Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Transl Psychiatry ; 8(1): 139, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30065280

ABSTRACT

Withdrawal symptoms are common upon discontinuation of psychiatric medications. Catatonia, a neuropsychiatric condition proposed to be associated with gamma-aminobutyric acid (GABA) hypoactivity due to its robust response to benzodiazepines, has been described as a withdrawal syndrome in case reports but is not a well-recognized phenomenon. The authors undertook a review of withdrawal catatonia with an aim to understand its presentation as well as the medications and psychoactive substances it is associated with. The review identified 55 cases of withdrawal catatonia, the majority of which occurred upon discontinuation of benzodiazepines (24 cases) and discontinuation of clozapine (20 cases). No other antipsychotic medications were identified as having been associated with the onset of a catatonic episode within 2 weeks following their discontinuation. Increasing GABA activity and resultant GABA receptor adaptations with prolonged use is postulated as a shared pharmacological mechanism between clozapine and benzodiazepines that underlie their association with withdrawal catatonia. The existing evidence for clozapine's activity on the GABA system is reviewed. The clinical presentations of benzodiazepine withdrawal catatonia and clozapine withdrawal catatonia appear to differ and reasons for this are explored. One reason is that benzodiazepines act directly on GABAA receptors as allosteric agonists, while clozapine has more complex and indirect interactions, primarily through effects on receptors located on GABA interneurons. Another possible reason for the difference in clinical presentation is that clozapine withdrawal catatonia may also involve receptor adaptations in non-GABA receptors such as dopamine and acetylcholine. The findings from our review have implications for the treatment of withdrawal catatonia, and treatment recommendations are provided. Further research understanding the uniqueness of clozapine withdrawal catatonia among antipsychotic medication may give some insight as to clozapine's differential mechanism of action.


Subject(s)
Benzodiazepines/adverse effects , Catatonia/chemically induced , Clozapine/adverse effects , GABA Antagonists/adverse effects , Substance Withdrawal Syndrome/diagnosis , Humans
2.
J Vis Exp ; (136)2018 06 12.
Article in English | MEDLINE | ID: mdl-29985308

ABSTRACT

Pentylenetetrazole (PTZ) is a GABA-A receptor antagonist. An intraperitoneal injection of PTZ into an animal induces an acute, severe seizure at a high dose, whereas sequential injections of a subconvulsive dose have been used for the development of chemical kindling, an epilepsy model. A single low-dose injection of PTZ induces a mild seizure without convulsion. However, repetitive low-dose injections of PTZ decrease the threshold to evoke a convulsive seizure. Finally, continuous low-dose administration of PTZ induces a severe tonic-clonic seizure. This method is simple and widely applicable to investigate the pathophysiology of epilepsy, which is defined as a chronic disease that involves repetitive seizures. This chemical kindling protocol causes repetitive seizures in animals. With this method, vulnerability to PTZ-mediated seizures or the degree of aggravation of epileptic seizures was estimated. These advantages have led to the use of this method for screening anti-epileptic drugs and epilepsy-related genes. In addition, this method has been used to investigate neuronal damage after epileptic seizures because the histological changes observed in the brains of epileptic patients also appear in the brains of chemical-kindled animals. Thus, this protocol is useful for conveniently producing animal models of epilepsy.


Subject(s)
Brain/drug effects , GABA Antagonists/therapeutic use , Kindling, Neurologic/pathology , Pentylenetetrazole/therapeutic use , Seizures/chemically induced , Animals , Disease Models, Animal , GABA Antagonists/adverse effects , GABA Antagonists/pharmacology , Injections, Intraperitoneal , Male , Mice , Pentylenetetrazole/adverse effects , Pentylenetetrazole/pharmacology
3.
Free Radic Biol Med ; 114: 33-39, 2018 01.
Article in English | MEDLINE | ID: mdl-28993272

ABSTRACT

Down syndrome (DS), also known as trisomy 21, is the most common genetic cause of intellectual disability. It is also a model human disease for exploring consequences of gene dosage imbalance on complex phenotypes. Learning and memory impairments linked to intellectual disabilities in DS could result from synaptic plasticity deficits and excitatory-inhibitory alterations leading to changes in neuronal circuitry in the brain of affected individuals. Increasing number of studies in mouse and cellular models converge towards the assumption that excitatory-inhibitory imbalance occurs in DS, likely early during development. Thus increased inhibition appears to be a common trend that could explain synaptic and circuit disorganization. Interestingly using several potent pharmacological tools, preclinical studies strongly demonstrated that cognitive deficits could be restored in mouse models of DS. Clinical trials have not yet provided robust data for therapeutic application and additional studies are needed. Here we review the literature and our own published work emphasizing the over-inhibition hypothesis in DS and their links with gene dosage imbalance paving the way for future basic and clinical research.


Subject(s)
Cognition Disorders/chemically induced , Down Syndrome/drug therapy , GABA Antagonists/adverse effects , Receptors, GABA-A/chemistry , Signal Transduction/drug effects , Animals , Disease Models, Animal , Humans , Receptors, GABA-A/drug effects
4.
Synapse ; 71(7)2017 07.
Article in English | MEDLINE | ID: mdl-28245513

ABSTRACT

ONO-2952, a novel antagonist of translocator protein 18 kDa (TSPO), binds with high affinity to TSPO in rat brain and human tumor cell line membrane preparations. This study used the TSPO-specific PET radioligand [11 C]PBR28 to confirm binding of ONO-2952 to brain TSPO in human subjects, and evaluate brain TSPO occupancy and its relationship with ONO-2952 plasma concentration. Sixteen healthy subjects received a single oral dose of 200, 60, 20, or 6 mg ONO-2952 (n = 4 per dose). Two PET scans with [11 C]PBR28 were conducted ≤7 days apart: at baseline and 24 h after ONO-2952 administration. [11 C]PBR28 regional distribution volume (VT ) was derived with kinetic modeling using the arterial input function and a two tissue compartment model. Nonspecific binding (VND ) was obtained on an individual basis for each subject using linear regression as the x-intercept of the Lassen plot. The binding potential relative to VND (BPND ) was derived as the difference between VT in the ROI (VT ROI) and VND , normalized to VND ; BPND = (VT ROI - VND )/VND . TSPO occupancy was calculated as the change in BPND (ΔBPND ) from individual's baseline scan to the on-medication scan to the baseline BPND value. TSPO occupancy by ONO-2952 was dose dependent between 20-200 mg, approaching saturation at 200 mg both in the whole brain and in 15 anatomic regions of interest (ROI). Estimated Ki values ranged from 24.1 to 72.2 nM. This open-label, single-center, single-dose study demonstrated engagement of ONO-2952 to brain TSPO. The relationship between pharmacokinetics and TSPO occupancy observed in this study support the hypothesis that ONO-2952 could potentially modulate neurosteroid production by binding to brain TSPO.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Cyclopropanes/pharmacology , GABA Antagonists/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Receptors, GABA/metabolism , Acetamides , Adult , Carbon Radioisotopes , Cyclopropanes/adverse effects , Cyclopropanes/blood , Dose-Response Relationship, Drug , Female , Follow-Up Studies , GABA Antagonists/adverse effects , GABA Antagonists/blood , Heterocyclic Compounds, 4 or More Rings/adverse effects , Heterocyclic Compounds, 4 or More Rings/blood , Humans , Linear Models , Magnetic Resonance Imaging , Male , Middle Aged , Positron-Emission Tomography , Pyridines , Radiopharmaceuticals , Young Adult
6.
Epilepsy Behav ; 57(Pt A): 95-104, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26930359

ABSTRACT

Resistance to antiepileptic drugs (AEDs) is a major problem in epilepsy therapy, so that development of more effective AEDs is an unmet clinical need. Several rat and mouse models of epilepsy with spontaneous difficult-to-treat seizures exist, but because testing of antiseizure drug efficacy is extremely laborious in such models, they are only rarely used in the development of novel AEDs. Recently, the use of acute seizure tests in epileptic rats or mice has been proposed as a novel strategy for evaluating novel AEDs for increased antiseizure efficacy. In the present study, we compared the effects of five AEDs (valproate, phenobarbital, diazepam, lamotrigine, levetiracetam) on the pentylenetetrazole (PTZ) seizure threshold in mice that were made epileptic by pilocarpine. Experiments were started 6 weeks after a pilocarpine-induced status epilepticus. At this time, control seizure threshold was significantly lower in epileptic than in nonepileptic animals. Unexpectedly, only one AED (valproate) was less effective to increase seizure threshold in epileptic vs. nonepileptic mice, and this difference was restricted to doses of 200 and 300 mg/kg, whereas the difference disappeared at 400mg/kg. All other AEDs exerted similar seizure threshold increases in epileptic and nonepileptic mice. Thus, induction of acute seizures with PTZ in mice pretreated with pilocarpine does not provide an effective and valuable surrogate method to screen drugs for antiseizure efficacy in a model of difficult-to-treat chronic epilepsy as previously suggested from experiments with this approach in rats.


Subject(s)
Anticonvulsants/therapeutic use , GABA Antagonists/administration & dosage , Pentylenetetrazole/administration & dosage , Seizures/drug therapy , Animals , Diazepam/therapeutic use , Disease Models, Animal , Drug Resistance , Epilepsy/drug therapy , GABA Antagonists/adverse effects , Levetiracetam , Male , Mice , Pentylenetetrazole/adverse effects , Phenobarbital/therapeutic use , Pilocarpine , Piracetam/analogs & derivatives , Rats , Status Epilepticus/drug therapy , Valproic Acid/therapeutic use
7.
Regul Toxicol Pharmacol ; 75: 46-57, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26724267

ABSTRACT

The adverse outcome pathway (AOP) is a framework to mechanistically link molecular initiating events to adverse biological outcomes. From a regulatory perspective, it is of crucial importance to determine the confidence for the AOP in question as well as the quality of data available in supporting this evaluation. A weight of evidence approach has been proposed for this task, but many of the existing frameworks for weight of evidence evaluation are qualitative and there is not clear guidance regarding how weight of evidence should be calculated for an AOP. In this paper we advocate the use of a subject matter expertise driven approach for weight of evidence evaluation based on criteria and metrics related to data quality and the strength of causal linkages between key events. As a demonstration, we notionally determine weight of evidence scores for two AOPs: Non-competitive ionotropic GABA receptor antagonism leading to epileptic seizures, and Antagonist-binding and stabilization of a co-repressor to the peroxisome proliferator-activated receptor α (PPARα) signaling complex ultimately causing starvation-like weight loss.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Models, Biological , Animals , Emaciation/chemically induced , Epilepsy/chemically induced , GABA Antagonists/adverse effects , Humans , Membrane Transport Modulators/adverse effects , PPAR alpha/antagonists & inhibitors , Risk Assessment , Weight Loss/drug effects
8.
Biomark Med ; 9(11): 1225-39, 2015.
Article in English | MEDLINE | ID: mdl-26508561

ABSTRACT

Antagonism of ionotropic GABA receptors (iGABARs) can occur at three distinct types of receptor binding sites causing chemically induced epileptic seizures. Here we review three adverse outcome pathways, each characterized by a specific molecular initiating event where an antagonist competitively binds to active sites, negatively modulates allosteric sites or noncompetitively blocks ion channel on the iGABAR. This leads to decreased chloride conductance, followed by depolarization of affected neurons, epilepsy-related death and ultimately decreased population. Supporting evidence for causal linkages from the molecular to population levels is presented and differential sensitivity to iGABAR antagonists in different GABA receptors and organisms discussed. Adverse outcome pathways are poised to become important tools for linking mechanism-based biomarkers to regulated outcomes in next-generation risk assessment.


Subject(s)
Biomarkers/metabolism , GABA Antagonists/adverse effects , Nervous System/drug effects , Receptors, GABA/metabolism , Animals , Humans , Nervous System/metabolism
9.
Curr Pharm Des ; 21(23): 3325-42, 2015.
Article in English | MEDLINE | ID: mdl-26088116

ABSTRACT

This review paper discusses the central role of gamma-aminobutyric acid (GABA) in diverse physiological systems and functions and the therapeutic potential of the benzodiazepine antagonist flumazenil (Ro 15- 1788) for a wide range of disorders of the central nervous system (CNS). Our group and others have studied the potential of flumazenil as a treatment for benzodiazepine dependence. A small but growing body of research has indicated that flumazenil may also have clinical application in CNS disorders such as Parkinson's disease, idiopathic hypersomnia and amyotrophic lateral sclerosis. Despite this body of research the therapeutic potential of flumazenil remains poorly understood and largely unrealized. The purpose of this paper is not to provide an exhaustive review of all possible therapeutic applications for flumazenil but rather to stimulate research interest, and discussion of the exciting therapeutic potential of this drug for a range of chronic debilitating conditions.


Subject(s)
Central Nervous System Diseases/drug therapy , Central Nervous System/drug effects , Flumazenil/therapeutic use , GABA Antagonists/therapeutic use , Off-Label Use , gamma-Aminobutyric Acid/metabolism , Animals , Central Nervous System/metabolism , Central Nervous System/physiopathology , Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/physiopathology , Flumazenil/adverse effects , GABA Antagonists/adverse effects , Humans , Inappropriate Prescribing , Patient Safety , Practice Patterns, Physicians' , Risk Assessment , Risk Factors , Treatment Outcome
10.
Parasite ; 21: 42, 2014.
Article in English | MEDLINE | ID: mdl-25148564

ABSTRACT

The speed of efficacy of afoxolaner (NexGard) against Ctenocephalides felis fleas was evaluated in two studies. Study A assessed the efficacy against existing fleas whereas study B assessed the efficacy against new infesting fleas. In study A, 12 dogs were allocated to the untreated group and 20 dogs to the treated group. All dogs were infested by 100 fleas each at Day -1, treated at Day 0 and flea combed at 2 h or at 6 h post treatment. In study B, 6 dogs were allocated to the untreated group and 10 to the treated group. They were infested with 100 fleas each on Days 2, 7, 14, 21 and 28. Fleas were removed and counted at 6 h post-infestation. Immediate and persistent efficacies were evaluated by counting fleas on the dogs. To evaluate induced mortality after exposure on dogs, fleas collected alive were placed in an insectarium for 24 h and assessed for viability. The immediate efficacy on dogs was significant at 6 h with 100%. The induced death of the fleas collected live from dogs 2 h after exposure was 99.7%. Concerning new infesting fleas, the observed efficacy at 6 h and the induced mortality were significantly different (p < 0.05) from the control at all time-points. At 6 h, the prophylactic efficacy was > 97% at Day 2 and Day 8 and > 90% at Day 14. The induced mortality after 6 h of exposure on dogs varied between 73.3% and 100% for the whole study.


Subject(s)
Ctenocephalides , Dog Diseases/drug therapy , Flea Infestations/veterinary , Insecticides/therapeutic use , Isoxazoles/therapeutic use , Naphthalenes/therapeutic use , Animals , Ctenocephalides/drug effects , Dogs , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/adverse effects , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Flea Infestations/drug therapy , GABA Antagonists/administration & dosage , GABA Antagonists/adverse effects , GABA Antagonists/pharmacology , GABA Antagonists/therapeutic use , Insect Proteins/antagonists & inhibitors , Insecticides/administration & dosage , Insecticides/adverse effects , Insecticides/pharmacology , Isoxazoles/administration & dosage , Isoxazoles/adverse effects , Isoxazoles/pharmacology , Naphthalenes/administration & dosage , Naphthalenes/adverse effects , Naphthalenes/pharmacology , Time Factors , Treatment Outcome
11.
Pediatr Emerg Care ; 28(1): 55-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22217888

ABSTRACT

Oculogyric crisis is a neurologic adverse event characterized by bilateral dystonic, usually upward, conjugate eye deviations. Cefixime is a third-generation cephalosporin and is widely used in clinical practice in childhood. Confusion, encephalopathy, coma, myoclonus, nonconvulsive status epilepticus, and seizures have been described with the use of cephalosporins. We presented a cefixime-induced oculogyric crisis in a 7-year-old boy during the treatment of urinary tract infection, and this is the first case of cefixime-induced oculogyric crisis whose ocular symptoms gradually disappeared within 48 hours after the drug was discontinued.


Subject(s)
Anti-Bacterial Agents/adverse effects , Cefixime/adverse effects , Ocular Motility Disorders/chemically induced , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Attention Deficit Disorder with Hyperactivity/complications , Cefixime/pharmacology , Cefixime/therapeutic use , Child , Dopamine Antagonists/adverse effects , Dopamine Antagonists/pharmacology , GABA Antagonists/adverse effects , GABA Antagonists/pharmacology , Humans , Male , Neurologic Examination , Ocular Motility Disorders/complications , Ocular Motility Disorders/physiopathology , Postoperative Complications/drug therapy , Recurrence , Staphylococcal Infections/complications , Staphylococcal Infections/drug therapy , Urinary Tract Infections/complications , Urinary Tract Infections/drug therapy
12.
Neuroscience ; 201: 288-96, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22100272

ABSTRACT

In hippocampal Cornu Ammonis 1 (CA1) neurons, a prolonged depolarization evokes a train of action potentials followed by a prominent afterhyperpolarizing potential (AHP), which critically dampens neuronal excitability. Because it is not known whether epileptiform activity alters the AHP and whether any alteration of the AHP is independent of inhibition, we acutely induced epileptiform activity by bath application of the GABA(A) receptor blocker gabazine (5 µM) in the rat hippocampal slice preparation and studied its impact on the AHP using intracellular recordings. Following 10 min of gabazine wash-in, slices started to develop spontaneous epileptiform discharges. This disinhibition was accompanied by a significant shift of the resting membrane potential of CA1 neurons to more depolarized values. Prolonged depolarizations (600 ms) elicited a train of action potentials, the number of which was not different between baseline and gabazine treatment. However, the AHP following the train of action potentials was significantly reduced after 20 min of gabazine treatment. When the induction of epileptiform activity was prevented by co-application of 6-cyano-7-nitroquinoxaline-2,3-dione disodium (CNQX, 10 µM) and D-(-)-2-amino-5-phosphonopentanoic acid (D-AP5, 50 µM) to block α-amino-3-hydroxy-5-methylisoxazolepropionate (AMPA) and N-methyl-d-aspartate (NMDA) receptors, respectively, the AHP was preserved despite of GABA(A) receptor inhibition suggesting that the epileptiform activity was required to suppress the AHP. Moreover, the AHP was also preserved when the slices were treated with the protein kinase blockers H-9 (100 µM) and H-89 (1 µM). These results demonstrate that the AHP following a train of action potentials is rapidly suppressed by acutely induced epileptiform activity due to a phosphorylation process-presumably involving protein kinase A.


Subject(s)
Action Potentials/physiology , CA1 Region, Hippocampal/pathology , Epilepsy/pathology , Neural Inhibition/physiology , Neurons/physiology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Disease Models, Animal , Drug Interactions , Electric Stimulation , Enzyme Inhibitors/pharmacology , Epilepsy/chemically induced , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/adverse effects , In Vitro Techniques , Isoquinolines/pharmacology , Male , Neural Inhibition/drug effects , Pyridazines/adverse effects , Rats , Rats, Wistar , Statistics, Nonparametric , Sulfonamides/pharmacology , Time Factors
13.
J Child Adolesc Psychopharmacol ; 21(6): 565-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22136091

ABSTRACT

To date, placebo-controlled drug trials targeting the core social impairment of autistic disorder (autism) have had uniformly negative results. Given this, the search for new potentially novel agents targeting the core social impairment of autism continues. Acamprosate is U.S. Food and Drug Administration-approved drug to treat alcohol dependence. The drug likely impacts both gamma-aminobutyric acid and glutamate neurotransmission. This study describes our initial open-label experience with acamprosate targeting social impairment in youth with autism. In this naturalistic report, five of six youth (mean age, 9.5 years) were judged treatment responders to acamprosate (mean dose 1,110 mg/day) over 10 to 30 weeks (mean duration, 20 weeks) of treatment. Acamprosate was well tolerated with only mild gastrointestinal adverse effects noted in three (50%) subjects.


Subject(s)
Autistic Disorder/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , GABA Antagonists/therapeutic use , Social Behavior Disorders/drug therapy , Taurine/analogs & derivatives , Acamprosate , Autistic Disorder/complications , Child , Excitatory Amino Acid Antagonists/adverse effects , GABA Antagonists/adverse effects , Humans , Pilot Projects , Psychiatric Status Rating Scales/statistics & numerical data , Social Behavior Disorders/complications , Taurine/adverse effects , Taurine/therapeutic use
14.
Anaesthesist ; 60(10): 946-9, 2011 Oct.
Article in German | MEDLINE | ID: mdl-21728048

ABSTRACT

The increased use of phytotherapeutic drugs means that anesthetists are more often confronted with these drugs. In this case report possible problems which can occur are demonstrated exemplified by silexan. Silexan is a phytotherapeutic anxiolytic which is used in anxiety disorders. Because of its potential mechanism of action via the neurotransmitter gamma-aminobutyric acid (GABA) receptors interactions with narcotic drugs are possible. The case of an 18-year-old girl who underwent an operation under general anesthesia while taking silexan as long-term medication is presented. The desired depth of narcosis could only be reached by inhalative induction with sevoflurane after unsuccessful induction attempts using intravenous propofol and thiopental. Possible explanations for this route and inhalative induction as a possible alternative are discussed.


Subject(s)
Anesthetics/adverse effects , Oils, Volatile/adverse effects , Plant Oils/adverse effects , Adolescent , Anesthesia, Intravenous , Anesthetics, Intravenous/adverse effects , Arachnoid Cysts/surgery , Cerebellar Diseases/surgery , Drug Interactions , Female , GABA Antagonists/adverse effects , Humans , Hypnotics and Sedatives , Injections, Intravenous , Lavandula , Neurosurgical Procedures , Obesity/complications , Postoperative Nausea and Vomiting/prevention & control , Propofol/adverse effects , Receptors, GABA-A/drug effects , Thiopental
15.
PLoS Comput Biol ; 7(3): e1002016, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21483481

ABSTRACT

In the era of personalized medical practice, understanding the genetic basis of patient-specific adverse drug reaction (ADR) is a major challenge. Clozapine provides effective treatments for schizophrenia but its usage is limited because of life-threatening agranulocytosis. A recent high impact study showed the necessity of moving clozapine to a first line drug, thus identifying the biomarkers for drug-induced agranulocytosis has become important. Here we report a methodology termed as antithesis chemical-protein interactome (CPI), which utilizes the docking method to mimic the differences in the drug-protein interactions across a panel of human proteins. Using this method, we identified HSPA1A, a known susceptibility gene for CIA, to be the off-target of clozapine. Furthermore, the mRNA expression of HSPA1A-related genes (off-target associated systems) was also found to be differentially expressed in clozapine treated leukemia cell line. Apart from identifying the CIA causal genes we identified several novel candidate genes which could be responsible for agranulocytosis. Proteins related to reactive oxygen clearance system, such as oxidoreductases and glutathione metabolite enzymes, were significantly enriched in the antithesis CPI. This methodology conducted a multi-dimensional analysis of drugs' perturbation to the biological system, investigating both the off-targets and the associated off-systems to explore the molecular basis of an adverse event or the new uses for old drugs.


Subject(s)
Agranulocytosis/chemically induced , Clozapine/adverse effects , Drug-Related Side Effects and Adverse Reactions/chemically induced , Protein Interaction Mapping/methods , Benzodiazepines/pharmacology , Biomarkers/metabolism , Cell Line, Tumor , Computational Biology/methods , GABA Antagonists/adverse effects , HSP70 Heat-Shock Proteins/genetics , Humans , Olanzapine , Proteomics/methods , Reactive Oxygen Species , Retrospective Studies , Risk Factors
16.
J Neurosci ; 31(6): 2101-12, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21307247

ABSTRACT

Ischemic brain injury is a major problem associated with stroke. It has been increasingly recognized that acid-sensing ion channels (ASICs) contribute significantly to ischemic neuronal damage, but the underlying mechanism has remained elusive. Here, we show that extracellular spermine, one of the endogenous polyamines, exacerbates ischemic neuronal injury through sensitization of ASIC1a channels to extracellular acidosis. Pharmacological blockade of ASIC1a or deletion of the ASIC1 gene greatly reduces the enhancing effect of spermine in ischemic neuronal damage both in cultures of dissociated neurons and in a mouse model of focal ischemia. Mechanistically, spermine profoundly reduces desensitization of ASIC1a by slowing down desensitization in the open state, shifting steady-state desensitization to more acidic pH, and accelerating recovery between repeated periods of acid stimulation. Spermine-mediated potentiation of ASIC1a activity is occluded by PcTX1 (psalmotoxin 1), a specific ASIC1a inhibitor binding to its extracellular domain. Functionally, the enhanced channel activity is accompanied by increased acid-induced neuronal membrane depolarization and cytoplasmic Ca(2+) overload, which may partially explain the exacerbated neuronal damage caused by spermine. More importantly, blocking endogenous spermine synthesis significantly attenuates ischemic brain injury mediated by ASIC1a but not that by NMDA receptors. Thus, extracellular spermine contributes significantly to ischemic neuronal injury through enhancing ASIC1a activity. Our data suggest new neuroprotective strategies for stroke patients via inhibition of polyamine synthesis and subsequent spermine-ASIC interaction.


Subject(s)
Acidosis/physiopathology , Extracellular Fluid/drug effects , Infarction, Middle Cerebral Artery/pathology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Sodium Channels/metabolism , Spermine/pharmacology , Acid Sensing Ion Channels , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/deficiency , Amino Acid Transport System y+/metabolism , Animals , Biophysics , Brain Injuries/chemically induced , CHO Cells , Calcium/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Electric Stimulation , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/adverse effects , Glucose/deficiency , Hippocampus/cytology , Hydrogen-Ion Concentration , Hypoxia , L-Lactate Dehydrogenase/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Knockout , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/pathology , Oligonucleotides/pharmacology , Patch-Clamp Techniques/methods , Picrotoxin/adverse effects , Putrescine/pharmacology , Sodium Channels/genetics , Spermine/adverse effects , Tetrazolium Salts , Time Factors , Transfection , Valine/analogs & derivatives , Valine/pharmacology
17.
Bull Exp Biol Med ; 150(6): 665-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-22235411

ABSTRACT

The hyperkinekic effect (increase in spontaneous activity and development of choreomyoclonic hyperkinesis of the extremities and body) of picrotoxin injected into the rostral neostriatum of rats in a dose of 2 µg was reduced if the drug was injected together with ZnCl(2) in a concentration of 0.1 µg/µl. ZnCl(2) in a concentration of 1 µg/µl did not modulate the effects of picrotoxin, while in a concentration of 3 µg/µl it increased spontaneous motor activity in the open field test without affecting the symptoms of choreomyoclonic hyperkinesis.


Subject(s)
Chlorides/pharmacology , GABA Antagonists/adverse effects , Hyperkinesis/chemically induced , Neostriatum/drug effects , Picrotoxin/adverse effects , Zinc Compounds/pharmacology , Animals , Drug Synergism , GABA Antagonists/administration & dosage , GABA Antagonists/pharmacology , Male , Picrotoxin/administration & dosage , Picrotoxin/pharmacology , Rats , Rats, Wistar
18.
Epilepsy Res ; 92(2-3): 153-62, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20851574

ABSTRACT

Epileptic seizures typically result in delayed neuronal loss secondary to the initial damage and an up-regulation in connexin43 (Cx43). This study investigated the role of Cx43 gap junctions in lesion spread and cell loss following epileptiform activity. Epileptiform injury in hippocampal slice cultures was induced by 48 h exposure to 100 µM bicuculline methochloride (BMC). During the 24h recovery period following BMC treatment, lesion spread was observed in the CA1. A Cx43 mimetic peptide, applied during either the BMC treatment or recovery periods, produced concentration- and exposure time-dependent neuroprotection, as measured by propidium iodide uptake at the end of the recovery period. During the BMC period, peptide concentrations between 5 and 50 µM (sufficient to block hemichannels) had a protective effect while a substantial gap junction blockade with 500 µM peptide exacerbated the lesion. By contrast, all doses applied during the recovery period protected the CA1 region from further damage. The results indicate that while the slices are undergoing excessive neuronal firing and epileptic stress, gap junction communication appears to be essential for tissue survival but hemichannel opening may be damaging. Following epileptiform insult, however, gap junction communication plays a crucial role in the spread of neuronal damage. The findings from this study identify gap junction communication as a potential therapeutic target for epilepsy.


Subject(s)
CA1 Region, Hippocampal/pathology , Connexin 43/pharmacology , Nerve Degeneration/prevention & control , Animals , Animals, Newborn , Bicuculline/adverse effects , Bicuculline/analogs & derivatives , CA1 Region, Hippocampal/drug effects , Cell Death/drug effects , Culture Media, Serum-Free/adverse effects , Dose-Response Relationship, Drug , GABA Antagonists/adverse effects , In Vitro Techniques , Microtubule-Associated Proteins/metabolism , Nerve Degeneration/etiology , Peptides/chemistry , Peptides/pharmacology , Rats , Rats, Wistar , Time Factors
19.
Ross Fiziol Zh Im I M Sechenova ; 96(3): 283-92, 2010 Mar.
Article in Russian | MEDLINE | ID: mdl-20535998

ABSTRACT

Effects of the inhibitor of the expression of Heat shock proteins 70 kDa (Hsp70), quercetin on seizures and movement disorders induced by N-methyl D-aspartate (NMDA) or pentylenetetrazole in adult rats Wistar were investigated using behavioral methods. It was found that intraperitoneal injection of quercetin 4 hours before intraventricular microinjection of NMDA resulted in increased duration of tonic component of seizures, seizure and ataxia symptoms severity. Blockade of the expression of Hsp70 by quercetin increased the duration of clonic and tonic seizures and did not affect severity of seizures and ataxia symptoms, induced by intraperitoneal injection of pentylenetetrazole. Immunoblotting showed that injection of quercetin resulted in reduced content of the inducible form of Hsp70 in the hippocampus, thalamus and corpus callosum. The obtained results indicate proconvulsant effect of quercetin associated with the inhibition of Hsp70 expression. These data suggest involvement of Hp70 in regulation of central mechanisms of behavioral seizures and motor disorders induced by NMDA and pentylenetetrazole in rats.


Subject(s)
Antioxidants/pharmacology , Corpus Callosum/metabolism , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Quercetin/pharmacology , Seizures/metabolism , Animals , Ataxia/chemically induced , Ataxia/metabolism , Behavior, Animal/drug effects , Excitatory Amino Acid Agonists/adverse effects , Excitatory Amino Acid Agonists/pharmacology , GABA Antagonists/adverse effects , GABA Antagonists/pharmacology , HSP70 Heat-Shock Proteins , Male , N-Methylaspartate/adverse effects , N-Methylaspartate/pharmacology , Pentylenetetrazole/adverse effects , Pentylenetetrazole/pharmacology , Rats , Rats, Wistar , Seizures/chemically induced , Severity of Illness Index
20.
Clin Exp Pharmacol Physiol ; 37(4): 435-40, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19878215

ABSTRACT

1. The diuretic amiloride is known to modulate the activity of several types of ion channels and membrane receptors in addition to its inhibitory effects on many ion transport systems. However, the effects of amiloride on some important ion channels and receptors, such as GABA(A) receptors, in the central nervous system have not been characterized. 2. In the present study, we investigated the functional action of amiloride on native GABA(A) receptors in cultured neurons of rat inferior colliculus using whole-cell patch-clamp recordings. 3. Amiloride reversibly inhibited the amplitude of the GABA-induced current (I(GABA)) in a concentration-dependent manner (IC(50) 454 +/- 24 micromol/L) under conditions of voltage-clamp with a holding potential at -60 mV. The inhibition depended on drug application mode and was independent of membrane potential. Amiloride did not change the reversal potential of I(GABA). Moreover, amiloride induced a parallel right-ward shift in the concentration-response curve for I(GABA) without altering the maximal value and Hill coefficient. 4. The present study shows that amiloride competitively inhibits the current mediated by native GABA(A) receptors in the brain region, probably via a direct action on GABA-binding sites on the receptor. The findings suggest that the functional actions of amiloride on GABA(A) receptors may result in possible side-effects on the central nervous system in the case of direct application of this drug into the cerebrospinal fluid for treatment of diseases such as brain tumours.


Subject(s)
Amiloride/pharmacology , Diuretics/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Inferior Colliculi/drug effects , Neurons/drug effects , Synaptic Potentials/drug effects , Amiloride/adverse effects , Animals , Animals, Newborn , Cells, Cultured , Diuretics/adverse effects , GABA Antagonists/adverse effects , Inferior Colliculi/cytology , Kinetics , Patch-Clamp Techniques , Rats , Rats, Wistar , Sodium Channel Blockers/adverse effects , Sodium Channel Blockers/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...