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










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 388(2): 347-357, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37977809

ABSTRACT

Benzodiazepine pharmacoresistance develops when treatment of status epilepticus (SE) is delayed. This response may result from gamma-aminobutyric acid A receptors (GABAAR) internalization that follows prolonged SE; this receptor trafficking results in fewer GABAAR in the synapse to restore inhibition. Increase in synaptic N-methyl-D-aspartate receptors (NMDAR) also occurs in rodent models of SE. Lacosamide, a third-generation antiseizure medication (ASM), acts on the slow inactivation of voltage-gated sodium channels. Another ASM, rufinamide, similarly acts on sodium channels by extending the duration of time spent in the inactivation stage. Combination therapy of the benzodiazepine midazolam, NMDAR antagonist ketamine, and ASMs lacosamide (or rufinamide) was investigated for efficacy against soman (GD)-induced SE and neuropathology. Adult male rats implanted with telemetry transmitters for monitoring electroencephalographic (EEG) activity were exposed to a seizure-inducing dose of GD and treated with an admix of atropine sulfate and HI-6 1 minute later and with midazolam monotherapy or combination therapy 40 minutes after EEG seizure onset. Rats were monitored continuously for seizure activity for two weeks, after which brains were processed for assessment of neurodegeneration, neuronal loss, and neuroinflammatory responses. Simultaneous administration of midazolam, ketamine, and lacosamide (or rufinamide) was more protective against GD-induced SE compared with midazolam monotherapy. In general, lacosamide triple therapy had more positive outcomes on measures of epileptogenesis, EEG power integral, and the number of brain regions protected from neuropathology compared with rats treated with rufinamide triple therapy. Overall, both drugs were well tolerated in these combination models. SIGNIFICANCE STATEMENT: We currently report on improved efficacy of antiseizure medications lacosamide and rufinamide, each administered in combination with ketamine (NMDAR antagonist) and midazolam (benzodiazepine), in combatting soman (GD)-induced seizure, epileptogenesis, and brain pathology over that provided by midazolam monotherapy, or dual therapy of midazolam and lacosamide (or rufinamide) in rats. Administration of lacosamide as adjunct to midazolam and ketamine was particularly effective against GD-induced toxicity. However, protection was incomplete, suggesting the need for further study.


Subject(s)
Ketamine , Soman , Status Epilepticus , Triazoles , Rats , Male , Animals , Midazolam/therapeutic use , Midazolam/pharmacology , Lacosamide/adverse effects , Ketamine/pharmacology , Ketamine/therapeutic use , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Seizures/drug therapy , Benzodiazepines , Cholinergic Agents/adverse effects , gamma-Aminobutyric Acid
2.
J Pharmacol Exp Ther ; 388(2): 376-385, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37770198

ABSTRACT

Status epilepticus (SE) is a life-threatening development of self-sustaining seizures that becomes resistant to benzodiazepines when treatment is delayed. Benzodiazepine pharmacoresistance is thought in part to result from internalization of synaptic GABAA receptors, which are the main target of the drug. The naturally occurring neurosteroid allopregnanolone is a therapy of interest against SE for its ability to modulate all isoforms of GABAA receptors. Ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, has been partially effective in combination with benzodiazepines in mitigating SE-associated neurotoxicity. In this study, allopregnanolone as an adjunct to midazolam or midazolam-ketamine combination therapy was evaluated for efficacy against cholinergic-induced SE. Adult male rats implanted with electroencephalographic (EEG) telemetry devices were exposed to the organophosphorus chemical (OP) soman (GD) and treated with an admix of atropine sulfate and HI-6 at 1 minute after exposure followed by midazolam, midazolam-allopregnanolone, or midazolam-ketamine-allopregnanolone 40 minutes after seizure onset. Neurodegeneration, neuronal loss, and neuroinflammation were assessed 2 weeks after GD exposure. Seizure activity, EEG power integral, and epileptogenesis were also compared among groups. Overall, midazolam-ketamine-allopregnanolone combination therapy was effective in reducing cholinergic-induced toxic signs and neuropathology, particularly in the thalamus and hippocampus. Higher dosage of allopregnanolone administered in combination with midazolam and ketamine was also effective in reducing EEG power integral and epileptogenesis. The current study reports that there is a promising potential of neurosteroids in combination with benzodiazepine and ketamine treatments in a GD model of SE. SIGNIFICANCE STATEMENT: Allopregnanolone, a naturally occurring neurosteroid, reduced pathologies associated with soman (GD) exposure such as epileptogenesis, neurodegeneration, and neuroinflammation, and suppressed GD-induced toxic signs when used as an adjunct to midazolam and ketamine in a delayed treatment model of soman-induced status epilepticus (SE) in rats. However, protection was incomplete, suggesting that further studies are needed to identify optimal combinations of antiseizure medications and routes of administration for maximal efficacy against cholinergic-induced SE.


Subject(s)
Ketamine , Neurosteroids , Soman , Status Epilepticus , Rats , Male , Animals , Midazolam/pharmacology , Midazolam/therapeutic use , Ketamine/pharmacology , Ketamine/therapeutic use , Pregnanolone/adverse effects , Soman/toxicity , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Neuroinflammatory Diseases , Neurosteroids/therapeutic use , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Seizures/drug therapy , Benzodiazepines , Cholinergic Agents/adverse effects , Receptors, GABA-A , gamma-Aminobutyric Acid
3.
Epilepsia Open ; 6(4): 757-769, 2021 12.
Article in English | MEDLINE | ID: mdl-34657398

ABSTRACT

OBJECTIVE: Cholinergic-induced status epilepticus (SE) is associated with a loss of synaptic gamma-aminobutyric acid A receptors (GABAA R) and an increase in N-methyl-D-aspartate receptors (NMDAR) and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) that may contribute to pharmacoresistance when treatment with benzodiazepine antiseizure medication is delayed. The barbiturate phenobarbital enhances inhibitory neurotransmission by binding to a specific site in the GABAA R to increase the open state of the channel, decrease neuronal excitability, and reduce glutamate-induced currents through AMPA/kainate receptors. We hypothesized that phenobarbital as an adjunct to midazolam would augment the amelioration of soman-induced SE and associated neuropathological changes and that further protection would be provided by the addition of an NMDAR antagonist. METHODS: We investigated the efficacy of combining antiseizure medications to include a benzodiazepine and a barbiturate allosteric GABAA R modulator (midazolam and phenobarbital, respectively) to correct loss of inhibition, and ketamine to reduce excitation caused by increased synaptic localization of NMDAR and AMPAR, which are NMDA-dependent. Rats implanted with transmitters to record electroencephalographic (EEG) activity were exposed to soman and treated with atropine sulfate and HI-6 one min after exposure and with antiseizure medication(s) 40 minutes after seizure onset. RESULTS: The triple therapy combination of phenobarbital, midazolam, and ketamine administered at 40 minutes after seizure onset effectively prevented soman-induced epileptogenesis and reduced neurodegeneration. In addition, dual therapy with phenobarbital and midazolam or ketamine was more effective than monotherapy (midazolam or phenobarbital) in reducing cholinergic-induced toxicity. SIGNIFICANCE: Benzodiazepine efficacy is drastically reduced with time after seizure onset and inversely related to seizure duration. To overcome pharmacoresistance in severe benzodiazepine-refractory cholinergic-induced SE, simultaneous drug combination to include drugs that target both the loss of inhibition (eg, midazolam, phenobarbital) and the increased excitatory response (eg, ketamine) is more effective than benzodiazepine or barbiturate monotherapy.


Subject(s)
Ketamine , Soman , Animals , Anticonvulsants/therapeutic use , Brain/pathology , Drug Therapy, Combination , Ketamine/pharmacology , Midazolam/pharmacology , Midazolam/therapeutic use , Phenobarbital/pharmacology , Rats , Soman/toxicity
4.
Int J Mol Sci ; 22(4)2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33672922

ABSTRACT

The identification of improved medical countermeasures against exposure to chemical warfare nerve agents (CWNAs), a class of organophosphorus compounds, is dependent on the choice of animal model used in preclinical studies. CWNAs bind to acetylcholinesterase and prevent the catalysis of acetylcholine, causing a plethora of peripheral and central physiologic manifestations, including seizure. Rodents are widely used to elucidate the effects of CWNA-induced seizure, albeit with a caveat: they express carboxylesterase activity in plasma. Carboxylesterase, an enzyme involved in the detoxification of some organophosphorus compounds, plays a scavenging role and decreases CWNA availability, thus exerting a protective effect. Furthermore, species-specific amino acid differences in acetylcholinesterase confound studies that use oximes or other compounds to restore its function after inhibition by CWNA. The creation of a human acetylcholinesterase knock-in/serum carboxylesterase knockout (C57BL/6-Ces1ctm1.1LocAChEtm1.1Loc/J; a.k.a KIKO) mouse may facilitate better modeling of CWNA toxicity in a small rodent species. The current studies characterize the effects of exposure to soman, a highly toxic CWNA, and evaluate the efficacy of anti-seizure drugs in this newly developed KIKO mouse model. Data demonstrate that a combination of midazolam and ketamine reduces seizure duration and severity, eliminates the development of spontaneous recurrent seizures, and protects certain brain regions from neuronal damage in a genetically modified model with human relevance to organophosphorus compound toxicity. This new animal model and the results of this study and future studies using it will enhance medical countermeasures development for both defense and homeland security purposes.


Subject(s)
Acetylcholinesterase/metabolism , Carboxylesterase/metabolism , Disease Models, Animal , Medical Countermeasures , Soman/toxicity , Acetylcholinesterase/genetics , Anesthetics/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Carboxylesterase/genetics , Chemical Warfare Agents/toxicity , Humans , Ketamine/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Midazolam/pharmacology , Seizures/chemically induced , Seizures/physiopathology , Seizures/prevention & control
5.
Epilepsy Behav ; 111: 107229, 2020 10.
Article in English | MEDLINE | ID: mdl-32575012

ABSTRACT

Delayed treatment of cholinergic seizure results in benzodiazepine-refractory status epilepticus (SE) that is thought, at least in part, to result from maladaptive trafficking of N-methyl-d-aspartate (NMDA) and gamma-aminobutyric acid type A (GABAA) receptors, the effects of which may be ameliorated by combination therapy with the NMDA receptor antagonist ketamine. Our objective was to establish whether ketamine and midazolam dual therapy would improve outcome over midazolam monotherapy following soman (GD) exposure when evaluated in a mouse model that, similar to humans, lacks plasma carboxylesterase, greatly reducing endogenous scavenging of GD. In the current study, continuous cortical electroencephalographic activity was evaluated in male and female plasma carboxylesterase knockout mice exposed to a seizure-inducing dose of GD and treated with midazolam or with midazolam and ketamine combination at 40 min after seizure onset. Ketamine and midazolam combination reduced GD-induced lethality, seizure severity, and the number of mice that developed spontaneous recurrent seizure (SRS) compared with midazolam monotherapy. In addition, ketamine-midazolam combination treatment reduced GD-induced neuronal degeneration and microgliosis. These results support that combination of antiepileptic drug therapies aimed at correcting the maladaptive GABAA and NMDA receptor trafficking reduces the detrimental effects of GD exposure. Ketamine may be a beneficial adjunct to midazolam in reducing the epileptogenesis and neuroanatomical damage that follows nerve agent exposure and pharmacoresistant SE.


Subject(s)
Brain/pathology , Carboxylesterase/blood , Ketamine/administration & dosage , Midazolam/administration & dosage , Soman/toxicity , Status Epilepticus/blood , Animals , Anticonvulsants/administration & dosage , Brain/drug effects , Carboxylesterase/deficiency , Drug Therapy, Combination , Electroencephalography/methods , Female , Male , Mice , Mice, Knockout , Seizures/blood , Seizures/chemically induced , Seizures/drug therapy , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy
6.
Toxicol Sci ; 164(1): 142-152, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29596688

ABSTRACT

Elderly individuals compose a large percentage of the world population; however, few studies have addressed the efficacy of current medical countermeasures (MCMs) against the effects of chemical warfare nerve agent exposure in aged populations. We evaluated the efficacy of the anticonvulsant diazepam in an old adult rat model of soman (GD) poisoning and compared the toxic effects to those observed in young adult rats when anticonvulsant treatment is delayed. After determining their respective median lethal dose (LD50) of GD, we exposed young adult and old adult rats to an equitoxic 1.2 LD50 dose of GD followed by treatment with atropine sulfate and the oxime HI-6 at 1 min after exposure, and diazepam at 30 min after seizure onset. Old adult rats that presented with status epilepticus were more susceptible to developing spontaneous recurrent seizures (SRSs). Neuropathological analysis revealed that in rats of both age groups that developed SRS, there was a significant reduction in the density of mature neurons in the piriform cortex, thalamus, and amygdala, with more pronounced neuronal loss in the thalamus of old adult rats compared with young adult rats. Furthermore, old adult rats displayed a reduced density of cells expressing glutamic acid decarboxylase 67, a marker of GABAergic interneurons, in the basolateral amygdala and piriform cortex, and a reduction of astrocyte activation in the piriform cortex. Our observations demonstrate the reduced effectiveness of current MCM in an old adult animal model of GD exposure and strongly suggest the need for countermeasures that are more tailored to the vulnerabilities of an aging population.


Subject(s)
Aging/pathology , Anticonvulsants/therapeutic use , Chemical Warfare Agents/poisoning , Neurons/pathology , Seizures/pathology , Soman/poisoning , Animals , Anticonvulsants/administration & dosage , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Cell Count , Disease Models, Animal , Dose-Response Relationship, Drug , Electroencephalography , Lethal Dose 50 , Male , Medical Countermeasures , Neurons/drug effects , Rats, Inbred F344 , Seizures/chemically induced , Seizures/prevention & control , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Status Epilepticus/prevention & control , Telemetry , Time Factors
7.
Pharmacol Biochem Behav ; 98(1): 120-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21144858

ABSTRACT

Exposure to soman (GD) can result in prolonged seizures and subsequent neuropathology in a variety of brain regions including the amygdala and hippocampus. Both regions are believed to play important roles in the development and expression of fear conditioning. The purpose of this experiment was to test these conditioning tasks as a possible behavioral correlate of the observed neuropathology. Male rats were exposed to GD (1.0 or 1.2×LD50) or saline followed with injections of atropine sulfate, the oxime HI-6 and diazepam. Fear conditioning was conducted on post-exposure day (PED) 8 followed by measuring freezing to contextual and auditory conditioned stimuli on PED 9 and 10 respectively. Contextual and auditory fear conditioning was severely impaired in both the 1.0×LD50 and 1.2×LD50 GD groups. Both GD groups spent less time freezing than controls when returned to the context in which conditioning occurred. The 1.0×LD50 and 1.2×LD50 groups had very low levels of freezing following presentation of the auditory conditioned stimulus. Neuronal fiber degeneration was present in the piriform cortex, thalamus, and amygdala in GD-exposed animals regardless of dose. The present study suggests that contextual and auditory fear conditioning is impaired in GD-exposed rats possibly due to neuropathology observed in the hippocampus, amygdala and thalamus.


Subject(s)
Conditioning, Psychological/drug effects , Fear/drug effects , Soman/toxicity , Acoustic Stimulation , Amygdala/drug effects , Amygdala/pathology , Amygdala/physiopathology , Animals , Cholinesterase Inhibitors/toxicity , Conditioning, Psychological/physiology , Fear/physiology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Rats , Rats, Sprague-Dawley , Thalamus/drug effects , Thalamus/pathology , Thalamus/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...