1
|
Gorecki L, Pejchal J, Torruellas C, Korabecny J, Soukup O. Midazolam - A diazepam replacement for the management of nerve agent-induced seizures. Neuropharmacology 2024; 261:110171. [PMID: 39362626 DOI: 10.1016/j.neuropharm.2024.110171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/27/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
A benzodiazepine, diazepam, has been the leading antidote for seizures caused by nerve agents, the most toxic chemical weapons of mass destruction, since the 1960s. However, its limitations have often brought questions about its usefulness. Extensive effort has been devoted into exploring alternatives, such as other benzodiazepines, anticholinergics, or glutamate antagonists. However, only few showed clear clinical benefit. The only two options to ultimately reach clinical milestones are Avizafone, a water-soluble prodrug of diazepam adopted by the French and UK armed forces, and intramuscular midazolam, adopted by the US Army. The recently FDA-approved new intramuscular application of midazolam brought several advantages, such as rapid onset of action, short duration with predictable pharmacokinetics, increased water solubility for aqueous injectable solutions, and prolonged storage stability. Herein, we discuss the pitfalls and prospects of using midazolam as a substitute in anticonvulsant therapy with a particular focus on military purposes in combat casualty care. We have also considered and discussed several other alternatives that are currently at the experimental level. Recent studies have shown the superiority of midazolam over other benzodiazepines in the medical management of poisoned casualties. While its use in emergency care is straightforward, the proper dose for soldiers under battlefield conditions is questionable due to its sedative effects.
Collapse
Affiliation(s)
- Lukas Gorecki
- University of Defence, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; University Hospital Hradec Kralove, Biomedical Research Centre, Sokolska 581, 500 05, Hradec Kralove, Czech Republic.
| | - Jaroslav Pejchal
- University of Defence, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Carilyn Torruellas
- U. S. Army CCDC Chemical Biological Center, Aberdeen Proving Ground, MD, 21010-5424, USA
| | - Jan Korabecny
- University of Defence, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; University Hospital Hradec Kralove, Biomedical Research Centre, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- University of Defence, Military Faculty of Medicine, Department of Toxicology and Military Pharmacy, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; University Hospital Hradec Kralove, Biomedical Research Centre, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| |
Collapse
|
2
|
Cornelissen AS, van den Berg RM, Klaassen SD, de Koning JC, Langenberg JP, de Lange ECM, Joosen MJA. Synergistic polytherapy for the broad-spectrum treatment of chemically-induced seizures in rats. Toxicol Appl Pharmacol 2024; 493:117137. [PMID: 39476875 DOI: 10.1016/j.taap.2024.117137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/12/2024] [Accepted: 10/27/2024] [Indexed: 11/11/2024]
Abstract
Chemically-induced seizures, as a result of exposure to a neurotoxic compound, present a serious health concern. Compounds can elicit seizure activity through disruption of neuronal signaling by neurotransmitters, either by mimicking, modulating or antagonizing their action at the receptor or interfering with their metabolism. Benzodiazepines, such as diazepam and midazolam, and barbiturates are the mainstay of treatment of seizures. However, chemically-induced seizures are often persistent, requiring repeated treatment and increased doses of anticonvulsants, which in turn may lead to severe adverse effects such as respiratory depression. Here, we investigated the potential of rational polytherapy consisting of the benzodiazepine midazolam and the selective α2-adrenergic agonist dexmedetomidine as an improved, generically applicable anticonvulsant treatment regimen. Therapeutic efficacy was evaluated against two experimental paradigm compounds that induce persistent seizures in rats, the rodenticide TETS and the nerve agent soman. Following exposure, both TETS and soman elicited profound seizure activity and convulsions, associated with substantial mortality. Treatment with midazolam or dexmedetomidine alone provided no or limited suppression of seizure activity and improvement of survival at 4 h. Polytherapy consisting of midazolam and dexmedetomidine showed excellent anticonvulsant efficacy. Even at low doses, polytherapy showed a profound effect that lasted for the duration of the experiment. Analysis of the dose-response relationships confirmed presence of synergy. Administration of polytherapy in non-exposed animals did not indicate aggravation of adverse effects on respiration or heart rate. Even though more research is needed for the translation to clinical use, polytherapy consisting of midazolam and dexmedetomidine shows promise for the broad-spectrum treatment of (chemically-induced) seizures in emergency situations.
Collapse
Affiliation(s)
- Alex S Cornelissen
- Department of CBRN Protection, TNO Defence, Safety and Security, Lange Kleiweg 137, 2288 GJ Rijswijk, the Netherlands; Predictive Pharmacology group, Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Gorlaeus Laboratory, Einsteinweg 55, 2333 CC Leiden, the Netherlands.
| | - Roland M van den Berg
- Department of CBRN Protection, TNO Defence, Safety and Security, Lange Kleiweg 137, 2288 GJ Rijswijk, the Netherlands
| | - Steven D Klaassen
- Department of CBRN Protection, TNO Defence, Safety and Security, Lange Kleiweg 137, 2288 GJ Rijswijk, the Netherlands
| | - Jelle C de Koning
- Department of CBRN Protection, TNO Defence, Safety and Security, Lange Kleiweg 137, 2288 GJ Rijswijk, the Netherlands
| | - Jan P Langenberg
- Department of CBRN Protection, TNO Defence, Safety and Security, Lange Kleiweg 137, 2288 GJ Rijswijk, the Netherlands
| | - Elizabeth C M de Lange
- Predictive Pharmacology group, Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Gorlaeus Laboratory, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | - Marloes J A Joosen
- Department of CBRN Protection, TNO Defence, Safety and Security, Lange Kleiweg 137, 2288 GJ Rijswijk, the Netherlands
| |
Collapse
|
3
|
Aroniadou-Anderjaska V, Figueiredo TH, De Araujo Furtado M, Pidoplichko VI, Lumley LA, Braga MFM. Alterations in GABA A receptor-mediated inhibition triggered by status epilepticus and their role in epileptogenesis and increased anxiety. Neurobiol Dis 2024; 200:106633. [PMID: 39117119 DOI: 10.1016/j.nbd.2024.106633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
The triggers of status epilepticus (SE) in non-epileptic patients can vary widely, from idiopathic causes to exposure to chemoconvulsants. Regardless of its etiology, prolonged SE can cause significant brain damage, commonly resulting in the development of epilepsy, which is often accompanied by increased anxiety. GABAA receptor (GABAAR)-mediated inhibition has a central role among the mechanisms underlying brain damage and the ensuing epilepsy and anxiety. During SE, calcium influx primarily via ionotropic glutamate receptors activates signaling cascades which trigger a rapid internalization of synaptic GABAARs; this weakens inhibition, exacerbating seizures and excitotoxicity. GABAergic interneurons are more susceptible to excitotoxic death than principal neurons. During the latent period of epileptogenesis, the aberrant reorganization in synaptic interactions that follow interneuronal loss in injured brain regions, leads to the formation of hyperexcitable, seizurogenic neuronal circuits, along with disturbances in brain oscillatory rhythms. Reduction in the spontaneous, rhythmic "bursts" of IPSCs in basolateral amygdala neurons is likely to play a central role in anxiogenesis. Protecting interneurons during SE is key to preventing both epilepsy and anxiety. Antiglutamatergic treatments, including antagonism of calcium-permeable AMPA receptors, can be expected to control seizures and reduce excitotoxicity not only by directly suppressing hyperexcitation, but also by counteracting the internalization of synaptic GABAARs. Benzodiazepines, as delayed treatment of SE, have low efficacy due to the reduction and dispersion of their targets (the synaptic GABAARs), but also because themselves contribute to further reduction of available GABAARs at the synapse; furthermore, benzodiazepines may be completely ineffective in the immature brain.
Collapse
Affiliation(s)
- Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Marcio De Araujo Furtado
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Volodymyr I Pidoplichko
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Lucille A Lumley
- U.S. Army Medical Research Institute of Chemical Defense, Aberdeen, Proving Ground, MD, USA.
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
4
|
Shih TM, Munoz C, Acon-Chen C, Keith ZM. Pharmacology of Adenosine A 1 Receptor Agonist in a Humanized Esterase Mouse Seizure Model Following Soman Intoxication. Neurotox Res 2024; 42:41. [PMID: 39230655 PMCID: PMC11374867 DOI: 10.1007/s12640-024-00717-z] [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: 01/14/2024] [Revised: 06/14/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Recently a novel genetically modified mouse strain with serum carboxylesterase knocked-out and the human acetylcholinesterase gene knocked-in (KIKO) was created to simulate human responses to nerve agent (NA) exposure and its standard medical treatment. A1 adenosine receptor (A1AR) agonist N-bicyclo-(2.2.1)-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA) alone is a potent anticonvulsant and neuroprotectant (A/N) in both rat and KIKO mouse soman (GD) seizure models. In this study we utilized the KIKO mouse to evaluate further the basic pharmacologic A/N effects of ENBA as an adjunct to standard NA medical treatments (i.e., atropine sulfate, pralidoxime chloride [2-PAM], and midazolam). Male mice, implanted with cortical electroencephalographic (EEG) electrodes, were pretreated with asoxime (HI-6) and exposed to an epileptogenic dose of GD (33 µg/kg, s.c.) or saline (sham exposure) and then treated 15 min after seizure onset with ENBA at 15 mg/kg, i.p. (a minimum efficacy dose in suppressing NA-induced seizure) alone or as an adjunct to standard medical treatments. We collected EEG activity, seizure suppression outcomes, daily body temperature and weight, heart rate, toxic signs, neuropathology, and lethality data for up to 14 days. Without ENBA, death from NA exposure was 45%, while with ENBA, either alone or in combination with midazolam, the survival improved to 80% and 90%, respectively. Additionally, seizure was suppressed quickly and permanently, toxic signs, hypothermia, and bradycardia recovered by 48 h, and no neuropathology was evident. Our findings confirmed that ENBA is a potent A/N adjunct for delayed medical treatments of NA exposure.
Collapse
Affiliation(s)
- Tsung-Ming Shih
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Crystal Munoz
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9162, USA
| | - Cindy Acon-Chen
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
| | - Zora-Maya Keith
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
| |
Collapse
|
5
|
Harkins J, Langston J, Keith ZM, Munoz C, Acon-Chen C, Shih TM. Learning and memory function preserved by delayed A 1 adenosine receptor agonist treatment following soman intoxication in rats and a humanized esterase mouse model. Neuropharmacology 2024; 253:109983. [PMID: 38704023 PMCID: PMC11132435 DOI: 10.1016/j.neuropharm.2024.109983] [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: 01/09/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
Exposure to organophosphorus compounds, such as soman (GD), cause widespread toxic effects, sustained status epilepticus, neuropathology, and death. The A1 adenosine receptor agonist N-bicyclo-(2.2.1)-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), when given 1 min after GD exposure, provides neuroprotection and prevents behavioral impairments. Here, we tested the ability of ENBA at delayed treatment times to improve behavioral outcomes via a two-way active avoidance task in two male animal models, each consisting of saline and GD exposure groups. In a rat model, animals received medical treatments (atropine sulfate [A], 2-PAM [P], and midazolam [MDZ]) or AP + MDZ + ENBA at 15 or 30 min after seizure onset and were subjected to behavioral testing for up to 14 days. In a human acetylcholinesterase knock-in serum carboxylesterase knock-out mouse model, animals received AP, AP + MDZ, AP + ENBA, or AP + MDZ + ENBA at 15 min post seizure onset and were subjected to the behavioral task on days 7 and 14. In rats, the GD/AP + MDZ + ENBA group recovered to saline-exposed avoidance levels while the GD/AP + MDZ group did not. In mice, in comparison with GD/AP + MDZ group, the GD/AP + MDZ + ENBA showed decreases in escape latency, response latency, and pre-session crossings, as well as increases in avoidances. In both models, only ENBA-treated groups showed control level inter-trial interval crossings by day 14. Our findings suggest that ENBA, alone and as an adjunct to medical treatments, can improve behavioral and cognitive outcomes when given at delayed time points after GD intoxication.
Collapse
Affiliation(s)
- Joshua Harkins
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Jeffrey Langston
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Zora-Maya Keith
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Crystal Munoz
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Cindy Acon-Chen
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Tsung-Ming Shih
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| |
Collapse
|
6
|
Keith ZM, Munoz C, Acon-Chen C, Shih TM. Seizure suppression and neuroprotection in soman-exposed rats following delayed intramuscular treatment of adenosine A 1 receptor agonist as an adjunct to standard medical treatment. Toxicol Appl Pharmacol 2024; 488:116970. [PMID: 38777098 DOI: 10.1016/j.taap.2024.116970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/01/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024]
Abstract
Soman produces excitotoxic effects by inhibiting acetylcholinesterase in the cholinergic synapses and neuromuscular junctions, resulting in soman-induced sustained status epilepticus (SSE). Our previous work showed delayed intramuscular (i.m.) treatment with A1 adenosine receptor agonist N-bicyclo-[2.2.1]-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA) alone suppressed soman-induced SSE and prevented neuropathology. Using this same rat soman seizure model, we tested if delayed therapy with ENBA (60 mg/kg, i.m.) would terminate seizure, protect neuropathology, and aid in survival when given in conjunction with current standard medical countermeasures (MCMs): atropine sulfate, 2-PAM, and midazolam (MDZ). Either 15- or 30-min following soman-induced SSE onset, male rats received atropine and 2-PAM plus either MDZ or MDZ + ENBA. Electroencephalographic (EEG) activity, physiologic parameters, and motor function were recorded. Either 2- or 14-days following exposure surviving rats were euthanized and perfused for histology. All animals treated with MDZ + ENBA at both time points had 100% EEG seizure termination and reduced total neuropathology compared to animals treated with MDZ (2-day, p = 0.015 for 15-min, p = 0.002 for 30-min; 14-day, p < 0.001 for 15-min, p = 0.006 for 30-min), showing ENBA enhanced MDZ's anticonvulsant and neuroprotectant efficacy. However, combined MDZ + ENBA treatment, when compared to MDZ treatment groups, had a reduction in the 14-day survival rate regardless of treatment time, indicating possible enhancement of MDZ's neuronal inhibitory effects by ENBA. Based on our findings, ENBA shows promise as an anticonvulsant and neuroprotectant in a combined treatment regimen following soman exposure; when given as an adjunct to standard MCMs, the dose of ENBA needs to be adjusted.
Collapse
Affiliation(s)
- Zora-Maya Keith
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA.
| | - Crystal Munoz
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA.
| | - Cindy Acon-Chen
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA.
| | - Tsung-Ming Shih
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA.
| |
Collapse
|
7
|
Hobson BA, Rowland DJ, Dou Y, Saito N, Harmany ZT, Bruun DA, Harvey DJ, Chaudhari AJ, Garbow JR, Lein PJ. A longitudinal MRI and TSPO PET-based investigation of brain region-specific neuroprotection by diazepam versus midazolam following organophosphate-induced seizures. Neuropharmacology 2024; 251:109918. [PMID: 38527652 PMCID: PMC11250911 DOI: 10.1016/j.neuropharm.2024.109918] [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: 11/29/2023] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/27/2024]
Abstract
Acute poisoning with organophosphorus cholinesterase inhibitors (OPs), such as OP nerve agents and pesticides, can cause life threatening cholinergic crisis and status epilepticus (SE). Survivors often experience significant morbidity, including brain injury, acquired epilepsy, and cognitive deficits. Current medical countermeasures for acute OP poisoning include a benzodiazepine to mitigate seizures. Diazepam was long the benzodiazepine included in autoinjectors used to treat OP-induced seizures, but it is now being replaced in many guidelines by midazolam, which terminates seizures more quickly, particularly when administered intramuscularly. While a direct correlation between seizure duration and the extent of brain injury has been widely reported, there are limited data comparing the neuroprotective efficacy of diazepam versus midazolam following acute OP intoxication. To address this data gap, we used non-invasive imaging techniques to longitudinally quantify neuropathology in a rat model of acute intoxication with the OP diisopropylfluorophosphate (DFP) with and without post-exposure intervention with diazepam or midazolam. Magnetic resonance imaging (MRI) was used to monitor neuropathology and brain atrophy, while positron emission tomography (PET) with a radiotracer targeting translocator protein (TSPO) was utilized to assess neuroinflammation. Animals were scanned at 3, 7, 28, 65, 91, and 168 days post-DFP and imaging metrics were quantitated for the hippocampus, amygdala, piriform cortex, thalamus, cerebral cortex and lateral ventricles. In the DFP-intoxicated rat, neuroinflammation persisted for the duration of the study coincident with progressive atrophy and ongoing tissue remodeling. Benzodiazepines attenuated neuropathology in a region-dependent manner, but neither benzodiazepine was effective in attenuating long-term neuroinflammation as detected by TSPO PET. Diffusion MRI and TSPO PET metrics were highly correlated with seizure severity, and early MRI and PET metrics were positively correlated with long-term brain atrophy. Collectively, these results suggest that anti-seizure therapy alone is insufficient to prevent long-lasting neuroinflammation and tissue remodeling.
Collapse
Affiliation(s)
- Brad A Hobson
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA; Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA.
| | - Douglas J Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA.
| | - Yimeng Dou
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA.
| | - Naomi Saito
- Department of Public Health Sciences, University of California, Davis, School of Medicine, California 95616, USA.
| | - Zachary T Harmany
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA.
| | - Donald A Bruun
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA.
| | - Danielle J Harvey
- Department of Public Health Sciences, University of California, Davis, School of Medicine, California 95616, USA.
| | - Abhijit J Chaudhari
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA; Department of Radiology, University of California, Davis, School of Medicine, California 95817, USA.
| | - Joel R Garbow
- Biomedical Magnetic Resonance Center, Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, 63110, USA.
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA.
| |
Collapse
|
8
|
Lazar S, Neufeld-Cohen A, Egoz I, Baranes S, Gez R, Glick P, Cohen M, Gutman H, Chapman S, Gore A. Efficacy of a combined anti-seizure treatment against cholinergic established status epilepticus following a sarin nerve agent insult in rats. Toxicol Appl Pharmacol 2024; 484:116870. [PMID: 38395364 DOI: 10.1016/j.taap.2024.116870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/11/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
The development of refractory status epilepticus (SE) following sarin intoxication presents a therapeutic challenge. Here, we evaluated the efficacy of delayed combined double or triple treatment in reducing abnormal epileptiform seizure activity (ESA) and the ensuing long-term neuronal insult. SE was induced in rats by exposure to 1.2 LD50 sarin followed by treatment with atropine and TMB4 (TA) 1 min later. Double treatment with ketamine and midazolam or triple treatment with ketamine, midazolam and levetiracetam was administered 30 min post-exposure, and the results were compared to those of single treatment with midazolam alone or triple treatment with ketamine, midazolam, and valproate, which was previously shown to ameliorate this neurological insult. Toxicity and electrocorticogram activity were monitored during the first week, and behavioral evaluations were performed 2 weeks post-exposure, followed by biochemical and immunohistopathological analyses. Both double and triple treatment reduced mortality and enhanced weight recovery compared to TA-only treatment. Triple treatment and, to a lesser extent, double treatment significantly ameliorated the ESA duration. Compared to the TA-only or the TA+ midazolam treatment, both double and triple treatment reduced the sarin-induced increase in the neuroinflammatory marker PGE2 and the brain damage marker TSPO and decreased gliosis, astrocytosis and neuronal damage. Finally, both double and triple treatment prevented a change in behavior, as measured in the open field test. No significant difference was observed between the efficacies of the two triple treatments, and both triple combinations completely prevented brain injury (no differences from the naïve rats). Delayed double and, to a greater extent, triple treatment may serve as an efficacious delayed therapy, preventing brain insult propagation following sarin-induced refractory SE.
Collapse
Affiliation(s)
- Shlomi Lazar
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel.
| | - Adi Neufeld-Cohen
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Inbal Egoz
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Shlomi Baranes
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Rellie Gez
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Pnina Glick
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Maayan Cohen
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Hila Gutman
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Shira Chapman
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel
| | - Ariel Gore
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona 74100, Israel.
| |
Collapse
|
9
|
Reddy DS. Neurosteroids as Novel Anticonvulsants for Refractory Status Epilepticus and Medical Countermeasures for Nerve Agents: A 15-Year Journey to Bring Ganaxolone from Bench to Clinic. J Pharmacol Exp Ther 2024; 388:273-300. [PMID: 37977814 PMCID: PMC10801762 DOI: 10.1124/jpet.123.001816] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/05/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023] Open
Abstract
This article describes recent advances in the use of neurosteroids as novel anticonvulsants for refractory status epilepticus (RSE) and as medical countermeasures (MCs) for organophosphates and chemical nerve agents (OPNAs). We highlight a comprehensive 15-year journey to bring the synthetic neurosteroid ganaxolone (GX) from bench to clinic. RSE, including when caused by nerve agents, is associated with devastating morbidity and permanent long-term neurologic dysfunction. Although recent approval of benzodiazepines such as intranasal midazolam and intranasal midazolam offers improved control of acute seizures, novel anticonvulsants are needed to suppress RSE and improve neurologic function outcomes. Currently, few anticonvulsant MCs exist for victims of OPNA exposure and RSE. Standard-of-care MCs for postexposure treatment include benzodiazepines, which do not effectively prevent or mitigate seizures resulting from nerve agent intoxication, leaving an urgent unmet medical need for new anticonvulsants for RSE. Recently, we pioneered neurosteroids as next-generation anticonvulsants that are superior to benzodiazepines for treatment of OPNA intoxication and RSE. Because GX and related neurosteroids that activate extrasynaptic GABA-A receptors rapidly control seizures and offer robust neuroprotection by reducing neuronal damage and neuroinflammation, they effectively improve neurologic outcomes after acute OPNA exposure and RSE. GX has been selected for advanced, Biomedical Advanced Research and Development Authority-supported phase 3 trials of RSE and nerve agent seizures. In addition, in mechanistic studies of neurosteroids at extrasynaptic receptors, we identified novel synthetic analogs with features that are superior to GX for current medical needs. Development of new MCs for RSE is complex, tedious, and uncertain due to scientific and regulatory challenges. Thus, further research will be critical to fill key gaps in evaluating RSE and anticonvulsants in vulnerable (pediatric and geriatric) populations and military persons. SIGNIFICANCE STATEMENT: Following organophosphate and nerve agent intoxication, refractory status epilepticus (RSE) occurs despite benzodiazepine treatment. RSE occurs in 40% of status epilepticus patients, with a 35% mortality rate and significant neurological morbidity in survivors. To treat RSE, neurosteroids are better anticonvulsants than benzodiazepines. Our pioneering use of neurosteroids for RSE and nerve agents led us to develop ganaxolone as a novel anticonvulsant and neuroprotectant with significantly improved neurological outcomes. This article describes the bench-to-bedside journey of bringing neurosteroid therapy to patients, with ganaxolone leading the way.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
10
|
Singh T, Ramakrishnan S, Wu X, Reddy DS. Sex Differences in Organophosphate Model of Benzodiazepine-Refractory Status Epilepticus and Neuronal Damage. J Pharmacol Exp Ther 2024; 388:313-324. [PMID: 37770202 PMCID: PMC10801723 DOI: 10.1124/jpet.123.001747] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 10/03/2023] Open
Abstract
Sex differences are common in human epilepsy. Although men are more susceptible to seizure than women, the mechanisms underlying sex-specific vulnerabilities to seizure are unclear. The organophosphate (OP) diisopropylfluorophosphate (DFP) is known to cause neurotoxicity and status epilepticus (SE), a serious neurologic condition that causes prolonged seizures and brain damage. Current therapies for OP poisoning and SE do not consider neuronal variations between male and female brains. Therefore, we investigated sex-dependent differences in electrographic seizure activity and neuronal injury using the DFP model of refractory SE in rats. Electroencephalogram recordings were used to monitor DFP-induced SE, and the extent of brain injury was determined using fluoro-jade-B staining to detect cellular necrosis. After DFP exposure, we observed striking sex-dependent differences in SE and seizure activity patterns as well as protective responses to midazolam treatment. Following acute DFP exposure, male animals displayed more severe SE with intense epileptiform spiking and greater mortality than females. In contrast, we observed significantly more injured cells and cellular necrosis in the hippocampus and other brain regions in females than in males. We also observed extensive neuronal injury in the somatosensory cortex of males. The anticonvulsant effect of midazolam against SE was limited in this model and found to be similar in males and females. However, unlike males, females exhibited substantially more protection against neuronal damage after midazolam treatment. Overall, these results demonstrate significant sex-dependent differences in DFP-induced refractory SE and neuronal damage patterns, suggesting that it may be possible to develop sex-specific neuroprotective strategies for OP intoxication and refractory SE. SIGNIFICANCE STATEMENT: Sex-dependent differences in neurotoxicity and status epilepticus (SE) are key biological variables after organophosphate (OP) exposure. Here, we investigated sex-dependent differences in SE and brain injury after acute diisopropylfluorophosphate exposure. Male rats had more severe SE and less survival than females, while females had more neuronal damage. Females had more neuroprotection to midazolam than males, while both sexes had similar but partial anticonvulsant effects. These findings suggest that a sex-specific therapeutic approach may prevent neurological complications of OP-induced SE.
Collapse
Affiliation(s)
- Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics and Institute of Pharmacology and Neurotherapeutics, Texas A&M University School of Medicine, Bryan, Texas
| |
Collapse
|
11
|
Ramakrishnan S, Singh T, Reddy DS. Protective Activity of Novel Hydrophilic Synthetic Neurosteroids on Organophosphate Status Epilepticus-induced Chronic Epileptic Seizures, Non-Convulsive Discharges, High-Frequency Oscillations, and Electrographic Ictal Biomarkers. J Pharmacol Exp Ther 2024; 388:386-398. [PMID: 38050069 PMCID: PMC10801763 DOI: 10.1124/jpet.123.001817] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/06/2023] Open
Abstract
Nerve agents and organophosphates (OP) are neurotoxic chemicals that induce acute seizures, status epilepticus (SE), and mortality. Long-term neurologic and neurodegenerative effects manifest months to years after OP exposure. Current benzodiazepine anticonvulsants are ineffective in preventing such long-term neurobehavioral and neuropathological changes. New and effective anticonvulsants are needed for OP intoxication, especially for mitigating the long-term sequelae after acute exposure. We developed neurosteroids as novel anticonvulsants and neuroprotectants in OP exposure models. In this study, we evaluated the long-term efficacy of novel synthetic neurosteroids in preventing the development of chronic epilepsy and hyperexcitable ictal events in a rat OP model of SE. Rats were exposed to the OP nerve agent surrogate diisopropylfluorophosphate (DFP), and the experimental groups were treated with the synthetic neurosteroid valaxanolone (VX) or lysaxanolone (LX) 40 minutes post-exposure in conjunction with midazolam. Video-electroencephalography was monitored for two months to assess spontaneous recurrent seizures (SRS), epileptiform discharges, interictal spikes, and high-frequency oscillations (HFOs). Within 60 days of DFP exposure, rats developed chronic epilepsy characterized by frequent SRS, epileptiform discharges, and HFOs. LX treatment was associated with a dose-dependent reduction of epilepsy occurrence and overall seizure burden with a significant decrease in SRS and epileptiform discharges. It also significantly reduced the occurrence of epileptic biomarkers of HFOs and interictal spikes, indicating potential disease-modifying activity. Similarly, the neurosteroid analog VX also significantly attenuated SRS, discharges, HFOs, and ictal events. These results demonstrate the long-term protective effects of synthetic neurosteroids in the OP-exposed post-SE model, indicating their disease-modifying potential to prevent epilepsy and ictal abnormalities. SIGNIFICANCE STATEMENT: The effects of nerve agents and organophosphate (OP) exposure are persistent, and survivors suffer from a number of devastating, chronic neurological dysfunctions. Currently, there is no specific therapy for preventing this disastrous impact of OP exposure. We propose synthetic neurosteroids that activate tonic inhibition provide viable options for preventing the long-term neurological effects of OP intoxication. The results from this study reveal the disease-modifying potential of two novel synthetic neurosteroids in preventing epileptogenesis and chronic epileptic seizures after OP-induced SE.
Collapse
Affiliation(s)
- Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics (S.R., T.S., D.S.R.) and Institute of Pharmacology and Neurotherapeutics (D.S.R.), School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics (S.R., T.S., D.S.R.) and Institute of Pharmacology and Neurotherapeutics (D.S.R.), School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics (S.R., T.S., D.S.R.) and Institute of Pharmacology and Neurotherapeutics (D.S.R.), School of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
12
|
Puthillathu N, Moffett JR, Korotcov A, Bosomtwi A, Vengilote R, Krishnan JKS, Johnson EA, Arun P, Namboodiri AM. Brief isoflurane administration as an adjunct treatment to control organophosphate-induced convulsions and neuropathology. Front Pharmacol 2023; 14:1293280. [PMID: 38230376 PMCID: PMC10790757 DOI: 10.3389/fphar.2023.1293280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/21/2023] [Indexed: 01/18/2024] Open
Abstract
Organophosphate-based chemical agents (OP), including nerve agents and certain pesticides such as paraoxon, are potent acetylcholinesterase inhibitors that cause severe convulsions and seizures, leading to permanent central nervous system (CNS) damage if not treated promptly. The current treatment regimen for OP poisoning is intramuscular injection of atropine sulfate with an oxime such as pralidoxime (2-PAM) to mitigate cholinergic over-activation of the somatic musculature and autonomic nervous system. This treatment does not provide protection against CNS cholinergic overactivation and therefore convulsions require additional medication. Benzodiazepines are the currently accepted treatment for OP-induced convulsions, but the convulsions become refractory to these GABAA agonists and repeated dosing has diminishing effectiveness. As such, adjunct anticonvulsant treatments are needed to provide improved protection against recurrent and prolonged convulsions and the associated excitotoxic CNS damage that results from them. Previously we have shown that brief, 4-min administration of 3%-5% isoflurane in 100% oxygen has profound anticonvulsant and CNS protective effects when administered 30 min after a lethal dose of paraoxon. In this report we provide an extended time course of the effectiveness of 5% isoflurane delivered for 5 min, ranging from 60 to 180 min after a lethal dose of paraoxon in rats. We observed substantial effectiveness in preventing neuronal loss as shown by Fluoro-Jade B staining when isoflurane was administered 1 h after paraoxon, with diminishing effectiveness at 90, 120 and 180 min. In vivo magnetic resonance imaging (MRI) derived T2 and mean diffusivity (MD) values showed that 5-min isoflurane administration at a concentration of 5% prevents brain edema and tissue damage when administered 1 h after a lethal dose of paraoxon. We also observed reduced astrogliosis as shown by GFAP immunohistochemistry. Studies with continuous EEG monitoring are ongoing to demonstrate effectiveness in animal models of soman poisoning.
Collapse
Affiliation(s)
- Narayanan Puthillathu
- Department of Anatomy, Physiology, and Genetics, Neuroscience Program and Molecular and Cell Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - John R. Moffett
- Department of Anatomy, Physiology, and Genetics, Neuroscience Program and Molecular and Cell Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Alexandru Korotcov
- Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, United States
| | - Asamoah Bosomtwi
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, United States
| | - Ranjini Vengilote
- Department of Anatomy, Physiology, and Genetics, Neuroscience Program and Molecular and Cell Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jishnu K. S. Krishnan
- Department of Anatomy, Physiology, and Genetics, Neuroscience Program and Molecular and Cell Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Erik A. Johnson
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, Gunpowder, MD, United States
| | - Peethambaran Arun
- Department of Anatomy, Physiology, and Genetics, Neuroscience Program and Molecular and Cell Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Aryan M. Namboodiri
- Department of Anatomy, Physiology, and Genetics, Neuroscience Program and Molecular and Cell Biology Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
13
|
Shih TM. A novel genetically modified mouse seizure model for evaluating anticonvulsive and neuroprotective efficacy of an A 1 adenosine receptor agonist following soman intoxication. Toxicol Appl Pharmacol 2023; 464:116437. [PMID: 36849019 PMCID: PMC10228141 DOI: 10.1016/j.taap.2023.116437] [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: 07/04/2022] [Revised: 01/27/2023] [Accepted: 02/19/2023] [Indexed: 02/27/2023]
Abstract
Recently a novel humanized mouse strain has been successfully generated, in which serum carboxylesterase (CES) knock out (KO) mice (Es1-/-) were further genetically modified by knocking in (KI), or adding, the gene that encodes the human form of acetylcholinesterase (AChE). The resulting human AChE KI and serum CES KO (or KIKO) mouse strain should not only exhibit organophosphorus nerve agent (NA) intoxication in a manner more similar to humans, but also display AChE-specific treatment responses more closely mimicking those of humans to facilitate data translation to pre-clinic trials. In this study, we utilized the KIKO mouse to develop a seizure model for NA medical countermeasure investigation, and then applied it to evaluate the anticonvulsant and neuroprotectant (A/N) efficacy of a specific A1 adenosine receptor (A1AR) agonist, N-bicyclo-(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), which has been shown in a rat seizure model to be a potent A/N compound. Male mice surgically implanted with cortical electroencephalographic (EEG) electrodes a week earlier were pretreated with HI-6 and challenged with various doses (26 to 47 μg/kg, SC) of soman (GD) to determine a minimum effective dose (MED) that induced sustained status epilepticus (SSE) activity in 100% of animals while causing minimum lethality at 24 h. The GD dose selected was then used to investigate the MED doses of ENBA when given either immediately following SSE initiation (similar to wartime military first aid application) or at 15 min after ongoing SSE seizure activity (applicable to civilian chemical attack emergency triage). The selected GD dose of 33 μg/kg (1.4 x LD50) generated SSE in 100% of KIKO mice and produced only 30% mortality. ENBA at a dose as little as 10 mg/kg, IP, caused isoelectric EEG activity within minutes after administration in naïve un-exposed KIKO mice. The MED doses of ENBA to terminate GD-induced SSE activity were determined to be 10 and 15 mg/kg when treatment was given at the time of SSE onset and when seizure activity was ongoing for 15 min, respectively. These doses were much lower than in the non-genetically modified rat model, which required an ENBA dose of 60 mg/kg to terminate SSE in 100% GD-exposed rats. At MED doses, all mice survived for 24 h, and no neuropathology was observed when the SSE was stopped. The findings confirmed that ENBA is a potent A/N for both immediate and delayed (i.e., dual purposed) therapy to victims of NA exposure and serves as a promising neuroprotective antidotal and adjunctive medical countermeasure candidate for pre-clinical research and development for human application.
Collapse
Affiliation(s)
- Tsung-Ming Shih
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen, Proving Ground, MD 21010-5400, USA..
| |
Collapse
|
14
|
Massey N, Vasanthi SS, Samidurai M, Gage M, Rao N, Meyer C, Thippeswamy T. 1400 W, a selective inducible nitric oxide synthase inhibitor, mitigates early neuroinflammation and nitrooxidative stress in diisopropylfluorophosphate-induced short-term neurotoxicity rat model. Front Mol Neurosci 2023; 16:1125934. [PMID: 37008784 PMCID: PMC10064070 DOI: 10.3389/fnmol.2023.1125934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
Organophosphate nerve agent (OPNA) exposure induces acute and long-term neurological deficits. OPNA exposure at sub-lethal concentrations induces irreversible inhibition of acetylcholinesterase and cholinergic toxidrome and develops status epilepticus (SE). Persistent seizures have been associated with increased production of ROS/RNS, neuroinflammation, and neurodegeneration. A total of 1400W is a novel small molecule, which irreversibly inhibits inducible nitric oxide synthase (iNOS) and has been shown to effectively reduce ROS/RNS generation. In this study, we investigated the effects of 1400W treatment for a week or two weeks at 10 mg/kg or 15 mg/kg per day in the rat diisopropylfluorophosphate (DFP) model. 1400W significantly reduced the number of microglia, astroglia, and NeuN+FJB positive cells compared to the vehicle in different regions of the brain. 1400W also significantly reduced nitrooxidative stress markers and proinflammatory cytokines in the serum. However, neither of the two concentrations of 1400W for two weeks of treatment had any significant effect on epileptiform spike rate and spontaneous seizures during the treatment period in mixed sex cohorts, males, or females. No significant sex differences were found in response to DFP exposure or 1400W treatment. In conclusion, 1400W treatment at 15 mg/kg per day for two weeks was more effective in significantly reducing DFP-induced nitrooxidative stress, neuroinflammatory and neurodegenerative changes.
Collapse
|
15
|
Figueiredo TH, Aroniadou-Anderjaska V, Pidoplichko VI, Apland JP, Braga MFM. Antiseizure and Neuroprotective Efficacy of Midazolam in Comparison with Tezampanel (LY293558) against Soman-Induced Status Epilepticus. TOXICS 2022; 10:409. [PMID: 35893842 PMCID: PMC9330837 DOI: 10.3390/toxics10080409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 12/12/2022]
Abstract
Acute exposure to nerve agents induces status epilepticus (SE), which can cause death or long-term brain damage. Diazepam is approved by the FDA for the treatment of nerve agent-induced SE, and midazolam (MDZ) is currently under consideration to replace diazepam. However, animal studies have raised questions about the neuroprotective efficacy of benzodiazepines. Here, we compared the antiseizure and neuroprotective efficacy of MDZ (5 mg/kg) with that of tezampanel (LY293558; 10 mg/kg), an AMPA/GluK1 receptor antagonist, administered 1 h after injection of the nerve agent, soman (1.2 × LD50), in adult male rats. Both of the anticonvulsants promptly stopped SE, with MDZ having a more rapid effect. However, SE reoccurred to a greater extent in the MDZ-treated group, resulting in a significantly longer total duration of SE within 24 h post-exposure compared with the LY293558-treated group. The neuroprotective efficacy of the two drugs was studied in the basolateral amygdala, 30 days post-exposure. Significant neuronal and inter-neuronal loss, reduced ratio of interneurons to the total number of neurons, and reduction in spontaneous inhibitory postsynaptic currents accompanied by increased anxiety were found in the MDZ-treated group. The rats treated with LY293558 did not differ from the control rats (not exposed to soman) in any of these measurements. Thus, LY293558 has significantly greater efficacy than midazolam in protecting against prolonged seizures and brain damage caused by acute nerve agent exposure.
Collapse
Affiliation(s)
- Taiza H. Figueiredo
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.H.F.); (V.A.-A.); (V.I.P.)
| | - Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.H.F.); (V.A.-A.); (V.I.P.)
- Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Volodymyr I. Pidoplichko
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.H.F.); (V.A.-A.); (V.I.P.)
| | - James P. Apland
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Aberdeen, MD 21010, USA;
| | - Maria F. M. Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.H.F.); (V.A.-A.); (V.I.P.)
- Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
16
|
Morgan JE, Wilson SC, Travis BJ, Bagri KH, Pagarigan KT, Belski HM, Jackson C, Bounader KM, Coppola JM, Hornung EN, Johnson JE, McCarren HS. Refractory and Super-Refractory Status Epilepticus in Nerve Agent-Poisoned Rats Following Application of Standard Clinical Treatment Guidelines. Front Neurosci 2021; 15:732213. [PMID: 34566572 PMCID: PMC8462486 DOI: 10.3389/fnins.2021.732213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
Nerve agents (NAs) induce a severe cholinergic crisis that can lead to status epilepticus (SE). Current guidelines for treatment of NA-induced SE only include prehospital benzodiazepines, which may not fully resolve this life-threatening condition. This study examined the efficacy of general clinical protocols for treatment of SE in the specific context of NA poisoning in adult male rats. Treatment with both intramuscular and intravenous benzodiazepines was entirely insufficient to control SE. Second line intervention with valproate (VPA) initially terminated SE in 35% of rats, but seizures always returned. Phenobarbital (PHB) was more effective, with SE terminating in 56% of rats and 19% of rats remaining seizure-free for at least 24 h. The majority of rats demonstrated refractory SE (RSE) and required treatment with a continuous third-line anesthetic. Both ketamine (KET) and propofol (PRO) led to high levels of mortality, and nearly all rats on these therapies had breakthrough seizure activity, demonstrating super-refractory SE (SRSE). For the small subset of rats in which SE was fully resolved, significant improvements over controls were observed in recovery metrics, behavioral assays, and brain pathology. Together these data suggest that NA-induced SE is particularly severe, but aggressive treatment in the intensive care setting can lead to positive functional outcomes for casualties.
Collapse
Affiliation(s)
- Julia E Morgan
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Sara C Wilson
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Benjamin J Travis
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Kathryn H Bagri
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Kathleen T Pagarigan
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Hannah M Belski
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Cecelia Jackson
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Kevin M Bounader
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Jessica M Coppola
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Eden N Hornung
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - James E Johnson
- Comparative Pathology Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| | - Hilary S McCarren
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, United States
| |
Collapse
|
17
|
Barker BS, Spampanato J, McCarren HS, Berger K, Jackson CE, Yeung DT, Dudek FE, McDonough JH. The K v7 Modulator, Retigabine, is an Efficacious Antiseizure Drug for Delayed Treatment of Organophosphate-induced Status Epilepticus. Neuroscience 2021; 463:143-158. [PMID: 33836243 PMCID: PMC8142924 DOI: 10.1016/j.neuroscience.2021.03.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
Benzodiazepines are the primary treatment option for organophosphate (OP)-induced status epilepticus (SE), but these antiseizure drugs (ASDs) lose efficacy as treatment is delayed. In the event of a mass civilian or military exposure, significant treatment delays are likely. New ASDs that combat benzodiazepine-resistant, OP-induced SE are critically needed, particularly if they can be efficacious after a long treatment delay. This study evaluated the efficacy of the Kv7 channel modulator, retigabine, as a novel therapy for OP-induced SE. Adult, male rats were exposed to soman or diisopropyl fluorophosphate (DFP) to elicit SE and monitored by electroencephalogram (EEG) recording. Retigabine was administered alone or adjunctive to midazolam (MDZ) at delays of 20- or 40-min in the soman model, and 60-min in the DFP model. Following EEG recordings, rats were euthanized and brain tissue was collected for Fluoro-Jade B (FJB) staining to quantify neuronal death. In the DFP model, MDZ + 15 mg/kg retigabine suppressed seizure activity and was neuroprotective. In the soman model, MDZ + 30 mg/kg retigabine suppressed seizures at 20- and 40-min delays. Without MDZ, 15 mg/kg retigabine provided partial antiseizure and neuroprotectant efficacy in the DFP model, while 30 mg/kg without MDZ failed to attenuate soman-induced SE. At 60 mg/kg, retigabine without MDZ strongly reduced seizure activity and neuronal degeneration against soman-induce SE. This study demonstrates the antiseizure and neuroprotective efficacy of retigabine against OP-induced SE. Our data suggest retigabine could be a useful adjunct to standard-of-care and has potential for use in the absence of MDZ.
Collapse
Affiliation(s)
- Bryan S Barker
- United States Army Medical Research Institute of Chemical Defense, Medical Toxicology Research Division, Neuroscience Department, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Jay Spampanato
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA.
| | - Hilary S McCarren
- United States Army Medical Research Institute of Chemical Defense, Medical Toxicology Research Division, Neuroscience Department, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Kyle Berger
- United States Army Medical Research Institute of Chemical Defense, Medical Toxicology Research Division, Neuroscience Department, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Cecelia E Jackson
- United States Army Medical Research Institute of Chemical Defense, Medical Toxicology Research Division, Neuroscience Department, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - David T Yeung
- National Institutes of Health/National Institute of Allergy and Infectious Disease, Bethesda, MD 20892, USA
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - John H McDonough
- United States Army Medical Research Institute of Chemical Defense, Medical Toxicology Research Division, Neuroscience Department, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| |
Collapse
|
18
|
Loughery TN, Whitten KA, Acon-Chen C, Shih TM. Intramuscularly administered A1 adenosine receptor agonists as delayed treatment for organophosphorus nerve agent-induced Status Epilepticus. Toxicol Appl Pharmacol 2021; 419:115515. [PMID: 33798593 DOI: 10.1016/j.taap.2021.115515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/22/2021] [Accepted: 03/28/2021] [Indexed: 11/20/2022]
Abstract
Exposure to organophosphorus nerve agents (NAs) like sarin (GB) and soman (GD) can lead to sustained seizure activity, or status epilepticus (SE). Previous research has shown that activation of A1 adenosine receptors (A1ARs) can inhibit neuronal excitability, which could aid in SE termination. Two A1AR agonists, 2-Chloro-N6-cyclopentyladenosine (CCPA) and N-Bicyclo(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), were effective in terminating GD-induced SE in rats when administered via intraperitoneal (IP) injection. However, IP injection is not a clinically relevant route of administration. This study evaluated the efficacy of these agonists in terminating NA-induced SE when administered via intramuscular (IM) route. Adult male rats were exposed subcutaneously (SC) to either GB (150 μg/kg) or GD (90 μg/kg) and were treated with ENBA or CCPA at 15, 30, or 60 min after seizure onset or left untreated. Up to 7 days after exposure, deeply anesthetized rats were euthanized and perfused brains were removed for histologic assessment of neuropathology (i.e., neuronal damage) in six brain regions (amygdala, cerebral cortex, piriform cortex, thalamus, dorsal hippocampus, and ventral hippocampus). A total neuropathy score (0-24) was determined for each rat by adding the scores from each of the six regions. The higher the total score the more severe the neuropathology. With the GB model and 60 min treatment delay, ENBA-treated rats experienced 78.6% seizure termination (N = 14) and reduced neuropathology (11.6 ± 2.6, N = 5), CCPA-treated rats experienced 85.7% seizure termination (N = 14) and slightly reduced neuropathology (20.7 ± 1.8, N = 6), and untreated rats experienced no seizure termination (N = 13) and severe neuropathology (22.3 ± 1.0, N = 4). With the GD model and 60 min treatment delay, ENBA-treated rats experienced 92.9% seizure termination (N = 14) and reduced neuropathology (13.96 ± 1.8, N = 9), CCPA-treated rats experienced 78.6% seizure termination (N = 14) and slightly reduced neuropathology (22.0 ± 0.9, N = 10); and untreated rats experienced 16.7% seizure termination (N = 12) and severe neuropathology (22.0 ± 1.8, N = 5). While ENBA and CCPA both demonstrate a clear ability to terminate SE when administered up to 60 min after seizure onset, ENBA offers more neuroprotection, making it a promising candidate for NA-induced SE.
Collapse
Affiliation(s)
- Tara N Loughery
- Neuroscience Department, Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Kimberly A Whitten
- Comparative Pathology Department, Research Support Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Cindy Acon-Chen
- Neuroscience Department, Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA
| | - Tsung-Ming Shih
- Neuroscience Department, Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010-5400, USA.
| |
Collapse
|
19
|
Gore A, Neufeld-Cohen A, Egoz I, Baranes S, Gez R, Efrati R, David T, Dekel Jaoui H, Yampolsky M, Grauer E, Chapman S, Lazar S. Neuroprotection by delayed triple therapy following sarin nerve agent insult in the rat. Toxicol Appl Pharmacol 2021; 419:115519. [PMID: 33823148 DOI: 10.1016/j.taap.2021.115519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 02/08/2023]
Abstract
The development of refractory status epilepticus (SE) induced by sarin intoxication presents a therapeutic challenge. In our current research we evaluate the efficacy of a delayed combined triple treatment in ending the abnormal epileptiform seizure activity (ESA) and the ensuing of long-term neuronal insult. SE was induced in male Sprague-Dawley rats by exposure to 1.2LD50 sarin insufficiently treated by atropine and TMB4 (TA) 1 min later. Triple treatment of ketamine, midazolam and valproic acid was administered 30 min or 1 h post exposure and was compared to a delayed single treatment with midazolam alone. Toxicity and electrocorticogram activity were monitored during the first week and behavioral evaluation performed 3 weeks post exposure followed by brain biochemical and immunohistopathological analyses. The addition of both single and triple treatments reduced mortality and enhanced weight recovery compared to the TA-only treated group. The triple treatment also significantly minimized the duration of the ESA, reduced the sarin-induced increase in the neuroinflammatory marker PGE2, the brain damage marker TSPO, decreased the gliosis, astrocytosis and neuronal damage compared to the TA+ midazolam or only TA treated groups. Finally, the triple treatment eliminated the sarin exposed increased open field activity, as well as impairing recognition memory as seen in the other experimental groups. The delayed triple treatment may serve as an efficient therapy, which prevents brain insult propagation following sarin-induced refractory SE, even if treatment is postponed for up to 1 h.
Collapse
Affiliation(s)
- Ariel Gore
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel.
| | - Adi Neufeld-Cohen
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel
| | - Inbal Egoz
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel
| | - Shlomi Baranes
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel
| | - Rellie Gez
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel
| | - Rahav Efrati
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel
| | - Tse'ela David
- The Veterinary Center for Pre-clinical Research, Israel Institute for Biological, Chemical and Environmental Sciences, Ness- Ziona 74100, Israel
| | - Hani Dekel Jaoui
- The Veterinary Center for Pre-clinical Research, Israel Institute for Biological, Chemical and Environmental Sciences, Ness- Ziona 74100, Israel
| | - Michael Yampolsky
- The Veterinary Center for Pre-clinical Research, Israel Institute for Biological, Chemical and Environmental Sciences, Ness- Ziona 74100, Israel
| | - Ettie Grauer
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel
| | - Shira Chapman
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel
| | - Shlomi Lazar
- Department of Pharmacology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness-Ziona 74100, Israel.
| |
Collapse
|
20
|
Reddy DS, Zaayman M, Kuruba R, Wu X. Comparative profile of refractory status epilepticus models following exposure of cholinergic agents pilocarpine, DFP, and soman. Neuropharmacology 2021; 191:108571. [PMID: 33878303 DOI: 10.1016/j.neuropharm.2021.108571] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/24/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022]
Abstract
Status epilepticus (SE) is a medical emergency with continuous seizure activity that causes profound neuronal damage, morbidity, or death. SE incidents can arise spontaneously but mostly are elicited by seizurogenic triggers. Chemoconvulsants such as the muscarinic agonist pilocarpine and, organophosphates (OP) such as the pesticide diisopropylfluorophosphate (DFP) and, the nerve agent soman, can induce SE. Pilocarpine, DFP, and soman share a common feature of cholinergic crisis that transitions into a state of refractory SE, but their comparative profiles remain unclear. Here, we evaluated the comparative convulsant profile of pilocarpine, DFP, and soman to produce refractory SE and brain damage in rats. Behavioral and electrographic seizures were monitored for 24 h after exposure, and the extent of brain injury was determined by histological markers of neuronal injury and degeneration. Seizures were elicited rather slowly after pilocarpine as compared to DFP or soman, which caused rapid onset of spiking that swiftly developed into persistent SE. Time-course of SE activity after DFP was comparable to that after soman, a potent nerve agent. Diazepam controlled pilocarpine-induced SE, but it was ineffective in reducing OP-induced SE. All three agents produced modestly different degrees of neuronal injury and neurodegeneration in the brain. These results reveal distinct convulsant and neuronal injury patterns following exposure to cholinergic agonists, OP pesticides, and nerve agents. A battery of SE models, especially SE induced by cholinergic agents and other etiologies including epilepsy and brain tumors, is essential to identify novel anticonvulsant therapies for the management of refractory SE.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center College of Medicine, Bryan, TX, 77807, USA.
| | - Marcus Zaayman
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Ramkumar Kuruba
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center College of Medicine, Bryan, TX, 77807, USA
| |
Collapse
|
21
|
Maupu C, Enderlin J, Igert A, Oger M, Auvin S, Hassan-Abdi R, Soussi-Yanicostas N, Brazzolotto X, Nachon F, Dal Bo G, Dupuis N. Diisopropylfluorophosphate-induced status epilepticus drives complex glial cell phenotypes in adult male mice. Neurobiol Dis 2021; 152:105276. [PMID: 33529768 DOI: 10.1016/j.nbd.2021.105276] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/14/2020] [Accepted: 01/24/2021] [Indexed: 12/11/2022] Open
Abstract
Organophosphate pesticides and nerve agents (OPs), are characterized by cholinesterase inhibition. In addition to severe peripheral symptoms, high doses of OPs can lead to seizures and status epilepticus (SE). Long lasting seizure activity and subsequent neurodegeneration promote neuroinflammation leading to profound pathological alterations of the brain. The aim of this study was to characterize neuroinflammatory responses at key time points after SE induced by the OP, diisopropylfluorophosphate (DFP). Immunohistochemistry (IHC) analysis and RT-qPCR on cerebral tissue are often insufficient to identity and quantify precise neuroinflammatory alterations. To address these needs, we performed RT-qPCR quantification after whole brain magnetic-activated cell-sorting (MACS) of CD11B (microglia/infiltrated macrophages) and GLAST (astrocytes)-positive cells at 1, 4, 24 h and 3 days post-SE. In order to compare these results to those obtained by IHC, we performed, classical Iba1 (microglia/infiltrated macrophages) and GFAP (astrocytes) IHC analysis in parallel, focusing on the hippocampus, a brain region affected by seizure activity and neurodegeneration. Shortly after SE (1-4 h), an increase in pro-inflammatory (M1-like) markers and A2-specific markers, proposed as neurotrophic, were observed in CD11B and GLAST-positive isolated cells, respectively. Microglial cells successively expressed immuno-regulatory (M2b-like) and anti-inflammatory (M2a-like) at 4 h and 24 h post-SE induction. At 24 h and 3 days, A1-specific markers, proposed as neurotoxic, were increased in isolated astrocytes. Although IHC analysis presented no modification in terms of percentage of marked area and cell number at 1 and 4 h after SE, at 24 h and 3 days after SE, microglial and astrocytic activation was visible by IHC as an increase in Iba1 and GFAP-positive area and Iba1-positive cells in DFP animals when compared to the control. Our work identified sequential microglial and astrocytic phenotype activation. Although the role of each phenotype in SE cerebral outcomes requires further study, targeting specific markers at specific time point could be a beneficial strategy for DFP-induced SE treatment.
Collapse
Affiliation(s)
- Clémence Maupu
- Département de Toxicologie et risques chimiques, Institut de recherche biomédicale des armées, BP73, F-91223 Brétigny sur Orge cedex, France
| | - Julie Enderlin
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France; Service de neurologie pédiatrique, AP-HP, Hôpital Robert Debré, F-75019 Paris, France
| | - Alexandre Igert
- Département de Toxicologie et risques chimiques, Institut de recherche biomédicale des armées, BP73, F-91223 Brétigny sur Orge cedex, France
| | - Myriam Oger
- Unité Imagerie, Institut de recherche biomédicale des armées, BP73, F-91223 Brétigny sur Orge cedex, France
| | - Stéphane Auvin
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France; Service de neurologie pédiatrique, AP-HP, Hôpital Robert Debré, F-75019 Paris, France
| | | | | | - Xavier Brazzolotto
- Département de Toxicologie et risques chimiques, Institut de recherche biomédicale des armées, BP73, F-91223 Brétigny sur Orge cedex, France
| | - Florian Nachon
- Département de Toxicologie et risques chimiques, Institut de recherche biomédicale des armées, BP73, F-91223 Brétigny sur Orge cedex, France
| | - Grégory Dal Bo
- Département de Toxicologie et risques chimiques, Institut de recherche biomédicale des armées, BP73, F-91223 Brétigny sur Orge cedex, France
| | - Nina Dupuis
- Département de Toxicologie et risques chimiques, Institut de recherche biomédicale des armées, BP73, F-91223 Brétigny sur Orge cedex, France.
| |
Collapse
|
22
|
Itin C, Liani M, Zur M, Gorenbein P. Stability of Diazepam Solution for Injection Following Long-Term Storage in an Ambient Temperature of the Mediterranean Climate. Pharm Res 2021; 38:361-367. [PMID: 33404991 DOI: 10.1007/s11095-020-02984-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE Diazepam is utilized as a convulsion antidote following nerve gas attacks. As an emergency medicine, it requires storage at ambient temperatures which often doesn't meet manufacturers' requirements, leading to an early invalidation of the product. Current work investigated this issue. METHODS Long-term stability of diazepam ampoules for injection stored in an ambient temperature of the Mediterranean climate for ~10 years vs storage at room temperature was studied. RESULTS Diazepam assay and pH remained within pharmacopeial specifications irrespective of storage conditions. A major degradation product 2-methylamino-5-chlorobenzophenone (MACB) showed a clear trend of accumulation as a function of storage time, exceeding the permitted limit at ~2 years, irrespective of storage conditions. A strong correlation between the discoloration of the solutions and the concentration of MACB was obtained. Intravenous administration of MACB to rats at doses ~2200-fold higher than permissible specification levels caused neither mortality nor any toxicological nor post-mortem findings. CONCLUSIONS Regarding the parameters tested: diazepam assay, MACB assay, and pH, storing ampoules of diazepam solution for injection in field conditions of high temperatures of the Mediterranean climate did not cause accelerated degradation as compared to room temperature. These findings open an option for the usage of expired ampoules in special scenarios.
Collapse
Affiliation(s)
- Constantin Itin
- Pharmacy, Biomedical Engineering & Medical Supplies Branch, Israel Defense Forces (IDF) Medical Corps, Ramat Gan, Israel.
| | - Maor Liani
- Pharmacy, Biomedical Engineering & Medical Supplies Branch, Israel Defense Forces (IDF) Medical Corps, Ramat Gan, Israel
| | - Meital Zur
- Pharmacy, Biomedical Engineering & Medical Supplies Branch, Israel Defense Forces (IDF) Medical Corps, Ramat Gan, Israel
| | - Pavel Gorenbein
- Pharmacy, Biomedical Engineering & Medical Supplies Branch, Israel Defense Forces (IDF) Medical Corps, Ramat Gan, Israel
| |
Collapse
|
23
|
Reddy SD, Wu X, Kuruba R, Sridhar V, Reddy DS. Magnetic resonance imaging analysis of long-term neuropathology after exposure to the nerve agent soman: correlation with histopathology and neurological dysfunction. Ann N Y Acad Sci 2020; 1480:116-135. [PMID: 32671850 PMCID: PMC7708405 DOI: 10.1111/nyas.14431] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/09/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022]
Abstract
Nerve agents (NAs) produce acute and long-term brain injury and dysfunction, as evident from the Japan and Syria incidents. Magnetic resonance imaging (MRI) is a versatile technique to examine such chronic anatomical, functional, and neuronal damage in the brain. The objective of this study was to investigate long-term structural and neuronal lesion abnormalities in rats exposed to acute soman intoxication. T2-weighted MRI images of 10 control and 17 soman-exposed rats were acquired using a Siemens MRI system at 90 days after soman exposure. Quantification of brain tissue volumes and T2 signal intensity was conducted using the Inveon Research Workplace software and the extent of damage was correlated with histopathology and cognitive function. Soman-exposed rats showed drastic hippocampal atrophy with neuronal loss and reduced hippocampal volume (HV), indicating severe damage, but had similar T2 relaxation times to the control group, suggesting limited scarring and fluid density changes despite the volume decrease. Conversely, soman-exposed rats displayed significant increases in lateral ventricle volumes and T2 times, signifying strong cerebrospinal fluid expansion in compensation for tissue atrophy. The total brain volume, thalamic volume, and thalamic T2 time were similar in both groups, however, suggesting that some brain regions remained more intact long-term after soman intoxication. The MRI neuronal lesions were positively correlated with the histological markers of neurodegeneration and neuroinflammation 90 days after soman exposure. The predominant MRI hippocampal atrophy (25%) was highly consistent with massive reduction (35%) of neuronal nuclear antigen-positive (NeuN+ ) principal neurons and parvalbumin-positive (PV+ ) inhibitory interneurons within this brain region. The HV was significantly correlated with both inflammatory markers of GFAP+ astrogliosis and IBA1+ microgliosis. The reduced HV was also directly correlated with significant memory deficits in the soman-exposed cohort, confirming a possible neurobiological basis for neurological dysfunction. Together, these findings provide powerful insight on long-term region-specific neurodegenerative patterns after soman exposure and demonstrate the feasibility of in vivo neuroimaging to monitor neuropathology, predict the risk of neurological deficits, and evaluate response to medical countermeasures for NAs.
Collapse
Affiliation(s)
- Sandesh D Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
- Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Ramkumar Kuruba
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Vidya Sridhar
- Texas A&M Institute for Preclinical Studies, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
24
|
Dhir A, Bruun DA, Guignet M, Tsai Y, González E, Calsbeek J, Vu J, Saito N, Tancredi DJ, Harvey DJ, Lein PJ, Rogawski MA. Allopregnanolone and perampanel as adjuncts to midazolam for treating diisopropylfluorophosphate-induced status epilepticus in rats. Ann N Y Acad Sci 2020; 1480:183-206. [PMID: 32915470 PMCID: PMC7756871 DOI: 10.1111/nyas.14479] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/29/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
Abstract
Combinations of midazolam, allopregnanolone, and perampanel were assessed for antiseizure activity in a rat diisopropylfluorophosphate (DFP) status epilepticus model. Animals receiving DFP followed by atropine and pralidoxime exhibited continuous high-amplitude rhythmical electroencephalography (EEG) spike activity and behavioral seizures for more than 5 hours. Treatments were administered intramuscularly 40 min after DFP. Seizures persisted following midazolam (1.8 mg/kg). The combination of midazolam with either allopregnanolone (6 mg/kg) or perampanel (2 mg/kg) terminated EEG and behavioral status epilepticus, but the onset of the perampanel effect was slow. The combination of midazolam, allopregnanolone, and perampanel caused rapid and complete suppression of EEG and behavioral seizures. In the absence of DFP, animals treated with the three-drug combination were sedated but not anesthetized. Animals that received midazolam alone exhibited spontaneous recurrent EEG seizures, whereas those that received the three-drug combination did not, demonstrating antiepileptogenic activity. All combination treatments reduced neurodegeneration as assessed with Fluoro-Jade C staining to a greater extent than midazolam alone, and most reduced astrogliosis as assessed by GFAP immunoreactivity but had mixed effects on markers of microglial activation. We conclude that allopregnanolone, a positive modulator of the GABAA receptor, and perampanel, an AMPA receptor antagonist, are potential adjuncts to midazolam in the treatment of benzodiazepine-refractory organophosphate nerve agent-induced status epilepticus.
Collapse
Affiliation(s)
- Ashish Dhir
- Department of Neurology, School of MedicineUniversity of California, DavisSacramentoCalifornia
| | - Donald A. Bruun
- Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisDavisCalifornia
| | - Michelle Guignet
- Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisDavisCalifornia
| | - Yi‐Hua Tsai
- Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisDavisCalifornia
| | - Eduardo González
- Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisDavisCalifornia
| | - Jonas Calsbeek
- Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisDavisCalifornia
| | - Joan Vu
- Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisDavisCalifornia
| | - Naomi Saito
- Department of Public Health Sciences, School of MedicineUniversity of California, DavisDavisCalifornia
| | - Daniel J. Tancredi
- Department of Pediatrics, School of MedicineUniversity of California, DavisSacramentoCalifornia
| | - Danielle J. Harvey
- Department of Public Health Sciences, School of MedicineUniversity of California, DavisDavisCalifornia
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary MedicineUniversity of California, DavisDavisCalifornia
| | - Michael A. Rogawski
- Department of Neurology, School of MedicineUniversity of California, DavisSacramentoCalifornia
| |
Collapse
|
25
|
Lane M, Carter D, Pescrille JD, Aracava Y, Fawcett WP, Basinger GW, Pereira EFR, Albuquerque EX. Oral Pretreatment with Galantamine Effectively Mitigates the Acute Toxicity of a Supralethal Dose of Soman in Cynomolgus Monkeys Posttreated with Conventional Antidotes. J Pharmacol Exp Ther 2020; 375:115-126. [PMID: 32759369 DOI: 10.1124/jpet.120.265843] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022] Open
Abstract
Earlier reports suggested that galantamine, a drug approved to treat mild-to-moderate Alzheimer's disease (AD), and other centrally acting reversible acetylcholinesterase (AChE) inhibitors can serve as adjunct pretreatments against poisoning by organophosphorus compounds, including the nerve agent soman. The present study was designed to determine whether pretreatment with a clinically relevant oral dose of galantamine HBr mitigates the acute toxicity of 4.0×LD50 soman (15.08 µg/kg) in Macaca fascicularis posttreated intramuscularly with the conventional antidotes atropine (0.4 mg/kg), 2-pyridine aldoxime methyl chloride (30 mg/kg), and midazolam (0.32 mg/kg). The pharmacokinetic profile and maximal degree of blood AChE inhibition (∼25%-40%) revealed that the oral doses of 1.5 and 3.0 mg/kg galantamine HBr in these nonhuman primates (NHPs) translate to human-equivalent doses that are within the range used for AD treatment. Subsequent experiments demonstrated that 100% of NHPs pretreated with either dose of galantamine, challenged with soman, and posttreated with conventional antidotes survived 24 hours. By contrast, given the same posttreatments, 0% and 40% of the NHPs pretreated, respectively, with vehicle and pyridostigmine bromide (1.2 mg/kg, oral), a peripherally acting reversible AChE inhibitor approved as pretreatment for military personnel at risk of exposure to soman, survived 24 hours after the challenge. In addition, soman caused extensive neurodegeneration in the hippocampi of saline- or pyridostigmine-pretreated NHPs, but not in the hippocampi of galantamine-pretreated animals. To our knowledge, this is the first study to demonstrate the effectiveness of clinically relevant oral doses of galantamine to prevent the acute toxicity of supralethal doses of soman in NHPs. SIGNIFICANCE STATEMENT: This is the first study to demonstrate that a clinically relevant oral dose of galantamine effectively prevents lethality and neuropathology induced by a supralethal dose of the nerve agent soman in Cynomolgus monkeys posttreated with conventional antidotes. These findings are of major significance for the continued development of galantamine as an adjunct pretreatment against nerve agent poisoning.
Collapse
Affiliation(s)
- Malcolm Lane
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| | - D'Arice Carter
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| | - Joseph D Pescrille
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| | - Yasco Aracava
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| | - William P Fawcett
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| | - G William Basinger
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| | - Edna F R Pereira
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| | - Edson X Albuquerque
- Division of Translational Toxicology, Department of Epidemiology and Public Health (M.L., D.C., J.D.P., Y.A., W.P.F., E.F.R.P., E.X.A.) and Department of Pharmacology (E.F.R.P., E.X.A.), University of Maryland School of Medicine, Baltimore, Maryland; and Countervail Corp., Charlotte, North Carolina (G.W.B.)
| |
Collapse
|
26
|
Reddy DS, Perumal D, Golub V, Habib A, Kuruba R, Wu X. Phenobarbital as alternate anticonvulsant for organophosphate-induced benzodiazepine-refractory status epilepticus and neuronal injury. Epilepsia Open 2020; 5:198-212. [PMID: 32524045 PMCID: PMC7278559 DOI: 10.1002/epi4.12389] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 02/08/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022] Open
Abstract
Objective Organophosphates (OPs) such as diisopropylfluorophosphate (DFP) and soman are lethal chemical agents that can produce seizures, refractory status epilepticus (SE), and brain damage. There are few optimal treatments for late or refractory SE. Phenobarbital is a second‐line drug for SE, usually after lorazepam, diazepam, or midazolam have failed to stop SE. Practically, 40 minutes or less is often necessary for first responders to arrive and assist in a chemical incident. However, it remains unclear whether administration of phenobarbital 40 minutes after OP intoxication is still effective. Here, we investigated the efficacy of phenobarbital treatment at 40 minutes postexposure to OP intoxication. Methods Acute refractory SE was induced in rats by DFP injection as per a standard paradigm. After 40 minutes, subjects were given phenobarbital intramuscularly (30‐100 mg/kg) and progression of seizure activity was monitored by video‐EEG recording. The extent of brain damage was assessed 3 days after DFP injections by neuropathology analysis of neurodegeneration and neuronal injury by unbiased stereology. Results Phenobarbital produced a dose‐dependent seizure protection. A substantial decrease in SE was evident at 30 and 60 mg/kg, and a complete seizure termination was noted at 100 mg/kg within 40 minutes after treatment. Neuropathology findings showed significant neuroprotection in 100 mg/kg cohorts in brain regions associated with SE. Although higher doses resulted in greater protection against refractory SE and neuronal damage, they did not positively correlate with improved survival rate. Moreover, phenobarbital caused serious adverse effects including anesthetic or comatose state and even death. Significance Phenobarbital appears as an alternate anticonvulsant for OP‐induced refractive SE in hospital settings. A careful risk‐benefit analysis is required because of negative outcomes on survival and cardio‐respiratory function. However, the need for sophisticated support and critical monitoring in hospital may preclude its use as medical countermeasure in mass casualty situations.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics College of Medicine Texas A&M University Health Science Center Bryan TX USA
| | - Dheepthi Perumal
- Department of Neuroscience and Experimental Therapeutics College of Medicine Texas A&M University Health Science Center Bryan TX USA
| | - Victoria Golub
- Department of Neuroscience and Experimental Therapeutics College of Medicine Texas A&M University Health Science Center Bryan TX USA
| | - Andy Habib
- Department of Neuroscience and Experimental Therapeutics College of Medicine Texas A&M University Health Science Center Bryan TX USA
| | - Ramkumar Kuruba
- Department of Neuroscience and Experimental Therapeutics College of Medicine Texas A&M University Health Science Center Bryan TX USA
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics College of Medicine Texas A&M University Health Science Center Bryan TX USA
| |
Collapse
|
27
|
Saberi F, Bahrami F, Saberi M, Mashhadi Akbar Boojar M. The pro-convulsant effects of diazinon low dose in male rats under amygdala kindling. Drug Chem Toxicol 2020; 45:625-632. [PMID: 32249606 DOI: 10.1080/01480545.2020.1746801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Organophosphates can damage the brain in systemic intoxication. In this study, the effects of a minimum toxic dose (MTD) of diazinon (DZ) on amygdala afterdischarge threshold (ADT), kindling acquisition and kindled seizure parameters were evaluated. Intact male rats were stereotactically implanted with a tripolar and two monopolar electrodes in the amygdala and dura respectively. After recovery, animals received daily either, olive oil (control), 15 or 30 mg/kg (MTD) of DZ intraperitoneally, and ADT, afterdischarge duration (ADD) at each stage (S1 to S5) of kindling and number of trials for kindling acquisition were determined daily. Also, the effect of DZ on stage 4 latency (S4L), ADD, stage 5 duration (S5D) and the activity of the red blood cholinesterase (ChE) were evaluated. The ADT was lower and the ADD was longer significantly in DZ treated group in comparison to control (p < 0.01) and the number of trials to reach each stage of kindling acquisition was reduced (p < 0.001). The total amount of ADDs during the kindling procedure increased significantly 5 days after DZ treatment. While the S4L was reduced, the S5D increased significantly after DZ treatment. The ChE activity was inhibited significantly after 20 min of DZ treatment and continued till 24 h (p < 0.01). Data indicate that even half of the MTD of DZ could increase the sensitivity and excitability of the CNS to the epileptic activity at least via reduction of stimulation threshold and AD prolongation. Furthermore, repeated exposure to the low concentrations of organophosphates may be pro-convulsant and should be restricted.
Collapse
Affiliation(s)
- Fatemeh Saberi
- Department of Dermatology, Razi Skin Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Bahrami
- Department of Physiology and Biophysics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehdi Saberi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdi Mashhadi Akbar Boojar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Evaluation of fosphenytoin, levetiracetam, and propofol as treatments for nerve agent-induced seizures in pediatric and adult rats. Neurotoxicology 2020; 79:58-66. [PMID: 32220603 DOI: 10.1016/j.neuro.2020.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/21/2020] [Accepted: 03/14/2020] [Indexed: 10/24/2022]
Abstract
Multiple recent instances of nerve agent (NA) exposure in civilian populations have occurred, resulting in a variety of negative effects and lethality in both adult and pediatric populations. Seizures are a prominent effect of NAs that can result in neurological damage and contribute to their lethality. Current anticonvulsant treatments for NAs are approved for adults, but no approved pediatric treatments exist. Further, the vast majority of NA-related research in animals has been conducted in adult male subjects. There is a need for research that includes female and pediatric populations in testing. In this project, adult and pediatric male and female rats were challenged with sarin or VX and then treated with fosphenytoin, levetiracetam, or propofol. In this study, fosphenytoin and levetiracetam failed to terminate seizure activity when animals were treated 5 min after seizure onset. Propofol was effective, exhibiting high efficacy and potency for terminating seizure activity quickly in pediatric and adult animals, suggesting it may be an effective anticonvulsant for NA-induced seizures in pediatric populations.
Collapse
|
29
|
Kundrick E, Marrero-Rosado B, Stone M, Schultz C, Walker K, Lee-Stubbs RB, de Araujo Furtado M, Lumley LA. Delayed midazolam dose effects against soman in male and female plasma carboxylesterase knockout mice. Ann N Y Acad Sci 2020; 1479:94-107. [PMID: 32027397 DOI: 10.1111/nyas.14311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/13/2020] [Accepted: 01/19/2020] [Indexed: 01/30/2023]
Abstract
Chemical warfare nerve agent exposure leads to status epilepticus that may progress to epileptogenesis and severe brain pathology when benzodiazepine treatment is delayed. We evaluated the dose-response effects of delayed midazolam (MDZ) on toxicity induced by soman (GD) in the plasma carboxylesterase knockout (Es1-/- ) mouse, which, similar to humans, lacks plasma carboxylesterase. Initially, we compared the median lethal dose (LD50 ) of GD exposure in female Es1-/- mice across estrous with male mice and observed a greater LD50 during estrus compared with proestrus or with males. Subsequently, male and female GD-exposed Es1-/- mice treated with a dose range of MDZ 40 min after seizure onset were evaluated for survivability, seizure activity, and epileptogenesis. GD-induced neuronal loss and microglial activation were evaluated 2 weeks after exposure. Similar to our previous observations in rats, delayed treatment with MDZ dose-dependently increased survival and reduced seizure severity in GD-exposed mice, but was unable to prevent epileptogenesis, neuronal loss, or gliosis. These results suggest that MDZ is beneficial against GD exposure, even when treatment is delayed, but that adjunct therapies to enhance protection need to be identified. The Es1-/- mouse GD exposure model may be useful to screen for improved medical countermeasures against nerve agent exposure.
Collapse
Affiliation(s)
- Erica Kundrick
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Brenda Marrero-Rosado
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Michael Stone
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Caroline Schultz
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Katie Walker
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Robyn B Lee-Stubbs
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | | | - Lucille A Lumley
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| |
Collapse
|
30
|
Bibi D, Bialer M. Pharmacokinetic and pharmacodynamic analysis of (2S,3S)‐
sec
‐butylpropylacetamide (SPD) in rats and pigs—A CNS‐active stereoisomer of SPD. Epilepsia 2020; 61:149-156. [DOI: 10.1111/epi.16411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/20/2019] [Accepted: 11/20/2019] [Indexed: 12/21/2022]
Affiliation(s)
- David Bibi
- Institute of Drug Research School of Pharmacy Faculty of Medicine The Hebrew University of Jerusalem Jerusalem Israel
| | - Meir Bialer
- Institute of Drug Research School of Pharmacy Faculty of Medicine The Hebrew University of Jerusalem Jerusalem Israel
- Affiliated with the David R. Bloom Center for Pharmacy The Hebrew University of Jerusalem Jerusalem Israel
| |
Collapse
|
31
|
Barker BS, Spampanato J, McCarren HS, Smolik M, Jackson CE, Hornung EN, Yeung DT, Dudek FE, McDonough JH. Screening for Efficacious Anticonvulsants and Neuroprotectants in Delayed Treatment Models of Organophosphate-induced Status Epilepticus. Neuroscience 2020; 425:280-300. [PMID: 31783100 PMCID: PMC6935402 DOI: 10.1016/j.neuroscience.2019.11.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 01/04/2023]
Abstract
Organophosphorus (OP) compounds are deadly chemicals that exert their intoxicating effects through the irreversible inhibition of acetylcholinesterase (AChE). In addition to an excess of peripheral ailments, OP intoxication induces status epilepticus (SE) which if left untreated may lead to permanent brain damage or death. Benzodiazepines are typically the primary therapies for OP-induced SE, but these drugs lose efficacy as treatment time is delayed. The CounterACT Neurotherapeutic Screening (CNS) Program was therefore established by the National Institutes of Health (NIH) to discover novel treatments that may be administered adjunctively with the currently approved medical countermeasures for OP-induced SE in a delayed treatment scenario. The CNS program utilizes in vivo EEG recordings and Fluoro-JadeB (FJB) histopathology in two established rat models of OP-induced SE, soman (GD) and diisopropylfluorophosphate (DFP), to evaluate the anticonvulsant and neuroprotectant efficacy of novel adjunct therapies when administered at 20 or 60 min after the induction of OP-induced SE. Here we report the results of multiple compounds that have previously shown anticonvulsant or neuroprotectant efficacy in other models of epilepsy or trauma. Drugs tested were ganaxolone, diazoxide, bumetanide, propylparaben, citicoline, MDL-28170, and chloroquine. EEG analysis revealed that ganaxolone demonstrated the most robust anticonvulsant activity, whereas all other drugs failed to attenuate ictal activity in both models of OP-induced SE. FJB staining demonstrated that none of the tested drugs had widespread neuroprotective abilities. Overall these data suggest that neurosteroids may represent the most promising anticonvulsant option for OP-induced SE out of the seven unique mechanisms tested here. Additionally, these results suggest that drugs that provide significant neuroprotection from OP-induced SE without some degree of anticonvulsant activity are elusive, which further highlights the necessity to continue screening novel adjunct treatments through the CNS program.
Collapse
Affiliation(s)
- Bryan S Barker
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA.
| | - Jay Spampanato
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Hilary S McCarren
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Melissa Smolik
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Cecelia E Jackson
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Eden N Hornung
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - David T Yeung
- Chemical Countermeasures Research Program, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - John H McDonough
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| |
Collapse
|
32
|
Abstract
This article describes current pursuits for developing novel antidotes for organophosphate (OP) intoxication. Recent mechanistic studies of benzodiazepine-resistant seizures have key consequences for victims of OP pesticide and nerve agent attacks. We uncovered why current therapies are not able to stop the OP-induced seizures and brain cell death and what type of drug might be better. OP exposure down regulates critical inhibitory GABA-A receptors, kills neurons, and causes massive neuroinflammation that will cause more neuronal death, which causes the problem of too few benzodiazepine receptors. The loss of inhibitory interneurons creates a self-sustaining seizure circuit and refractory status epilepticus. Thus, there is an urgent need for mechanism-based, new antidotes for OP intoxication. We have discovered neurosteroids as next-generation anticonvulsants superior to midazolam for the treatment of OP poisoning. Neurosteroids that activate both extrasynaptic and synaptic GABA-A receptors have the potential to stop seizures more effectively and safely than benzodiazepines. In addition, neurosteroids confers robust neuroprotection by reducing neuronal injury and neuroinflammation. The synthetic neurosteroid ganaxolone is being considered for advanced development as a future anticonvulsant for nerve agents. Experimental studies shows striking efficacy of ganaxolone and its analogs in OP exposure models. They are also effective in attenuating long-term neuropsychiatric deficits caused by OP exposure. Overall, neurosteroids represent rational anticonvulsants for OP intoxication, even when given late after exposure.
Collapse
|
33
|
Spampanato J, Pouliot W, Bealer SL, Roach B, Dudek FE. Antiseizure and neuroprotective effects of delayed treatment with midazolam in a rodent model of organophosphate exposure. Epilepsia 2019; 60:1387-1398. [PMID: 31125451 PMCID: PMC6662604 DOI: 10.1111/epi.16050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Exposure to organophosphates (OPs) and OP nerve agents (NAs) causes status epilepticus (SE) and irreversible brain damage. Rapid control of seizure activity is important to minimize neuronal injury and the resulting neurological and behavioral disorders; however, early treatment will not be possible after mass release of OPs or NAs. METHODS We utilized a delayed-treatment model of OP exposure in adult rats by administration of diisopropyl fluorophosphate (DFP) to study the relationship between the antiseizure and neuroprotective effects of the "standard-of-care" benzodiazepine, midazolam (MDZ), when given at 30, 60, and 120 minutes after SE onset. After electroencephalography (EEG) recordings, neural damage in serial brain sections was studied with Fluoro-Jade B staining. RESULTS MDZ-induced seizure suppression was equivalent in magnitude regardless of treatment delay (ie, seizure duration). When assessed globally (ie, normalized across 10 different brain regions) for each treatment delay, MDZ administration resulted in only nonsignificant reductions in neuronal death. However, when data for MDZ treatment were combined from all three delay times, a small but significant reduction in global neuronal death was detected when compared to vehicle treatment, which indicated that the substantive MDZ-induced seizure suppression led to only a small reduction in neuronal death. SIGNIFICANCE In conclusion, MDZ significantly reduced DFP-induced SE intensity when treatment was delayed 30, 60, and even up to 120 minutes; however, this reduction in seizure intensity had no detectable effect on neuronal death at each individual delay time. These data show that although MDZ suppressed seizures, additional neuroprotective therapies are needed to mitigate the effects of OP exposure.
Collapse
Affiliation(s)
- Jay Spampanato
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Wendy Pouliot
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Steven L Bealer
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Bonnie Roach
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Francis Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
34
|
Hoffman KM, Eisen MR, Chandler JK, Nelson MR, Johnson EA, McNutt PM. Retrograde activation of CB1R by muscarinic receptors protects against central organophosphorus toxicity. Neuropharmacology 2019; 155:113-120. [PMID: 31132436 DOI: 10.1016/j.neuropharm.2019.05.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/18/2019] [Accepted: 05/21/2019] [Indexed: 11/17/2022]
Abstract
The acute toxicity of organophosphorus-based compounds is primarily a result of acetylcholinesterase inhibition in the central and peripheral nervous systems. The resulting cholinergic crisis manifests as seizure, paralysis, respiratory failure and neurotoxicity. Though overstimulation of muscarinic receptors is the mechanistic basis of central organophosphorus (OP) toxicities, short-term changes in synapse physiology that precede OP-induced seizures have not been investigated in detail. To study acute effects of OP exposure on synaptic function, field excitatory postsynaptic potentials (fEPSPs) were recorded from Schaffer collateral synapses in the mouse hippocampus CA1 stratum radiatum during perfusion with various OP compounds. Administration of the OPs paraoxon, soman or VX rapidly and stably depressed fEPSPs via a presynaptic mechanism, while the non-OP proconvulsant tetramethylenedisulfotetramine had no effect on fEPSP amplitudes. OP-induced presynaptic long-term depression manifested prior to interictal spiking, occurred independent of recurrent firing, and did not require NMDA receptor currents, suggesting that it was not mediated by activity-dependent calcium uptake. Pharmacological dissection revealed that the presynaptic endocannabinoid type 1 receptor (CB1R) as well as postsynaptic M1 and M3 muscarinic acetylcholine receptors were necessary for OP-LTD. Administration of CB1R antagonists significantly reduced survival in mice after a soman challenge, revealing an acute protective role for endogenous CB1R signaling during OP exposure. Collectively these data demonstrate that the endocannabinoid system alters glutamatergic synaptic function during the acute response to OP acetylcholinesterase inhibitors.
Collapse
Affiliation(s)
- Katie M Hoffman
- Biological Sciences, Lehigh University, 27 Memorial Drive West, Bethlehem, PA, 18015, USA
| | - Margaret R Eisen
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD, 21010, USA
| | - Jessica K Chandler
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD, 21010, USA
| | - Marian R Nelson
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD, 21010, USA
| | - Erik A Johnson
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD, 21010, USA
| | - Patrick M McNutt
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD, 21010, USA.
| |
Collapse
|
35
|
Johnstone TBC, McCarren HS, Spampanato J, Dudek FE, McDonough JH, Hogenkamp D, Gee KW. Enaminone Modulators of Extrasynaptic α 4β 3δ γ-Aminobutyric Acid A Receptors Reverse Electrographic Status Epilepticus in the Rat After Acute Organophosphorus Poisoning. Front Pharmacol 2019; 10:560. [PMID: 31178732 PMCID: PMC6543275 DOI: 10.3389/fphar.2019.00560] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/03/2019] [Indexed: 01/22/2023] Open
Abstract
Seizures induced by organophosphorus nerve agent exposure become refractory to treatment with benzodiazepines because these drugs engage synaptic γ-aminobutyric acid-A receptors (GABAARs) that rapidly internalize during status epilepticus (SE). Extrasynaptic GABAARs, such as those containing α4β3δ subunits, are a putative pharmacological target to comprehensively manage nerve agent-induced seizures since they do not internalize during SE and are continuously available for activation. Neurosteroids related to allopregnanolone have been tested as a possible replacement for benzodiazepines because they target both synaptic and extrasynaptic GABAARs receptors. A longer effective treatment window, extended treatment efficacy, and enhanced neuroprotection represent significant advantages of neurosteroids over benzodiazepines. However, neurosteroid use is limited by poor physicochemical properties arising from the intrinsic requirement of the pregnane steroid core structure for efficacy rendering drug formulation problematic. We tested a non-steroidal enaminone GABAAR modulator that interacts with both synaptic and extrasynaptic GABAARs on a binding site distinct from neurosteroids or benzodiazepines for efficacy to control electrographic SE induced by diisopropyl fluorophosphate or soman intoxication in rats. Animals were treated with standard antidotes, and experimental therapeutic treatment was given following 1 h (diisopropyl fluorophosphate model) or 20 min (soman model) after SE onset. We found that the enaminone 2-261 had an extended duration of seizure termination (>10 h) in the diisopropyl fluorophosphate intoxication model in the presence or absence of midazolam (MDZ). 2-261 also moderately potentiated MDZ in the soman-induced seizure model but had limited efficacy as a stand-alone anticonvulsant treatment due to slow onset of action. 2-261 significantly reduced neuronal death in brain areas associated with either diisopropyl fluorophosphate- or soman-induced SE. 2-261 represents an alternate chemical template from neurosteroids for enhancing extrasynaptic α4β3δ GABAAR activity to reverse SE from organophosphorous intoxication.
Collapse
Affiliation(s)
- Timothy B C Johnstone
- Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Hilary S McCarren
- Neuroscience Department, Medical Toxicology Research Division, United States Army Research Institute of Chemical Defense, Aberdeen, MD, United States
| | - Jay Spampanato
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - John H McDonough
- Neuroscience Department, Medical Toxicology Research Division, United States Army Research Institute of Chemical Defense, Aberdeen, MD, United States
| | - Derk Hogenkamp
- Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Kelvin W Gee
- Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
36
|
In Vivo Evaluation of A1 Adenosine Agonists as Novel Anticonvulsant Medical Countermeasures to Nerve Agent Intoxication in a Rat Soman Seizure Model. Neurotox Res 2019; 36:323-333. [DOI: 10.1007/s12640-019-00034-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 10/26/2022]
|
37
|
Iyengar ARS, Pande AH. Is Human Paraoxonase 1 the Saviour Against the Persistent Threat of Organophosphorus Nerve Agents? Protein Pept Lett 2019; 26:471-478. [PMID: 30942142 DOI: 10.2174/0929866526666190403120259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 01/22/2023]
Abstract
Nerve agents have been used extensively in chemical warfare in the past. However, recent use of Novichok agents have reignited the debate on the threat posed by Organophosphorus Nerve Agents (OPNAs). The currently available therapy for OPNA toxicity is only symptomatic and is potentially ineffective in neutralizing OPNAs. Hence, there is a dire need to develop a prophylactic therapy for counteracting OPNA toxicity. In this regard, human paraoxonase 1 has emerged as the enzyme of choice. In this review, we have focussed upon the recent and past events of OPNA use, their mechanism of action and toxicity. Further, we have emphasized upon the potential of enzyme based therapy and the various advances in the development of paraoxonase 1 as a countermeasure for OPNA poisoning. Finally, we have elaborated the shortcomings of paraoxonase 1 and the work that needs to be undertaken in order to develop human paraoxonase 1 as a prophylactic against OPNA poisoning.
Collapse
Affiliation(s)
- A R Satvik Iyengar
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, 2308 NSW, Australia
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| |
Collapse
|
38
|
Aroniadou-Anderjaska V, Figueiredo TH, Apland JP, Braga MF. Targeting the glutamatergic system to counteract organophosphate poisoning: A novel therapeutic strategy. Neurobiol Dis 2019; 133:104406. [PMID: 30798006 DOI: 10.1016/j.nbd.2019.02.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/23/2019] [Accepted: 02/20/2019] [Indexed: 12/15/2022] Open
Abstract
One of the devastating effects of acute exposure to organophosphates, like nerve agents, is the induction of severe and prolonged status epilepticus (SE), which can cause death, or brain damage if death is prevented. Seizures after exposure are initiated by muscarinic receptor hyperstimulation-after inhibition of acetylcholinesterase by the organophosphorus agent and subsequent elevation of acetylcholine-but they are reinforced and sustained by glutamatergic hyperexcitation, which is the primary cause of brain damage. Diazepam is the FDA-approved anticonvulsant for the treatment of nerve agent-induced SE, and its replacement by midazolam is currently under consideration. However, clinical data derived from the treatment of SE of any etiology, as well as studies on the control of nerve agent-induced SE in animal models, have indicated that diazepam and midazolam control seizures only temporarily, their antiseizure efficacy is reduced as the latency of treatment from the onset of SE increases, and their neuroprotective efficacy is limited or absent. Here, we review data on the discovery of a novel anticonvulsant and neuroprotectant, LY293558, an AMPA/GluK1 receptor antagonist. Treatment of soman-exposed immature, young-adult, and aged rats with LY293558, terminates SE with limited recurrence of seizures, significantly protects from brain damage, and prevents long-term behavioral deficits, even when LY293558 is administered 1 h post-exposure. More beneficial effects and complete neuroprotection is obtained when LY293558 administration is combined with caramiphen, which antagonizes NMDA receptors. Further efficacy studies may bring the LY293558 + caramiphen combination therapy on the pathway to approval for human use.
Collapse
Affiliation(s)
- Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| | - James P Apland
- Neuroscience Program, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, United States of America.
| | - Maria F Braga
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America; Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| |
Collapse
|
39
|
Shih TM, Koenig JA, Acon Chen C. Comparative effects of scopolamine and phencynonate on organophosphorus nerve agent-induced seizure activity, neuropathology and lethality. Toxicol Mech Methods 2019; 29:322-333. [PMID: 30558458 DOI: 10.1080/15376516.2018.1558322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The efficacy of anticonvulsant therapies to stop seizure activities following organophosphorus nerve agents (NAs) has been documented as being time-dependent. We utilized the guinea pig NA-seizure model to compare the effectiveness of phencynonate (PCH) and scopolamine (SCP) when given at the early (at time of seizure onset) or late (40 min after seizure onset) phase of seizure progression. PCH possesses both anticholinergic and anti-NMDA activities, while SCP is a purely anti-muscarinic compound. Animals with cortical electrodes were pretreated with pyridostigmine bromide 30 min prior to exposure to a 2.0 x LD50 subcutaneous dose of a NA (GA, GB, GD, GF, VR, or VX), followed one min later with atropine sulfate and 2-PAM. At either early or late phase, animals were treated with either PCH or SCP and the 24-h anticonvulsant ED50 doses were determined. When administered at seizure onset, PCH, and SCP were both effective at terminating seizure activity against all NAs, with ED50 values for SCP generally being lower. At the 40 min time, ED50 values were obtained following GA, GD, GF, and VR challenges for SCP, but ED50 value was obtained only following GD for PCH, indicating a superior efficacy of SCP. When seizure activity was controlled, a significant improvement in weight loss, neuropathology, and survival was observed, regardless of treatment or NA. Overall, these results demonstrate the differing efficacies of these two similarly structured anticholinergic compounds with delayed administration and warrant further investigation into the timing and mechanisms of the seizure maintenance phase in different animal models.
Collapse
Affiliation(s)
- Tsung-Ming Shih
- a Medical Toxicology Research Division , US Army Medical Research Institute of Chemical Defense , Edgewood , MD , USA
| | - Jeffrey A Koenig
- a Medical Toxicology Research Division , US Army Medical Research Institute of Chemical Defense , Edgewood , MD , USA
| | - Cindy Acon Chen
- a Medical Toxicology Research Division , US Army Medical Research Institute of Chemical Defense , Edgewood , MD , USA
| |
Collapse
|
40
|
Haines KM, Matson LM, Dunn EN, Ardinger CE, Lee-Stubbs R, Bibi D, McDonough JH, Bialer M. Comparative efficacy of valnoctamide and sec-butylpropylacetamide (SPD) in terminating nerve agent-induced seizures in pediatric rats. Epilepsia 2019; 60:315-321. [PMID: 30615805 DOI: 10.1111/epi.14630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/02/2018] [Accepted: 12/03/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Children and adults are likely to be among the casualties in a civilian nerve agent exposure. This study evaluated the efficacy of valnoctamide (racemic-VCD), sec-butylpropylacetamide (racemic-SPD), and phenobarbital for stopping nerve agent seizures in both immature and adult rats. METHODS Female and male postnatal day (PND) 21, 28, and 70 (adult) rats, previously implanted with electroencephalography (EEG) electrodes were exposed to seizure-inducing doses of the nerve agents sarin or VX and EEG was recorded continuously. Five minutes after seizure onset, animals were treated with SPD, VCD, or phenobarbital. The up-down method was used over successive animals to determine the anticonvulsant median effective dose (ED50 ) of the drugs. RESULTS SPD-ED50 values in the VX model were the following: PND21, 53 mg/kg (male) and 48 mg/kg (female); PND28, 108 mg/kg (male) and 43 mg/kg (female); and PND70, 101 mg/kg (male) and 40 mg/kg (female). SPD-ED50 values in the sarin model were the following: PND21, 44 mg/kg (male) and 28 mg/kg (female); PND28, 79 mg/kg (male) and 34 mg/kg (female); and PND70, 53 mg/kg (male) and 53 mg/kg (female). VCD-ED50 values in the VX model were the following: PND21, 34 mg/kg (male) and 43 mg/kg (female); PND28, 165 mg/kg (male) and 59 mg/kg (female); and PND70, 87 mg/kg (male) and 91 mg/kg (female). VCD-ED50 values in the sarin model were the following: PND21, 45 mg/kg (male), 48 mg/kg (female); PND28, 152 mg/kg (male) 79 mg/kg (female); and PND70, 97 mg/kg (male) 79 mg/kg (female). Phenobarbital-ED50 values in the VX model were the following: PND21, 43 mg/kg (male) and 18 mg/kg (female); PND28, 48 mg/kg (male) and 97 mg/kg (female). Phenobarbital-ED50 values in the sarin model were the following: PND21, 32 mg/kg (male) and 32 mg/kg (female); PND28, 58 mg/kg (male) and 97 mg/kg (female); and PND70, 65 mg/kg (female). SIGNIFICANCE SPD and VCD demonstrated anticonvulsant activity in both immature and adult rats in the sarin- and VX-induced status epilepticus models. Phenobarbital was effective in immature rats, whereas in adult rats, higher doses were required that were accompanied by toxicity. Overall, significantly less drug was required to stop seizures in PND21 animals than in the older animals, and overall, males required higher amounts of drug than females.
Collapse
Affiliation(s)
- Kari M Haines
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Liana M Matson
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Emily N Dunn
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Cherish E Ardinger
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Robyn Lee-Stubbs
- Research Support Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - David Bibi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - John H McDonough
- Nerve Agent Countermeasures, Medical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Meir Bialer
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.,David R. Bloom Center for Pharmacy, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
41
|
Kuruba R, Wu X, Reddy DS. Benzodiazepine-refractory status epilepticus, neuroinflammation, and interneuron neurodegeneration after acute organophosphate intoxication. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2845-2858. [PMID: 29802961 PMCID: PMC6066461 DOI: 10.1016/j.bbadis.2018.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/30/2018] [Accepted: 05/22/2018] [Indexed: 12/19/2022]
Abstract
Nerve agents and some pesticides such as diisopropylfluorophosphate (DFP) cause neurotoxic manifestations that include seizures and status epilepticus (SE), which are potentially lethal and carry long-term neurological morbidity. Current antidotes for organophosphate (OP) intoxication include atropine, 2-PAM and diazepam (a benzodiazepine for treating seizures and SE). There is some evidence for partial or complete loss of diazepam anticonvulsant efficacy when given 30 min or later after exposure to an OP; this condition is known as refractory SE. Effective therapies for OP-induced SE are lacking and it is unclear why current therapies do not work. In this study, we investigated the time-dependent efficacy of diazepam in the nerve agent surrogate DFP model of OP intoxication on seizure suppression and neuroprotection in rats, following an early and late therapy. Diazepam (5 mg/kg, IM) controlled seizures when given 10 min after DFP exposure ("early"), but it was completely ineffective at 60 or 120 min ("late") after DFP. DFP-induced neuronal injury, neuroinflammation, and neurodegeneration of principal cells and GABAergic interneurons were significantly reduced by early but not late therapy. These findings demonstrate that diazepam failed to control seizures, SE and neuronal injury when given 60 min or later after DFP exposure, confirming the benzodiazepine-refractory SE and brain damage after OP intoxication. In addition, this study indicates that degeneration of inhibitory interneurons and inflammatory glial activation are potential mechanisms underlying these morbid outcomes of OP intoxication. Therefore, novel anticonvulsant and neuroprotectant antidotes, superior to benzodiazepines, are desperately needed for controlling nerve agent-induced SE and brain injury.
Collapse
Affiliation(s)
- Ramkumar Kuruba
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
42
|
Wu X, Kuruba R, Reddy DS. Midazolam-Resistant Seizures and Brain Injury after Acute Intoxication of Diisopropylfluorophosphate, an Organophosphate Pesticide and Surrogate for Nerve Agents. J Pharmacol Exp Ther 2018; 367:302-321. [PMID: 30115757 DOI: 10.1124/jpet.117.247106] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 08/14/2018] [Indexed: 12/15/2022] Open
Abstract
Organophosphates (OP) such as the pesticide diisopropylfluorophosphate (DFP) and the nerve agent sarin are lethal chemicals that induce seizures, status epilepticus (SE), and brain damage. Midazolam, a benzodiazepine modulator of synaptic GABA-A receptors, is currently considered as a new anticonvulsant for nerve agents. Here, we characterized the time course of protective efficacy of midazolam (0.2-5 mg/kg, i.m.) in rats exposed to DFP, a chemical threat agent and surrogate for nerve agents. Behavioral and electroencephalogram (EEG) seizures were monitored for 24 hours after DFP exposure. The extent of brain injury was determined 3 days after DFP exposure by unbiased stereologic analyses of valid markers of neurodegeneration and neuroinflammation. Seizures were elicited within ∼8 minutes after DFP exposure that progressively developed into persistent SE lasting for hours. DFP exposure resulted in massive neuronal injury or necrosis, neurodegeneration of principal cells and interneurons, and neuroinflammation as evident by extensive activation of microglia and astrocytes in the hippocampus, amygdala, and other brain regions. Midazolam controlled seizures, neurodegeneration, and neuroinflammation when given early (10 minutes) after DFP exposure, but it was less effective when given at 40 minutes or later. Delayed therapy (≥40 minutes), a simulation of the practical therapeutic window for first responders or hospital admission, was associated with reduced seizure protection and neuroprotection. These results strongly reaffirm that the DFP-induced seizures and brain damage are progressively resistant to delayed treatment with midazolam, confirming the benzodiazepine refractory SE after OP intoxication. Thus, novel anticonvulsants superior to midazolam or adjunct therapies that enhance its efficacy are needed for effective treatment of refractory SE.
Collapse
Affiliation(s)
- Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center College of Medicine, Bryan, Texas
| | - Ramkumar Kuruba
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center College of Medicine, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center College of Medicine, Bryan, Texas
| |
Collapse
|
43
|
Apland JP, Aroniadou-Anderjaska V, Figueiredo TH, Pidoplichko VI, Rossetti K, Braga MFM. Comparing the Antiseizure and Neuroprotective Efficacy of LY293558, Diazepam, Caramiphen, and LY293558-Caramiphen Combination against Soman in a Rat Model Relevant to the Pediatric Population. J Pharmacol Exp Ther 2018; 365:314-326. [PMID: 29467308 PMCID: PMC5878669 DOI: 10.1124/jpet.117.245969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
The currently Food and Drug Administration-approved anticonvulsant for the treatment of status epilepticus (SE) induced by nerve agents is the benzodiazepine diazepam; however, diazepam does not appear to offer neuroprotective benefits. This is of particular concern with respect to the protection of children because, in the developing brain, synaptic transmission mediated via GABAA receptors, the target of diazepam, is weak. In the present study, we exposed 21-day-old male rats to 1.2 × LD50 soman and compared the antiseizure, antilethality, and neuroprotective efficacy of diazepam (10 mg/kg), LY293558 (an AMPA/GluK1 receptor antagonist; 15 mg/kg), caramiphen (CRM, an antimuscarinic with NMDA receptor-antagonistic properties; 50 mg/kg), and LY293558 (15 mg/kg) + CRM (50 mg/kg), administered 1 hour after exposure. Diazepam, LY293558, and LY293558 + CRM, but not CRM alone, terminated SE; LY293558 + CRM treatment acted significantly faster and produced a survival rate greater than 85%. Thirty days after soman exposure, neurodegeneration in limbic regions was most severe in the CRM-treated group, minimal to severe-depending on the region-in the diazepam group, absent to moderate in the LY293558-treated group, and totally absent in the LY293558 + CRM group. Amygdala and hippocampal atrophy, a severe reduction in spontaneous inhibitory activity in the basolateral amygdala, and increased anxiety-like behavior in the open-field and acoustic startle response tests were present in the diazepam and CRM groups, whereas the LY293558 and LY293558 + CRM groups did not differ from controls. The combined administration of LY293558 and CRM, by blocking mainly AMPA, GluK1, and NMDA receptors, is a very effective anticonvulsant and neuroprotective therapy against soman in young rats.
Collapse
Affiliation(s)
- James P Apland
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Vassiliki Aroniadou-Anderjaska
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Taiza H Figueiredo
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Volodymyr I Pidoplichko
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Katia Rossetti
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Maria F M Braga
- Neuroscience Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.); and Departments of Anatomy, Physiology, and Genetics (V.A.-A., T.H.F., V.I.P., K.R., M.F.M.B.) and Psychiatry (V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
44
|
Rojas A, Wang W, Glover A, Manji Z, Fu Y, Dingledine R. Beneficial Outcome of Urethane Treatment Following Status Epilepticus in a Rat Organophosphorus Toxicity Model. eNeuro 2018; 5:ENEURO.0070-18.2018. [PMID: 29766039 PMCID: PMC5952304 DOI: 10.1523/eneuro.0070-18.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/07/2023] Open
Abstract
The efficacy of benzodiazepines to terminate electrographic status epilepticus (SE) declines the longer a patient is in SE. Therefore, alternative methods for ensuring complete block of SE and refractory SE are necessary. We compared the ability of diazepam and a subanesthetic dose of urethane to terminate prolonged SE and mitigate subsequent pathologies. Adult Sprague Dawley rats were injected with diisopropylfluorophosphate (DFP) to induce SE. Rats were administered diazepam (10 mg/kg, ip) or urethane (0.8 g/kg, s.c.) 1 h after DFP-induced SE and compared to rats that experienced uninterrupted SE. Large-amplitude and high-frequency spikes induced by DFP administration were quenched for at least 46 h in rats administered urethane 1 h after SE onset as demonstrated by cortical electroencephalography (EEG). By contrast, diazepam interrupted SE but seizures with high power in the 20- to 70-Hz band returned 6-10 h later. Urethane was more effective than diazepam at reducing hippocampal neurodegeneration, brain inflammation, gliosis and weight loss as measured on day 4 after SE. Furthermore, rats administered urethane displayed a 73% reduction in the incidence of spontaneous recurrent seizures after four to eight weeks and a 90% reduction in frequency of seizures in epileptic rats. By contrast, behavioral changes in the light/dark box, open field and a novel object recognition task were not improved by urethane. These findings indicate that in typical rodent SE models, it is the return of SE overnight, and not the initially intense 1-2 h of SE experience, that is largely responsible for neurodegeneration, accompanying inflammation, and the subsequent development of epilepsy.
Collapse
Affiliation(s)
- Asheebo Rojas
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Wenyi Wang
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Avery Glover
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Zahra Manji
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Yujiao Fu
- Department of Pharmacology, Emory University, Atlanta, GA 30322
| | | |
Collapse
|
45
|
McCarren HS, Arbutus JA, Ardinger C, Dunn EN, Jackson CE, McDonough JH. Dexmedetomidine stops benzodiazepine-refractory nerve agent-induced status epilepticus. Epilepsy Res 2018; 141:1-12. [PMID: 29414381 DOI: 10.1016/j.eplepsyres.2018.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/21/2017] [Accepted: 01/07/2018] [Indexed: 12/15/2022]
Abstract
Nerve agents are highly toxic chemicals that pose an imminent threat to soldiers and civilians alike. Nerve agent exposure leads to an increase in acetylcholine within the central nervous system, resulting in development of protracted seizures known as status epilepticus (SE). Currently, benzodiazepines are the standard of care for nerve agent-induced SE, but their efficacy quickly wanes as the time to treatment increases. Here, we examine the role of the α2-adrenoceptor in termination of nerve agent-induced SE using the highly specific agonist dexmedetomidine (DEX). Adult male rats were exposed to soman and entered SE as confirmed by electroencephalograph (EEG). We observed that administration of DEX in combination with the benzodiazepine midazolam (MDZ) 20 or 40 min after the onset of SE stopped seizures and returned processed EEG measurements to baseline levels. The protective effect of DEX was blocked by the α2-adrenoceptor antagonist atipamezole (ATI), but ATI failed to restore seizure activity after it was already halted by DEX in most cases, suggesting that α2-adrenoceptors may be involved in initiating SE cessation rather than merely suppressing seizure activity. Histologically, treatment with DEX + MDZ significantly reduced the number of dying neurons as measured by FluoroJade B in the amygdala, thalamus, and piriform cortex, but did not protect the hippocampus or parietal cortex even when SE was successfully halted. We conclude that DEX serves not just as a valuable potential addition to the anticonvulsant regimen for nerve agent exposure, but also as a tool for dissecting the neural circuitry that drives SE.
Collapse
Affiliation(s)
- Hilary S McCarren
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States.
| | - Julia A Arbutus
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Cherish Ardinger
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Emily N Dunn
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Cecelia E Jackson
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - John H McDonough
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| |
Collapse
|
46
|
Krishnan JK, Figueiredo TH, Moffett JR, Arun P, Appu AP, Puthillathu N, Braga MF, Flagg T, Namboodiri AM. Brief isoflurane administration as a post-exposure treatment for organophosphate poisoning. Neurotoxicology 2017; 63:84-89. [DOI: 10.1016/j.neuro.2017.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/19/2017] [Accepted: 09/18/2017] [Indexed: 10/18/2022]
|
47
|
Hobson BA, Rowland DJ, Supasai S, Harvey DJ, Lein PJ, Garbow JR. A magnetic resonance imaging study of early brain injury in a rat model of acute DFP intoxication. Neurotoxicology 2017; 66:170-178. [PMID: 29183789 DOI: 10.1016/j.neuro.2017.11.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 11/18/2022]
Abstract
Current treatments for seizures induced by organophosphates do not protect sufficiently against progressive neurodegeneration or delayed cognitive impairment. Developing more effective therapeutic approaches has been challenging because the pathogenesis of these delayed consequences is poorly defined. Using magnetic resonance imaging (MRI), we previously reported brain lesions that persist for months in a rat model of acute intoxication with the OP, diisopropylfluorophosphate (DFP). However, the early spatiotemporal progression of these lesions remains unknown. To address this data gap, we used in vivo MRI to longitudinally monitor brain lesions during the first 3 d following acute DFP intoxication. Adult male Sprague Dawley rats acutely intoxicated with DFP (4mg/kg, sc) were MR imaged at 6, 12, 18, 24, 48, 72h post-DFP, and their brains then taken for correlative histology to assess neurodegeneration using FluoroJade C (FJC) staining. Acute DFP intoxication elicited moderate-to-severe seizure activity. T2-weighted (T2w) anatomic imaging revealed prominent lesions within the thalamus, piriform cortex, cerebral cortex, hippocampus, corpus striatum, and substantia nigra that corresponded to neurodegeneration, evident as bands of FJC positive cells. Semi-quantitative assessment of lesion severity demonstrated significant regional variation in the onset and progression of injury, and suggested that lesion severity may be modulated by isoflurane anesthesia. These results imply that the timing of therapeutic intervention for attenuating brain injury following OP intoxication may be regionally dependent, and that longitudinal assessment of OP-induced damage by MRI may be a powerful tool for assessing therapeutic response.
Collapse
Affiliation(s)
- Brad A Hobson
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, United States.
| | - Douglas J Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA, 95616, United States.
| | - Suangsuda Supasai
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, United States.
| | - Danielle J Harvey
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, CA, 95616, United States.
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, United States.
| | - Joel R Garbow
- Biomedical Magnetic Resonance Laboratory, Mallinckrodt Institute of Radiology, School of Medicine,Washington University in St. Louis, St. Louis, MO, 63110, United States.
| |
Collapse
|
48
|
Apland JP, Aroniadou-Anderjaska V, Figueiredo TH, Prager EM, Olsen CH, Braga MFM. Susceptibility to Soman Toxicity and Efficacy of LY293558 Against Soman-Induced Seizures and Neuropathology in 10-Month-Old Male Rats. Neurotox Res 2017; 32:694-706. [PMID: 28776308 DOI: 10.1007/s12640-017-9789-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/13/2017] [Accepted: 07/21/2017] [Indexed: 12/23/2022]
Abstract
Acute nerve agent exposure causes prolonged status epilepticus (SE), leading to death or long-term brain damage. We have previously demonstrated that LY293558, an AMPA/GluK1 kainate receptor antagonist, terminates SE induced by the nerve agent soman and protects from long-term brain damage, in immature rats and young-adult rats, even if administered with a relatively long latency from the time of exposure. However, susceptibility to the lethal consequences of SE increases with age, and mortality by SE induced by soman is substantially greater in older animals. Therefore, in the present study, we compared the susceptibility to soman toxicity of 10-month-old male rats with that of young-adult male rats (42 to 50 days old) and examined the protective efficacy of LY293558 in the older group. A lower percentage of the 10-month-old rats developed SE after injection of 1.2 × LD50 soman, compared to the young adults, the latency to seizure onset was longer in the older rats, and seizure intensity did not differ between the two age groups. However, mortality rate in the older rats who developed SE was higher than in the young adults. Acetylcholinesterase activity in the amygdala, hippocampus, and piriform cortex did not differ between the two age groups. Administration of LY293558 at 20 or 60 min post-exposure suppressed SE, increased 24-h survival rate, decreased the long-term risk of death, reduced neuronal degeneration in the amygdala, hippocampus, piriform, and entorhinal cortices, and facilitated recovery from body weight loss. Thus, LY293558 is an effective countermeasure against soman toxicity also in older animals.
Collapse
Affiliation(s)
- James P Apland
- Neuroscience Program, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Aberdeen, MD, 21010, USA
| | - Vassiliki Aroniadou-Anderjaska
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Eric M Prager
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.,John Wiley and Sons, Inc., 111 River Street, Hoboken, NJ, 07030, USA
| | - Cara H Olsen
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA. .,Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
49
|
Althaus AL, McCarren HS, Alqazzaz A, Jackson C, McDonough JH, Smith CD, Hoffman E, Hammond RS, Robichaud AJ, Doherty JJ. The synthetic neuroactive steroid SGE-516 reduces status epilepticus and neuronal cell death in a rat model of soman intoxication. Epilepsy Behav 2017; 68:22-30. [PMID: 28109985 DOI: 10.1016/j.yebeh.2016.12.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/18/2016] [Accepted: 12/17/2016] [Indexed: 12/18/2022]
Abstract
Organophosphorus nerve agents (OPNAs) are irreversible inhibitors of acetylcholinesterase that pose a serious threat to public health because of their use as chemical weapons. Exposure to high doses of OPNAs can dramatically potentiate cholinergic synaptic activity and cause status epilepticus (SE). Current standard of care for OPNA exposure involves treatment with cholinergic antagonists, oxime cholinesterase reactivators, and benzodiazepines. However, data from pre-clinical models suggest that OPNA-induced SE rapidly becomes refractory to benzodiazepines. Neuroactive steroids (NAS), such as allopregnanolone, retain anticonvulsant activity in rodent models of benzodiazepine-resistant SE, perhaps because they modulate a broader variety of GABAA receptor subtypes. SGE-516 is a novel, next generation NAS and a potent and selective GABAA receptor positive allosteric modulator (PAM). The present study first established that SGE-516 reduced electrographic seizures in the rat lithium-pilocarpine model of pharmacoresistant SE. Then the anticonvulsant activity of SGE-516 was investigated in the soman-intoxication model of OPNA-induced SE. SGE-516 (5.6, 7.5, and 10mg/kg, IP) significantly reduced electrographic seizure activity compared to control when administered 20min after SE onset. When 10mg/kg SGE-516 was administered 40min after SE onset, seizure activity was still significantly reduced compared to control. In addition, all cohorts of rats treated with SGE-516 exhibited significantly reduced neuronal cell death as measured by FluoroJade B immunohistochemistry. These data suggest synthetic NASs that positively modulate both synaptic and extrasynaptic GABAA receptors may be candidates for further study in the treatment of OPNA-induced SE.
Collapse
Affiliation(s)
| | - Hilary S McCarren
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Aymen Alqazzaz
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Cecelia Jackson
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - John H McDonough
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Carl D Smith
- United States Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Ethan Hoffman
- Drug Discovery, Sage Therapeutics, Inc., Cambridge, MA, USA
| | | | | | | |
Collapse
|
50
|
Iyengar ARS, Pande AH. Organophosphate-Hydrolyzing Enzymes as First-Line of Defence Against Nerve Agent-Poisoning: Perspectives and the Road Ahead. Protein J 2016; 35:424-439. [DOI: 10.1007/s10930-016-9686-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|