptic El and control ddY mice, and on the occurrence of seizures in El mice. Simultaneous application of vigabatrin and tiagabine inhibited epileptiform discharges induced by high-K+ solution in the brain slices in an antagonistic fashion. The degree of inhibition by tiagabine after pre-treatment with vigabatrin was additive in ddY mice and synergistic in El mice. In Mg2+-free solution, co-treatment by the two drugs produced additive inhibition in slices from both mouse strains, but pre-treatment with vigabatrin produced synergistic inhibition in slices only from ddY mice. In the slices from El mice, a combination of drugs resulted in additive effects in both co- and pre-treatment by the drugs. Although these drugs are also effective in vivo at suppressing seizure occurrence in El mice, the combined application does not show synergistic effects, but rather is antagonistic under the experimental conditions in this particular variant of epilepsy. The synergistic inhibition of epileptiform discharges in brain slices may, in part, have originated from the complex interaction with GAT1. In experiments on the GAT1 expressed in oocytes it could be demonstrated that synergistic inhibition occurs only at low concentration (0.1 nM) of vigabatrin. This illustrates that the oocytes may form a powerful test system for drug screening and investigation of complex drug interactions. These results present a novel interpretation of synergistic inhibition of certain epileptic discharges using vigabatrin and another drug, and that for successful synergistic treatment of epilepsies carefully designed timed dosage regimens are essential.
Collapse
Affiliation(s)
- Y Fueta
- Department of Med. Tech., School of Health Sciences, Univ. Occupat./Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| | | | | |
Collapse
|
26
|
Abstract
In this article, we discuss and highlight some of the potential neurochemical underpinnings of bipolar disorder (BD) and epilepsy. Some similarities are found in both disorders, such as the episodic course of the illnesses, the possible mechanism of kindling, and the efficacy of some antiepileptic drugs (AEDs) in treatment, all pointing to a common underlying pathophysiology. Common mechanisms at the level of ion channels might include the antikindling and the calcium-antagonistic and potassium outward current-modulating properties of AEDs. However, future research on intracellular mechanisms might become decisive for a better understanding of the similarities between the disorders.
Collapse
Affiliation(s)
- Benedikt Amann
- Department of Psychiatry, University LMU Munich, Munich, Germany.
| | | |
Collapse
|
27
|
Morimoto K, Fahnestock M, Racine RJ. Kindling and status epilepticus models of epilepsy: rewiring the brain. Prog Neurobiol 2004; 73:1-60. [PMID: 15193778 DOI: 10.1016/j.pneurobio.2004.03.009] [Citation(s) in RCA: 625] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Accepted: 03/24/2004] [Indexed: 01/09/2023]
Abstract
This review focuses on the remodeling of brain circuitry associated with epilepsy, particularly in excitatory glutamate and inhibitory GABA systems, including alterations in synaptic efficacy, growth of new connections, and loss of existing connections. From recent studies on the kindling and status epilepticus models, which have been used most extensively to investigate temporal lobe epilepsy, it is now clear that the brain reorganizes itself in response to excess neural activation, such as seizure activity. The contributing factors to this reorganization include activation of glutamate receptors, second messengers, immediate early genes, transcription factors, neurotrophic factors, axon guidance molecules, protein synthesis, neurogenesis, and synaptogenesis. Some of the resulting changes may, in turn, contribute to the permanent alterations in seizure susceptibility. There is increasing evidence that neurogenesis and synaptogenesis can appear not only in the mossy fiber pathway in the hippocampus but also in other limbic structures. Neuronal loss, induced by prolonged seizure activity, may also contribute to circuit restructuring, particularly in the status epilepticus model. However, it is unlikely that any one structure, plastic system, neurotrophin, or downstream effector pathway is uniquely critical for epileptogenesis. The sensitivity of neural systems to the modulation of inhibition makes a disinhibition hypothesis compelling for both the triggering stage of the epileptic response and the long-term changes that promote the epileptic state. Loss of selective types of interneurons, alteration of GABA receptor configuration, and/or decrease in dendritic inhibition could contribute to the development of spontaneous seizures.
Collapse
Affiliation(s)
- Kiyoshi Morimoto
- Department of Neuropsychiatry, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | | | | |
Collapse
|
28
|
Yamashita H, Ohno K, Inami H, Shishikura JI, Sakamoto S, Okada M, Yamaguchi T. Suppression of fully kindled seizure and retardation of kindling acquisition by YM928 in the rat kindling model of epilepsy. Eur J Pharmacol 2004; 494:147-54. [PMID: 15212968 DOI: 10.1016/j.ejphar.2004.04.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2003] [Revised: 04/22/2004] [Accepted: 04/30/2004] [Indexed: 11/26/2022]
Abstract
We investigated the effects of 2-[N-(4-chlorophenyl)-N-methylamino]-4H-pyrido[3.2-e]-1,3-thiazin-4-one (YM928), a selective alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor antagonist, in the rat kindling model of complex partial seizures. YM928 (10 and 30 mg/kg p.o.) markedly suppressed the motor seizures and afterdischarge induced by electrical stimulation of the amygdala at generalized seizure-triggering threshold intensity. YM928 (10 mg/kg p.o.) did not induce apparent abnormal behavior, but did induce sedation at a dose of 30 mg/kg p.o. YM928 (30 mg/kg p.o.) showed a similar anticonvulsant effect at twice the threshold intensity as it did at threshold intensity. Diazepam (10 mg/kg p.o.) and phenobarbital (60 mg/kg p.o.) also exerted anticonvulsant activities. Diazepam (10 mg/kg) showed a similar effect at twice the threshold as at threshold, but the anticonvulsant effect of phenobarbital (60 mg/kg p.o.) was reversed when the stimulus was doubled. When YM928 (10 mg/kg p.o.) was administered 60 min before daily stimulation of the amygdala, the development of kindling seizure was significantly retarded. These results indicate that YM928 has anticonvulsant effects and suppresses kindling acquisition without sedative effects, and may be suitable as an antiepileptic drug for the treatment of complex partial seizures in humans.
Collapse
Affiliation(s)
- Hiroshi Yamashita
- Institute for Drug Discovery Research, Yamanouchi Pharmaceutical Co., Ltd. 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan.
| | | | | | | | | | | | | |
Collapse
|
29
|
Whitlow RD, Sacher A, Loo DDF, Nelson N, Eskandari S. The anticonvulsant valproate increases the turnover rate of gamma-aminobutyric acid transporters. J Biol Chem 2003; 278:17716-26. [PMID: 12595533 DOI: 10.1074/jbc.m207582200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Valproate is an important anticonvulsant currently in clinical use for the treatment of seizures. We used electrophysiological and tracer uptake methods to examine the effect of valproate on a gamma-aminobutyric acid (GABA) transporter (mouse GAT3) expressed in Xenopus laevis oocytes. In the absence of GABA, valproate (up to 50 mm) had no noticeable effect on the steady-state electrogenic properties of mGAT3. In the presence of GABA, however, valproate enhanced the GABA-evoked steady-state inward current in a dose-dependent manner with a half-maximal concentration of 4.6 +/- 0.5 mm. Maximal enhancement of the GABA-evoked current was 275 +/- 10%. Qualitatively similar observations were obtained for human GAT1 and mouse GAT4. The valproate enhancement did not alter the Na(+) or Cl(-) dependence of the steady-state GABA-evoked currents. Uptake experiments under voltage clamp suggested that the valproate enhancement of the GABA-evoked current was matched by an enhancement in GABA uptake. Thus, despite the increase in GABA-evoked current, ion/GABA co-transport remained tightly coupled. Uptake experiments indicated that valproate is not transported by mouse GAT3 in the absence or presence of GABA. Valproate also enhanced the rate of the partial steps involved in transporter presteady-state charge movements. We propose that valproate increases the turnover rate of GABA transporters by an allosteric mechanism. The data suggest that at its therapeutic concentration, valproate may enhance the activity of neuronal and glial GABA transporters by up to 10%.
Collapse
Affiliation(s)
- Richard D Whitlow
- Biological Sciences Department, California State Polytechnic University, Pomona, California 91768-4032, USA
| | | | | | | | | |
Collapse
|
30
|
Yang Y, Li Q, Yang T, Hussain M, Shuaib A. Reduced brain infarct volume and improved neurological outcome by inhibition of the NR2B subunit of NMDA receptors by using CP101,606-27 alone and in combination with rt-PA in a thromboembolic stroke model in rats. J Neurosurg 2003; 98:397-403. [PMID: 12593629 DOI: 10.3171/jns.2003.98.2.0397] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT A novel postsynaptic antagonist of N-methyl-D-aspartate (NMDA) receptors, CP-101,606-27 may attenuate the effects of focal ischemia. In current experiments, the authors investigated its neuroprotective effect alone and in combination with recombinant tissue plasminogen activator (rt-PA) in thromboembolic focal cerebral ischemia in rats. METHODS Forty-eight male Wistar rats underwent embolization of the right middle cerebral artery to produce focal cerebral ischemia. After random division into six groups (eight rats in each group), animals received: vehicle; low-dose (LD) CP-101, 606-27, 14.4 mg/kg; high-dose (HD) CP- 101,606-27, 28.8 mg/kg; rt-PA, 10 mg/kg; low-dose combination (LDC) CP- 101,606-27, 14.4 mg/kg plus rt-PA, 10 mg/kg; or high-dose combination (HDC) CP- 101,606-27, 28.8 mg/kg plus rt-PA, 10 mg/kg) 2 hours after induction of embolic stroke. Animals were killed 48 hours after the onset of focal ischemia. Brain infarction volume, neurobehavioral outcome, poststroke seizure activity, poststroke mortality, and intracranial hemorrhage incidence were observed and evaluated. Compared with vehicle-treated animals (39.4 +/- 8.6%) 2 hours posttreatment with CP-101,606-27 or rt-PA or in combination a significant reduction in the percentage of brain infarct volume was seen (LD CP-101,606-27: 20.8 +/- 14.3%, p < 0.05; HD CP-101,606-27: 10.9 +/- 3.2%, p < 0.001; rt-PA: 21.1 +/- 7.3%, p < 0.05; LDC, 18.6 +/- 11.5%, p < 0.05; and HDC: 15.2 +/- 10.1%, p < 0.05; compared with control: 39.4 +/- 8.6%). Combination of CP-101,606-27 with rt-PA did not show a significantly enhanced neuroprotective effect. Except for the control and LDC treatment groups, neurobehavioral outcome was significantly improved 24 hours after embolic stroke in animals in all other active therapeutic groups receiving CP-101,606-27 or rt-PA or in combination. The authors also observed that treatment with HD CP-101,606-27 decreased poststroke seizure activity. CONCLUSIONS The data in this study suggested that postischemia treatment with CP-101,606-27 is neuroprotective in the current stroke model; however, the authors also note that although rt-PA may offer modest protection when used alone, combination with CP-101,606-27 did not appear to enhance its effects.
Collapse
Affiliation(s)
- Yi Yang
- Acute Stroke Program, Neurology Division, University of Alberta Hospital, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
31
|
Fueta Y, Schwarz W, Ohno K, Endo Y, Mita T. Selective suppression of hippocampal region hyperexcitability related to seizure susceptibility in epileptic El mice by the GABA-transporter inhibitor tiagabine. Brain Res 2002; 947:212-7. [PMID: 12176163 DOI: 10.1016/s0006-8993(02)02927-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
High seizure susceptibility in El mice is associated with disinhibition in the dentate gyrus (DG) and paired-pulse facilitation in the CA3 area in hippocampal slices [Brain Res. 745 (1997) 165; Brain Res. 779 (1998) 324]. A decrease in gamma-aminobutyric acid (GABA)-mediated inhibition and an increase in excitatory inputs to the major neurons seem to be the responsible mechanisms, respectively, for these phenomena. In this study, we examined the effects of tiagabine, an inhibitor of GABA transporter, on hyperexcitation in vivo and in slice preparations. Tiagabine (0.3-0.5 mg/kg) suppressed the occurrence of seizures to about 20% of controls with an ED(50) value of about 0.17 mg/kg. In addition, perfusion of hippocampal slices with tiagabine (20 microM) counteracted the paired-pulse facilitation in the CA3 region over the entire range of interpulse intervals (P<0.05, two-way ANOVA) and reduced the disinhibition in the DG measured at 10 and 20 ms during short interpulse intervals (P<0.005, paired t-test). The CA1 region in the El mice, as well as in a non-epileptic parental strain of ddY mice did not respond to the drug. However, frequency potentiation of CA3 was enhanced in both strains (P<0.05, paired t-test). Our results suggest that within the hippocampus the antiepileptic action of tiagabine is selectively suppressive for hyperexcitability of DG and CA3, which are responsible for seizure-susceptibility in El mice.
Collapse
Affiliation(s)
- Yukiko Fueta
- Department of Medical Technology, School of Health Sciences, University of Occupational and Environmental Health, Iseigaoka 1-1, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| | | | | | | | | |
Collapse
|
32
|
Stress-induced deoxycorticosterone-derived neurosteroids modulate GABA(A) receptor function and seizure susceptibility. J Neurosci 2002. [PMID: 11978855 DOI: 10.1523/jneurosci.22-09-03795.2002] [Citation(s) in RCA: 208] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Stress affects seizure susceptibility in animals and humans, but the underlying mechanisms are obscure. Here, we provide evidence that GABA(A) receptor-modulating neurosteroids derived from deoxycorticosterone (DOC) play a role in stress-related changes in seizure control. DOC, an adrenal steroid whose synthesis is enhanced during stress, undergoes sequential metabolic reduction by 5alpha-reductase and 3alpha-hydroxysteroid oxidoreductase to form 5alpha-dihydrodeoxycorticosterone (DHDOC) and allotetrahydrodeoxycorticosterone (THDOC), a GABA(A) receptor-modulating neurosteroid with anticonvulsant properties. Acute swim stress in rats significantly elevated plasma THDOC concentrations and raised the pentylenetetrazol (PTZ) seizure threshold. Small systemic doses of DOC produced comparable increases in THDOC and PTZ seizure threshold. Pretreatment with finasteride, a 5alpha-reductase inhibitor that blocks the conversion of DOC to DHDOC, reversed the antiseizure effects of stress. DOC also elevated plasma THDOC levels and protected mice against PTZ, methyl-6,7-dimethoxy-4-ethyl-beta-carboline-3-carboxylate, picrotoxin, and amygdala-kindled seizures in mice (ED50 values, 84-97 mg/kg). Finasteride reversed the antiseizure activity of DOC (ED50, 7.2 mg/kg); partial antagonism was also obtained with indomethacin (100 mg/kg), an inhibitor of 3alpha-hydroxysteroid oxidoreductase. Finasteride had no effect on seizure protection by DHDOC and THDOC, whereas indomethacin partially reversed DHDOC but not THDOC. DHDOC, like THDOC, potentiated GABA-activated Cl- currents in cultured hippocampal neurons (< or =1 microm) and directly activated GABA(A) receptor currents (> or =1 microm), compatible with a role for DHDOC in the antiseizure activity of DOC. DOC is a mediator of the physiological effects of acute stress that could contribute to stress-induced changes in seizure susceptibility through its conversion to neurosteroids with modulatory actions on GABA(A) receptors including THDOC and possibly also DHDOC.
Collapse
|
33
|
Feng HJ, Naritoku DK, Randall ME, Faingold CL. Modulation of audiogenically kindled seizures by gamma-aminobutyric acid-related mechanisms in the amygdala. Exp Neurol 2001; 172:477-81. [PMID: 11716572 DOI: 10.1006/exnr.2001.7810] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Repetitive induction of audiogenic seizures (AGSs) ("AGS kindling") results in expansion of the AGS neuronal network from the brainstem to forebrain structures. AGSs in kindled genetically epilepsy-prone rats (GEPR-9s) exhibit a significant increase in the duration of posttonic clonus (PTC). The amygdala (AMG) does not appear to be a required network component before AGS kindling, but this structure is implicated in the seizure network after AGS kindling. gamma-Aminobutyric acid (GABA) is a major neurotransmitter in AMG, and histamine receptor activation is also reported to stimulate GABA release. The present study examined the effect on audiogenically kindled seizures of focal microinjections into the AMG of GEPR-9s. AGS kindling involved induction of 14 AGSs in GEPR-9s. Bilateral microinjection of a GABA(A) agonist, muscimol (0.3 nmol/side), into the AMG significantly reduced the duration of PTC, starting 0.5 h after drug infusion, with recovery by 24 h. Microinjection of histamine (60 nmol/side) suppressed PTC at 0.5 h, with total blockade at 24 h, but the seizure pattern did not revert to that observed before kindling until 120 h. This long duration suggests that mechanisms in addition to modulation of GABA function may be involved in the effect of histamine. The wild running and tonic components of AGS were never affected by microinjection of these agents into the AMG. These findings confirm previous work suggesting that the AMG is not a required nucleus in the AGS neuronal network before kindling. However, the AMG becomes critical in expansion of the seizure network during AGS kindling, and audiogenically kindled seizures are negatively modulated by increased GABA function.
Collapse
Affiliation(s)
- H J Feng
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois 62794, USA
| | | | | | | |
Collapse
|
34
|
Stokes AH, Bernard LP, Nicklas WJ, Zeevalk GD. Attenuation of malonate toxicity in primary mesencephalic cultures using the GABA transport blocker, NO-711. J Neurosci Res 2001; 64:43-52. [PMID: 11276050 DOI: 10.1002/jnr.1052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cultured rat mesencephalic neurons were used to assess the effects of gamma-aminobutyric acid (GABA) transport blockers on toxicity caused by malonate, a reversible, competitive inhibitor of succinate dehydrogenase. Previous studies utilizing an ex vivo chick retinal preparation have shown that GABA release and cell swelling are early consequences of acute energy impairment and that GABA transport blockers attenuate this toxicity. The present results demonstrate that the nonsubstrate GABA transport blocker, NO-711 (1 nM-1 microM), dose-dependently protected cultured mesencephalic dopamine (DA) and GABA neurons from malonate-induced toxicity. Similar protection was demonstrated with nipecotic acid (1 mM) and SKF89976A (100 nM), substrate and nonsubstrate GABA transport blockers, respectively. These compounds by themselves produced no signs of toxicity, although nipecotic acid caused a long-term decrease in GABA uptake not associated with toxicity. Compounds which decrease intracellular reactive oxygen species (ROS) are protective in this model, but NO-711 did not prevent the rise in intracellular ROS induced by malonate, indicating its protective effects were downstream of ROS production. Supplementation of malonate treated cultures with the GABA(A) agonist, muscimol (10 microM), increased the toxicity toward the DA and GABA neuron populations. Antagonists at the GABA(A) and glycine receptors provided partial protection to both the GABA and DA neurons. These findings suggest that the GABA transporter, GABA(A), and/or glycine channels contribute to cell damage associated with energy impairment in this model.
Collapse
Affiliation(s)
- A H Stokes
- Department of Neurology, Robert Wood Johnson Medical School-UMDNJ, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
35
|
Wang CX, Todd KG, Yang Y, Gordon T, Shuaib A. Patency of cerebral microvessels after focal embolic stroke in the rat. J Cereb Blood Flow Metab 2001; 21:413-21. [PMID: 11323527 DOI: 10.1097/00004647-200104000-00010] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In patients with thrombotic stroke, the occluded artery often reopens over time. This results through a natural dissolution of the occluding material, and fragments of the material may move downstream to obstruct distal arteries. The current study was undertaken to investigate the patency of brain microvessels at varying time intervals after injection of a preformed clot into the right internal carotid artery of rats. Cerebral microvessels in brain sections were visualized using immunohistochemistry for fibronectin (detecting existing microvessels) and Evans blue (visualizing perfused microvessels). The percentage of patent microvessels was calculated as the number of Evans blue-positive microvessels divided by the number of fibronectin-positive microvessels. In normal control animals, results showed that 98% +/- 3% (mean +/- SD) of microvessels in the cortex and 94% +/- 14% in the striatum were patent. In the ischemic animals, immediately after clot injection, microvessels in the cortex and striatum were occluded, mainly in the territory irrigated by the middle cerebral artery. One hour after clot injection, microvessels had reopened in most of the cortex but remained occluded in some portions of the striatum, possibly as a result of downstream movement of fragments formed from the original clot. By 3 hours after clot injection, microvessels in the cortex were patent in all animals, whereas in the striatum microvessels were patent in 50% of the animals. In the other 50%, small striatal perfusion deficits persisted. At 24 hours after clot injection, microvessels were patent in both the cortex and striatum of all animals except one. These findings suggest that intracerebral clots dissolve spontaneously in a relatively short period of time, but that fragments formed from the clot may obstruct more distal blood vessels. It is likely that clot fragments lodge in arteries with lower blood flow and poor collateral perfusion, where they continue to cause ischemia for a longer duration. These results may in part explain the resistance of the striatum to neuroprotective strategies used for the treatment of focal cerebral ischemia.
Collapse
Affiliation(s)
- C X Wang
- Department of Neurology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
36
|
Peña F, Tapia R. Seizures and neurodegeneration induced by 4-aminopyridine in rat hippocampus in vivo: role of glutamate- and GABA-mediated neurotransmission and of ion channels. Neuroscience 2001; 101:547-61. [PMID: 11113304 DOI: 10.1016/s0306-4522(00)00400-0] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Infusion of the K(+) channel blocker 4-aminopyridine in the hippocampus induces the release of glutamate, as well as seizures and neurodegeneration. Since an imbalance between excitation and inhibition, as well as alterations of ion channels, may be involved in these effects of 4-aminopyridine, we have studied whether they are modified by drugs that block glutamatergic transmission or ion channels, or drugs that potentiate GABA-mediated transmission. The drugs were administered to anesthetized rats subjected to intrahippocampal infusion of 4-aminopyridine through microdialysis probes, with simultaneous collection of dialysis perfusates and recording of the electroencephalogram, and subsequent histological analysis. Ionotropic glutamate receptor antagonists clearly diminished the intensity of seizures and prevented the neuronal damage, but did not alter substantially the enhancement of extracellular glutamate induced by 4-aminopyridine. None of the drugs facilitating GABA-mediated transmission, including uptake blockers, GABA-transaminase inhibitors and agonists of the A-type receptor, was able to reduce the glutamate release, seizures or neuronal damage produced by 4-aminopyridine. In contrast, nipecotate, which notably increased extracellular levels of the amino acid, potentiated the intensity of seizures and the neurodegeneration. GABA(A) receptor antagonists partially reduced the extracellular accumulation of glutamate induced by 4-aminopyridine, but did not exert any protective action. Tetrodotoxin largely prevented the increase of extracellular glutamate, the electroencephalographic epileptic discharges and the neuronal death in the CA1 and CA3 hippocampal regions. Valproate and carbamazepine, also Na(+) channel blockers that possess general anticonvulsant action, failed to modify the three effects of 4-aminopyridine studied. The N-type Ca(2+) channel blocker omega-conotoxin, the K(+) channel opener diazoxide, and the non-specific ion channel blocker riluzole diminished the enhancement of extracellular glutamate and slightly protected against the neurodegeneration. However, the two former compounds did not antagonize the 4-aminopyridine-induced epileptiform discharges, and riluzole instead markedly increased the intensity and duration of the disharges. Moreover, at the highest dose tested (8mg/kg, i.p.), riluzole caused a 75% mortality of the rats. We conclude that 4-aminopyridine stimulates the release of glutamate from nerve endings and that the resultant augmented extracellular glutamate is directly related to the neurodegeneration and is involved in the generation of epileptiform discharges through the concomitant overactivation of glutamate receptors. Under these conditions, a facilitated GABA-mediated transmission may paradoxically boost neuronal hyperexcitation. Riluzole, a drug used to treat amyotrophic lateral sclerosis, seems to be toxic when combined with neuronal hyperexcitation.
Collapse
Affiliation(s)
- F Peña
- Departamento de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, AP 70-253, 04510, D.F., Mexico City, Mexico
| | | |
Collapse
|
37
|
Ma Y, Hu JH, Zhao WJ, Fei J, Yu Y, Zhou XG, Mei ZT, Guo LH. Overexpression of gamma-aminobutyric acid transporter subtype I leads to susceptibility to kainic acid-induced seizure in transgenic mice. Cell Res 2001; 11:61-7. [PMID: 11305326 DOI: 10.1038/sj.cr.7290067] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is the principal inhibitory neurotransmitter, and the GABAergic synaptic transmission is normally terminated by the rapid uptake through GABA transporters. With transgenic mice ubiquitously overexpressing GABA transporter subtype I (GAT1), the present study explored the pathophysiological role of GAT1 in epileptogenesis. Though displaying no spontaneous seizure activity, these mice exhibit altered electroencephalographic patterns and increased susceptibility to seizure induced by kainic acid. In addition, the GABA(A) receptor and glutamate transporters are up-regulated in transgenic mice, which perhaps reflects a compensatory or corrective change to the elevated level of GAT1. These preliminary findings support the hypothesis that excitatory and inhibitory neurotransmission, and seizure susceptibility can be altered by neurotransmitter transporters.
Collapse
Affiliation(s)
- Y Ma
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Tiagabine (TGB), a recently approved anti-epileptic drug (AED), has a specific and unique mechanism of action involving the inhibition of gamma-aminobutyric acid (GABA) re-uptake into neurones and glia. TGB is potent and has linear and predictable pharmacokinetics. It does not induce or inhibit hepatic metabolism and has no clinically significant effects on the serum concentrations of other AEDs or commonly used non-AEDs. Double-blind, placebo-controlled studies in primarily hepatic enzyme-induced patients showed that TGB 30 - 56 mg/day is an effective add-on treatment for all subtypes of partial seizures. The most common adverse effects in the trials were dizziness, asthenia (weakness), somnolence, accidental injury, infection, headache, nausea and nervousness. These side effects were usually mild to moderate in severity and generally did not require medical intervention. Long-term safety studies show continued efficacy of TGB over time and no evidence of tolerance for efficacy. Open studies confirm that patients with medically refractory partial epilepsy can be successfully converted to TGB monotherapy and that TGB may be effective for other seizure types, such as infantile spasms.
Collapse
Affiliation(s)
- S C Schachter
- Office of Clinical Trials and Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
39
|
Chen Xu W, Yi Y, Qiu L, Shuaib A. Neuroprotective activity of tiagabine in a focal embolic model of cerebral ischemia. Brain Res 2000; 874:75-7. [PMID: 10936225 DOI: 10.1016/s0006-8993(00)02554-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gamma aminobutyric acid (GABA) agonists have been shown to have neuroprotective effects when used after focal or global cerebral ischemia. In this study, we evaluated the neuroprotective effects of a GABA re-uptake inhibitory agent, tiagabine, on focal ischemic brain injury in an embolic model in rats. Tiagabine, injected at 1 h after embolization, significantly reduced brain infarction volume, measured with 2,3,5-triphenyltetrazolium chloride (TTC) histological assay. There were varying degrees of neuroprotective effect exhibited in the other experimental groups however this did not reach significance. These results suggest that tiagabine is neuroprotective when administrated at an early period after the ischemic brain injury.
Collapse
Affiliation(s)
- W Chen Xu
- The Department of Medicine, Division of Neurology, 2E3.13 WMHSC, University of Alberta Hospital, T6G 2B7, Edmonton, Canada
| | | | | | | |
Collapse
|
40
|
Abstract
Gabitril (tiagabine) is a potent selective inhibitor of the principal neuronal gamma-aminobutyric acid (GABA) transporter (GAT-1) in the cortex and hippocampus. By slowing the reuptake of synaptically-released GABA, it prolongs inhibitory postsynaptic potentials. In animal models of epilepsy, tiagabine is particularly effective against kindled (limbic) seizures and against reflexly-induced generalized convulsive seizures. These data are predictive of its efficacy in complex partial seizures in humans. Possible clinical applications outside the field of epilepsy include bipolar disorder and pain.
Collapse
Affiliation(s)
- B S Meldrum
- Department of Clinical Neurosciences, Institute of Psychiatry, Kings College, London, United Kingdom
| | | |
Collapse
|
41
|
Abstract
Tiagabine (TGB) is a recently approved antiepileptic drug (AED) that inhibits y-aminobutyric acid (GABA) reuptake into neurons and glia, a mechanism of action that is specific and unique among the AEDs. TGB is potent and has linear and predictable pharmacokinetics. It has no clinically relevant effects on hepatic metabolism or serum concentrations of other AEDs, effects on laboratory values, or interactions with common non-AEDs. TGB is effective as add-on therapy for partial seizures in patients with medically refractory epilepsy in doses ranging from 30 to 56 mg daily. Conversion to TGB monotherapy can be achieved in patients with medically refractory epilepsy, although additional controlled studies are needed to confirm the efficacy of TGB as monotherapy and to establish the effective dosage range. In controlled studies, the most common adverse events of TGB are dizziness, asthenia, somnolence, accidental injury, infection, headache, nausea, and nervousness. These are usually mild to moderate in severity and almost always resolve without medical intervention.
Collapse
Affiliation(s)
- S C Schachter
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
42
|
Lee YS, Ha JH, Yong CS, Lee DU, Huh K, Kang YS, Lee SH, Jung MW, Kim JA. Inhibitory effects of constituents of Gastrodia elata Bl. on glutamate-induced apoptosis in IMR-32 human neuroblastoma cells. Arch Pharm Res 1999; 22:404-9. [PMID: 10489882 DOI: 10.1007/bf02979066] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The inhibitory effects of the constituents of Gastrodia elata Bl. (GE) on glutamate-induced apoptosis in human neuronal cells were investigated using IMR32 human neuroblastoma cells. Glutamate (GLU) induced DNA fragmentation, a hallmark of apoptosis, in a dose-dependent manner. GLU also induced a slow and sustained increase in intracellular Ca2+ concentration. Treatment with EGTA, an extracellular Ca2+ chelator, in a nominal Ca2+-free buffer solution abolished the GLU-induced intracellular Ca2+ increase, indicating that GLU stimulated Ca2+ influx pathway in the IMR32 cells. BAPTA, an intracellular Ca2+ chelator, significantly inhibited the GLU-induced apoptosis assessed by the flow cytometry measuring hypodiploid DNA content indicative of apoptosis, implying that intracellular Ca2+ rise may mediate the apoptotic action of GLU. Vanillin (VAN) and p-hydroxybenzaldehyde (p-HB), known constituents of GE, significantly inhibited both intracellular Ca2+ rise and apoptosis induced by GLU. These results suggest that the apoptosis-inhibitory actions of the constituents of GE may account, at least in part, for the basis of their antiepileptic activities. These results further suggest that intracellular Ca2+ signaling pathway may be a molecular target of the constituents of GE.
Collapse
Affiliation(s)
- Y S Lee
- Department of Physiology, College of Medicine, Kwandong University, Kangnung, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Otsuki K, Morimoto K, Sato K, Yamada N, Kuroda S. Effects of lamotrigine and conventional antiepileptic drugs on amygdala- and hippocampal-kindled seizures in rats. Epilepsy Res 1998; 31:101-12. [PMID: 9714501 DOI: 10.1016/s0920-1211(98)00018-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We investigated the anticonvulsant and adverse behavioral effects of lamotrigine (LTG), a novel antiepileptic drug (AED), as well as other conventional AEDs on kindled seizures in rats. We also applied an anticonvulsive dose of LTG in vivo to rats in which the hippocampus had been subjected to long-term potentiation (LTP). LTG potently attenuated limbic-kindled seizures in a dose-dependent fashion, at doses at which animals showed no adverse behavioral effects. LTG was effective in preventing kindled seizures for up to 24 h after a single i.p. administration. The anticonvulsant effects of LTG were reversed when the stimulus current was raised to two or three times the generalized seizure-triggering threshold. Among the AEDs examined, valproate and LTG were the only drugs that engendered a potent anticonvulsant effect without concomitant adverse behavioral effects. Although all of the other AEDs exhibited anticonvulsant effects with various potencies, they produced adverse effects such as sedation or motor ataxia. Furthermore, an anticonvulsant dose of LTG did not affect either the induction or maintenance of tetanus-induced LTP in the hippocampus. These results indicate that LTG potently suppresses limbic-kindled seizures by raising the seizure triggering-threshold in the kindling focus at doses that do not affect LTP in the hippocampus.
Collapse
Affiliation(s)
- K Otsuki
- Department of Neuropsychiatry, Okayama University Medical School, Japan.
| | | | | | | | | |
Collapse
|
44
|
|
45
|
|
46
|
Morimoto K, Sato H, Sato K, Sato S, Yamada N. BW1003C87, phenytoin and carbamazepine elevate seizure threshold in the rat amygdala-kindling model of epilepsy. Eur J Pharmacol 1997; 339:11-5. [PMID: 9450611 DOI: 10.1016/s0014-2999(97)01347-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We examined the anticonvulsant effects of BW1003C87 (5-(2,3,5-trichlorophenyl)-2,4-diaminopyrimidine ethane sulphonic acid), which is structurally related to the new antiepileptic drug, lamotrigine, and compared its effects to those of the conventional antiepileptic drugs, phenytoin and carbamazopine, using the rat amygdala-kindling model of epilepsy. BW1003C87 (2.5-10 mg/kg, i.p.) had potent and long-lasting (48 h after single administration) effects on amygdala-kindled seizures. The effects of BW1003C87 were completely reversed when the stimulus intensity was increased to 2 or 3 times the threshold determined. Since the same effects on seizure threshold were obtained for phenytoin and carbamazepine in the present study and for lamotrigine in our previous study, we propose that the principal mechanism of these antiepileptic drugs, which act primarily on voltage-sensitive Na+ channels, is significant elevation of the seizure threshold in epileptogenic foci and that BW1003C87 has a profile similar to that of these drugs.
Collapse
Affiliation(s)
- K Morimoto
- Department of Neuropsychiatry, Faculty of Medicine, Kagawa Medical University, Kita-gun, Japan
| | | | | | | | | |
Collapse
|