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] [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
|
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
|
4
|
Moghe M, Kim SS, Guan M, Rait A, Pirollo KF, Harford JB, Chang EH. scL-2PAM: A Novel Countermeasure That Ameliorates Neuroinflammation and Neuronal Losses in Mice Exposed to an Anticholinesterase Organophosphate. Int J Mol Sci 2024; 25:7539. [PMID: 39062781 PMCID: PMC11276659 DOI: 10.3390/ijms25147539] [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: 05/26/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Due to their inhibition of acetylcholinesterase, organophosphates are among the most toxic of chemicals. Pralidoxime (a.k.a 2-PAM) is the only acetylcholinesterase reactivator approved in the U.S., but 2-PAM only poorly traverses the blood-brain barrier. Previously, we have demonstrated that scL-2PAM, a nanoformulation designed to enter the brain via receptor-mediated transcytosis, is superior to unencapsulated 2-PAM for reactivating brain acetylcholinesterase, ameliorating cholinergic crisis, and improving survival rates for paraoxon-exposed mice. Here, we employ histology and transcriptome analyses to assess the ability of scL-2PAM to prevent neurological sequelae including microglial activation, expression of inflammatory cytokines, and ultimately loss of neurons in mice surviving paraoxon exposures. Levels of the mRNA encoding chemokine ligand 2 (CCL2) were significantly upregulated after paraoxon exposures, with CCL2 mRNA levels in the brain correlating well with the intensity and duration of cholinergic symptoms. Our nanoformulation of 2-PAM was found to be superior to unencapsulated 2-PAM in reducing the levels of the CCL2 transcript. Moreover, brain histology revealed that scL-2PAM was more effective than unencapsulated 2-PAM in preventing microglial activation and the subsequent loss of neurons. Thus, scL-2PAM appears to be a new and improved countermeasure for reducing neuroinflammation and mitigating brain damage in survivors of organophosphate exposures.
Collapse
Affiliation(s)
- Manish Moghe
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | - Sang-Soo Kim
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
- SynerGene Therapeutics, Inc., Potomac, MD 20854, USA;
| | - Miaoyin Guan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | - Antonina Rait
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | - Kathleen F. Pirollo
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | | | - Esther H. Chang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Nguyen DA, Stone MF, Schultz CR, de Araujo Furtado M, Niquet J, Wasterlain CG, Lumley LA. Evaluation of Midazolam-Ketamine-Allopregnanolone Combination Therapy against Cholinergic-Induced Status Epilepticus in Rats. J Pharmacol Exp Ther 2024; 388:376-385. [PMID: 37770198 PMCID: PMC10801769 DOI: 10.1124/jpet.123.001784] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023] Open
Abstract
Status epilepticus (SE) is a life-threatening development of self-sustaining seizures that becomes resistant to benzodiazepines when treatment is delayed. Benzodiazepine pharmacoresistance is thought in part to result from internalization of synaptic GABAA receptors, which are the main target of the drug. The naturally occurring neurosteroid allopregnanolone is a therapy of interest against SE for its ability to modulate all isoforms of GABAA receptors. Ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, has been partially effective in combination with benzodiazepines in mitigating SE-associated neurotoxicity. In this study, allopregnanolone as an adjunct to midazolam or midazolam-ketamine combination therapy was evaluated for efficacy against cholinergic-induced SE. Adult male rats implanted with electroencephalographic (EEG) telemetry devices were exposed to the organophosphorus chemical (OP) soman (GD) and treated with an admix of atropine sulfate and HI-6 at 1 minute after exposure followed by midazolam, midazolam-allopregnanolone, or midazolam-ketamine-allopregnanolone 40 minutes after seizure onset. Neurodegeneration, neuronal loss, and neuroinflammation were assessed 2 weeks after GD exposure. Seizure activity, EEG power integral, and epileptogenesis were also compared among groups. Overall, midazolam-ketamine-allopregnanolone combination therapy was effective in reducing cholinergic-induced toxic signs and neuropathology, particularly in the thalamus and hippocampus. Higher dosage of allopregnanolone administered in combination with midazolam and ketamine was also effective in reducing EEG power integral and epileptogenesis. The current study reports that there is a promising potential of neurosteroids in combination with benzodiazepine and ketamine treatments in a GD model of SE. SIGNIFICANCE STATEMENT: Allopregnanolone, a naturally occurring neurosteroid, reduced pathologies associated with soman (GD) exposure such as epileptogenesis, neurodegeneration, and neuroinflammation, and suppressed GD-induced toxic signs when used as an adjunct to midazolam and ketamine in a delayed treatment model of soman-induced status epilepticus (SE) in rats. However, protection was incomplete, suggesting that further studies are needed to identify optimal combinations of antiseizure medications and routes of administration for maximal efficacy against cholinergic-induced SE.
Collapse
Affiliation(s)
- Donna A Nguyen
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland (D.A.N., M.F.S., C.R.S., L.A.L.); BioSEaD, LLC, Rockville, Maryland (M.D.A.F.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (J.N., C.G.W.); and Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California (J.N., C.G.W.)
| | - Michael F Stone
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland (D.A.N., M.F.S., C.R.S., L.A.L.); BioSEaD, LLC, Rockville, Maryland (M.D.A.F.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (J.N., C.G.W.); and Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California (J.N., C.G.W.)
| | - Caroline R Schultz
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland (D.A.N., M.F.S., C.R.S., L.A.L.); BioSEaD, LLC, Rockville, Maryland (M.D.A.F.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (J.N., C.G.W.); and Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California (J.N., C.G.W.)
| | - Marcio de Araujo Furtado
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland (D.A.N., M.F.S., C.R.S., L.A.L.); BioSEaD, LLC, Rockville, Maryland (M.D.A.F.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (J.N., C.G.W.); and Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California (J.N., C.G.W.)
| | - Jerome Niquet
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland (D.A.N., M.F.S., C.R.S., L.A.L.); BioSEaD, LLC, Rockville, Maryland (M.D.A.F.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (J.N., C.G.W.); and Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California (J.N., C.G.W.)
| | - Claude G Wasterlain
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland (D.A.N., M.F.S., C.R.S., L.A.L.); BioSEaD, LLC, Rockville, Maryland (M.D.A.F.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (J.N., C.G.W.); and Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California (J.N., C.G.W.)
| | - Lucille A Lumley
- Neuroscience Department, US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland (D.A.N., M.F.S., C.R.S., L.A.L.); BioSEaD, LLC, Rockville, Maryland (M.D.A.F.); Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California (J.N., C.G.W.); and Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California (J.N., C.G.W.)
| |
Collapse
|
8
|
Singh T, Ramakrishnan S, Wu X, Reddy DS. A Pediatric Rat Model of Organophosphate-Induced Refractory Status Epilepticus: Characterization of Long-Term Epileptic Seizure Activity, Neurologic Dysfunction and Neurodegeneration. J Pharmacol Exp Ther 2024; 388:416-431. [PMID: 37977810 PMCID: PMC10801778 DOI: 10.1124/jpet.123.001794] [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: 06/14/2023] [Revised: 09/09/2023] [Accepted: 09/26/2023] [Indexed: 11/19/2023] Open
Abstract
Children are highly vulnerable to the neurotoxic effects of organophosphates (OPs), which can cause neuronal developmental defects, including intellectual disability, autism, epilepsy, and related comorbidities. Unfortunately, no specific pediatric OP neurotoxicity model currently exists. In this study, we developed and characterized a pediatric rat model of status epilepticus (SE) induced by the OP diisopropylfluorophosphate (DFP) and examined its impact on long-term neurological outcomes. Postnatal day 21 rats were exposed to a DFP regimen with standard antidotes. Progressive behavioral deteriorations were assessed over a three-month period. Development of epileptic seizures, ictal discharges, high-frequency oscillations (HFOs), and interictal spikes were monitored by video-electroencephalography recordings. Histology-stereology analysis was performed to assess neurodegeneration, neuroinflammation, and morphologic abnormalities. DFP-exposed, post-SE animals exhibited significantly elevated levels of anxiety and depression than age-matched controls at 1, 2, and 3 months post-exposure. DFP-exposed animals displayed aggressive behavior and a marked decline in object recognition memory, as well as prominent impairment in spatial learning and memory. DFP-exposed animals had striking electrographic abnormalities with the occurrence of displayed epileptic seizures, ictal discharges, HFOs, and interictal spikes, suggesting chronic epilepsy. Neuropathological analysis showed substantially fewer principal neurons and inhibitory interneurons with a marked increase in reactive microglia and neuroinflammation in the hippocampus and other brain regions. DFP-exposed animals also exhibited mossy fiber sprouting indicating impaired network formations. Long-term epileptic seizures and neuropsychiatric functional deficits induced by DFP were consistent with neuropathological defects. Collectively, this pediatric model displays many hallmarks of chronic sequelae reminiscent of children exposed to OPs, suggesting that it will be a valuable tool for investigating pathologic mechanisms and potential treatment strategies to attenuate long-term OP neurotoxicity. SIGNIFICANCE STATEMENT: Millions of children are exposed to organophosphates (OPs) used in agriculture or chemical incidents. This study investigated the long-term impact of neonatal exposure to the OP chemical diisopropylfluorophosphate (DFP) on neurobehavioral and neurodevelopmental outcomes in adulthood. DFP exposure caused long-lasting behavioral abnormalities, epileptic seizures, and bilateral brain defects with an array of neurological sequelae seen in children's OP neurotoxicity. Thus, this model provides a novel tool to explore therapeutic interventions that mitigate long-term neurotoxic effects of children exposed to OP-induced seizures and status epilepticus.
Collapse
Affiliation(s)
- Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (T.S., S.R., X.W., D.S.R.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (T.S., S.R., X.W., D.S.R.)
| | - Sreevidhya Ramakrishnan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (T.S., S.R., X.W., D.S.R.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (T.S., S.R., X.W., D.S.R.)
| | - Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (T.S., S.R., X.W., D.S.R.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (T.S., S.R., X.W., D.S.R.)
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, Texas (T.S., S.R., X.W., D.S.R.) and Institute of Pharmacology and Neurotherapeutics, Texas A&M University Health Science Center, Bryan, Texas (T.S., S.R., X.W., D.S.R.)
| |
Collapse
|
9
|
Vasanthi SS, Rao NS, Samidurai M, Massey N, Meyer C, Gage M, Kharate M, Almanza A, Wachter L, Mafuta C, Trevino L, Carlo AM, Bryant E, Corson BE, Wohlgemuth M, Ostrander M, Showman L, Wang C, Thippeswamy T. Disease-modifying effects of a glial-targeted inducible nitric oxide synthase inhibitor (1400W) in mixed-sex cohorts of a rat soman (GD) model of epilepsy. J Neuroinflammation 2023; 20:163. [PMID: 37438764 PMCID: PMC10337207 DOI: 10.1186/s12974-023-02847-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Acute exposure to seizurogenic organophosphate (OP) nerve agents (OPNA) such as diisopropylfluorophosphate (DFP) or soman (GD), at high concentrations, induce immediate status epilepticus (SE), reactive gliosis, neurodegeneration, and epileptogenesis as a consequence. Medical countermeasures (MCMs-atropine, oximes, benzodiazepines), if administered in < 20 min of OPNA exposure, can control acute symptoms and mortality. However, MCMs alone are inadequate to prevent OPNA-induced brain injury and behavioral dysfunction in survivors. We have previously shown that OPNA exposure-induced SE increases the production of inducible nitric oxide synthase (iNOS) in glial cells in both short- and long- terms. Treating with a water soluble and highly selective iNOS inhibitor, 1400W, for 3 days significantly reduced OPNA-induced brain changes in those animals that had mild-moderate SE in the rat DFP model. However, such mitigating effects and the mechanisms of 1400W are unknown in a highly volatile nerve agent GD exposure. METHODS Mixed-sex cohort of adult Sprague Dawley rats were exposed to GD (132 μg/kg, s.c.) and immediately treated with atropine (2 mg/kg, i.m) and HI-6 (125 mg/kg, i.m.). Severity of seizures were quantified for an hour and treated with midazolam (3 mg/kg, i.m.). An hour post-midazolam, 1400W (20 mg/kg, i.m.) or vehicle was administered daily for 2 weeks. After behavioral testing and EEG acquisition, animals were euthanized at 3.5 months post-GD. Brains were processed for neuroinflammatory and neurodegeneration markers. Serum and CSF were used for nitrooxidative and proinflammatory cytokines assays. RESULTS We demonstrate a significant long-term (3.5 months post-soman) disease-modifying effect of 1400W in animals that had severe SE for > 20 min of continuous convulsive seizures. 1400W significantly reduced GD-induced motor and cognitive dysfunction; nitrooxidative stress (nitrite, ROS; increased GSH: GSSG); proinflammatory cytokines in the serum and some in the cerebrospinal fluid (CSF); epileptiform spikes and spontaneously recurring seizures (SRS) in males; reactive gliosis (GFAP + C3 and IBA1 + CD68-positive glia) as a measure of neuroinflammation, and neurodegeneration (especially parvalbumin-positive neurons) in some brain regions. CONCLUSION These findings demonstrate the long-term disease-modifying effects of a glial-targeted iNOS inhibitor, 1400W, in a rat GD model by modulating reactive gliosis, neurodegeneration (parvalbumin-positive neurons), and neuronal hyperexcitability.
Collapse
Affiliation(s)
- Suraj S. Vasanthi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Nikhil S. Rao
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Manikandan Samidurai
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Nyzil Massey
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Christina Meyer
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Meghan Gage
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Mihir Kharate
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Aida Almanza
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Logan Wachter
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Candide Mafuta
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Lily Trevino
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Adriana M. Carlo
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Elijah Bryant
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Brooke E. Corson
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Morgan Wohlgemuth
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Morgan Ostrander
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Lucas Showman
- W.M. Keck Metabolomics Research Laboratory, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Chong Wang
- Department of Veterinary Diagnostic and Production Animal Medicine and Statistics, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, 50011 USA
| |
Collapse
|
10
|
Vasanthi SS, Rao NS, Samidurai M, Massey N, Meyer C, Gage M, Kharate M, Almanza A, Wachter L, Mafuta C, Trevino L, Carlo AM, Bryant E, Corson BE, Wohlgemuth M, Ostrander M, Wang C, Thippeswamy T. Disease-Modifying Effects of a Glial-targeted Inducible Nitric Oxide Synthase Inhibitor (1400W) in Mixed-sex Cohorts of a Rat Soman (GD) Model of Epilepsy. RESEARCH SQUARE 2023:rs.3.rs-2883247. [PMID: 37214912 PMCID: PMC10197763 DOI: 10.21203/rs.3.rs-2883247/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Background Acute exposure to seizurogenic organophosphate (OP) nerve agents (OPNA) such as diisopropylfluorophosphate (DFP) or soman (GD), at high concentrations, induce immediate status epilepticus (SE), reactive gliosis, neurodegeneration, and epileptogenesis as a consequence. Medical countermeasures (MCMs- atropine, oximes, benzodiazepines), if administered in < 20 minutes of OPNA exposure, can control acute symptoms and mortality. However, MCMs alone are inadequate to prevent OPNA-induced brain injury and behavioral dysfunction in survivors. We have previously shown that OPNA exposure-induced SE increases the production of inducible nitric oxide synthase (iNOS) in glial cells in both short- and long- terms. Treating with a water soluble and highly selective iNOS inhibitor, 1400W, for three days significantly reduced OPNA-induced brain changes in those animals that had mild-moderate SE in the rat DFP model. However, such mitigating effects and the mechanisms of 1400W are unknown in a highly volatile nerve agent GD exposure. Methods Mixed-sex cohort of adult Sprague Dawley rats were exposed to GD (132µg/kg, s.c.) and immediately treated with atropine (2mg/kg, i.m) and HI-6 (125mg/kg, i.m.). Severity of seizures were quantified for an hour and treated with midazolam (3mg/kg, i.m.). An hour post-midazolam, 1400W (20mg/kg, i.m.) or vehicle was administered daily for two weeks. After behavioral testing and EEG acquisition, animals were euthanized at 3.5 months post-GD. Brains were processed for neuroinflammatory and neurodegeneration markers. Serum and CSF were used for nitrooxidative and proinflammatory cytokines assays. Results We demonstrate a significant long-term (3.5 months post-soman) disease-modifying effect of 1400W in animals that had severe SE for > 20min of continuous convulsive seizures. 1400W significantly reduced GD-induced motor and cognitive dysfunction; nitrooxidative stress (nitrite, ROS; increased GSH: GSSG); proinflammatory cytokines in the serum and some in the cerebrospinal fluid (CSF); epileptiform spikes and spontaneously recurring seizures (SRS) in males; reactive gliosis (GFAP + C3 and IBA1 + CD68 positive glia) as a measure of neuroinflammation, and neurodegeneration (including parvalbumin positive neurons) in some brain regions. Conclusion These findings demonstrate the long-term disease-modifying effects of a glial-targeted iNOS inhibitor, 1400W, in a rat GD model by modulating reactive gliosis, neurodegeneration, and neuronal hyperexcitability.
Collapse
|
11
|
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
|
12
|
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
|
13
|
The Effects of Drying Techniques on Phytochemical Contents and Biological Activities on Selected Bamboo Leaves. Molecules 2022; 27:molecules27196458. [PMID: 36234995 PMCID: PMC9571890 DOI: 10.3390/molecules27196458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 09/24/2022] [Indexed: 11/07/2022] Open
Abstract
The therapeutic potential of bamboos has acquired global attention. Nonetheless, the biological activities of the plants are rarely considered due to limited available references in Sabah, Malaysia. Furthermore, the drying technique could significantly affect the retention and degradation of nutrients in bamboos. Consequently, the current study investigated five drying methods, namely, sun, shade, microwave, oven, and freeze-drying, of the leaves of six bamboo species, Bambusa multiplex, Bambusa tuldoides, Bambusa vulgaris, Dinochloa sublaevigata, Gigantochloa levis, and Schizostachyum brachycladum. The infused bamboo leaves extracts were analysed for their total phenolic content (TPC) and total flavonoid content (TFC). The antioxidant activities of the samples were determined via the 2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), and ferric reducing antioxidant power (FRAP) assays, whereas their toxicities were evaluated through the brine shrimp lethality assay (BSLA). The chemical constituents of the samples were determined using liquid chromatography−tandem mass spectrometry (LC-MS/MS). The freeze-drying method exhibited the highest phytochemical contents and antioxidant activity yield, excluding the B. vulgaris sample, in which the microwave-dried sample recorded the most antioxidant and phytochemical levels. The TPC and TFC results were within the 2.69 ± 0.01−12.59 ± 0.09 mg gallic acid equivalent (GAE)/g and 0.77 ± 0.01−2.12 ± 0.01 mg quercetin equivalent (QE)/g ranges, respectively. The DPPH and ABTS IC50 (half-maximal inhibitory concentration) were 2.92 ± 0.01−4.73 ± 0.02 and 1.89−0.01 to 3.47 ± 0.00 µg/mL, respectively, indicating high radical scavenging activities. The FRAP values differed significantly between the drying methods, within the 6.40 ± 0.12−36.65 ± 0.09 mg Trolox equivalent (TE)/g range. The phytochemical contents and antioxidant capacities exhibited a moderate correlation, revealing that the TPC and TFC were slightly responsible for the antioxidant activities. The toxicity assessment of the bamboo extracts in the current study demonstrated no toxicity against the BSLA based on the LC50 (lethal concentration 50) analysis at >1000 µg/mL. LC-MS analysis showed that alkaloid and pharmaceutical compounds influence antioxidant activities, as found in previous studies. The acquired information might aid in the development of bamboo leaves as functional food items, such as bamboo tea. They could also be investigated for their medicinal ingredients that can be used in the discovery of potential drugs.
Collapse
|
14
|
Rao NS, Meyer C, Vasanthi SS, Massey N, Samidurai M, Gage M, Putra M, Almanza AN, Wachter L, Thippeswamy T. DFP-Induced Status Epilepticus Severity in Mixed-Sex Cohorts of Adult Rats Housed in the Same Room: Behavioral and EEG Comparisons. Front Cell Dev Biol 2022; 10:895092. [PMID: 35620057 PMCID: PMC9127803 DOI: 10.3389/fcell.2022.895092] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 11/26/2022] Open
Abstract
Sex is a biological variable in experimental models. In our previous diisopropylfluorophosphate (DFP) studies, female rats required a higher dose of DFP to achieve a somewhat similar severity of status epilepticus (SE) as males. In those studies, male and female rats were bought separately from the same vendor, housed in different rooms, and the DFP used was from different batches. We had also shown that surgery for epidural electrodes implantation reduces the threshold for SE. Our recent study in the soman (GD) model using a mixed-sex cohort of rats housed individually but in the same room showed that females achieved significantly higher SE severity than males for the same dose of GD. In this study, we demonstrate that housing the mixed-sex cohorts in the same room and treating them with DFP (4 mg/kg, s.c.) from the same pool, though from different batches, yielded reproducible SE severity in both sexes and both telemetry (surgery) and non-telemetry (non-surgery) groups. We conducted experiments in four mixed-sex cohorts of adult Sprague-Dawley rats. In females, the surgery for implanting the telemetry devices reduced the latency to convulsive seizure (CS) and increased SE severity compared to non-telemetry females. However, there were no sex differences in latency or SE severity within telemetry or non-telemetry groups. Once animals reached CS stage ≥3, they remained in CS stage in both sexes until midazolam was administered. Midazolam (3 mg/kg, i.m.) treatment 1-one-hour post-DFP significantly reduced epileptiform spikes in both sexes. The mortality was only 2% in 24 h. Irrespective of sex or stage of estrous cycle or surgery, the animals had continuous convulsive SE for ∼40 min. In telemetry rats, electrographic changes correlated with behavioral seizures. However, there was a significant difference in SE severity and the latency between directly-observed behavioral CS and EEG-based CS quantification in both sexes. Overall, these results suggest that housing both sexes in the same room and treating with DFP in a mixed-sex cohort from the same pool of reagents will minimize variability in SE severity. Such rigorous experiments will yield better outcomes while testing disease-modifying agents in epilepsy models.
Collapse
Affiliation(s)
- Nikhil S Rao
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Christina Meyer
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Suraj S Vasanthi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Nyzil Massey
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Manikandan Samidurai
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Meghan Gage
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Marson Putra
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Aida N Almanza
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Logan Wachter
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
15
|
Gage M, Putra M, Gomez-Estrada C, Golden M, Wachter L, Gard M, Thippeswamy T. Differential Impact of Severity and Duration of Status Epilepticus, Medical Countermeasures, and a Disease-Modifier, Saracatinib, on Brain Regions in the Rat Diisopropylfluorophosphate Model. Front Cell Neurosci 2021; 15:772868. [PMID: 34720886 PMCID: PMC8555467 DOI: 10.3389/fncel.2021.772868] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/29/2022] Open
Abstract
Acute organophosphate (OP) toxicity poses a significant threat to both military and civilian personnel as it can lead to a variety of cholinergic symptoms including the development of status epilepticus (SE). Depending on its severity, SE can lead to a spectrum of neurological changes including neuroinflammation and neurodegeneration. In this study, we determined the impact of SE severity and duration on disease promoting parameters such as gliosis and neurodegeneration and the efficacy of a disease modifier, saracatinib (AZD0530), a Src/Fyn tyrosine kinase inhibitor. Animals were exposed to 4 mg/kg diisopropylfluorophosphate (DFP, s.c.) followed by medical countermeasures. We had five experimental groups: controls (no DFP), animals with no continuous convulsive seizures (CS), animals with ∼20-min continuous CS, 31-60-min continuous CS, and > 60-min continuous CS. These groups were then assessed for astrogliosis, microgliosis, and neurodegeneration 8 days after DFP exposure. The 31-60-min and > 60-min groups, but not ∼20-min group, had significantly upregulated gliosis and neurodegeneration in the hippocampus compared to controls. In the piriform cortex and amygdala, however, all three continuous CS groups had significant upregulation in both gliosis and neurodegeneration. In a separate cohort of animals that had ∼20 and > 60-min of continuous CS, we administered saracatinib for 7 days beginning three hours after DFP. There was bodyweight loss and mortality irrespective of the initial SE severity and duration. However, in survived animals, saracatinib prevented spontaneous recurrent seizures (SRS) during the first week in both severity groups. In the ∼20-min CS group, compared to the vehicle, saracatinib significantly reduced neurodegeneration in the piriform cortex and amygdala. There were no significant differences in the measured parameters between the naïve control and saracatinib on its own (without DFP) groups. Overall, this study demonstrates the differential effects of the initial SE severity and duration on the localization of gliosis and neurodegeneration. We have also demonstrated the disease-modifying potential of saracatinib. However, its’ dosing regimen should be optimized based on initial severity and duration of CS during SE to maximize therapeutic effects and minimize toxicity in the DFP model as well as in other OP models such as soman.
Collapse
Affiliation(s)
- Meghan Gage
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States.,Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States
| | - Marson Putra
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States.,Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States
| | - Crystal Gomez-Estrada
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Madison Golden
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Logan Wachter
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Megan Gard
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States.,Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States
| |
Collapse
|
16
|
Cornelissen AS, Garcia EE, Raulli RE, Laney J, Joosen MJA. A delayed treatment model for the evaluation of scopolamine for VX nerve agent intoxication. Toxicol Appl Pharmacol 2021; 427:115650. [PMID: 34273408 DOI: 10.1016/j.taap.2021.115650] [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: 04/04/2021] [Revised: 06/24/2021] [Accepted: 07/12/2021] [Indexed: 11/18/2022]
Abstract
Most research on medical countermeasures for nerve agent exposure assumes a military scenario, in which (autoinjector) treatment is envisaged to be available immediately. In a civilian setting however, treatment is delayed until arrival of first-aid responders. This may significantly affect treatment efficacy and the requirements for secondary intensive care. The aim of the current study was to develop a guinea pig model to evaluate the efficacy of delayed treatment following nerve agent exposure. We identified a trigger-to-treat based on a progressive stage of the toxidrome following VX exposure, which was associated with the subsiding of clonic movements. This paradigm resulted in treatment consistently being administered between 15 and 25 min post-exposure. Using the model, we investigated the potential for the anticholinergic scopolamine to act as a delayed treatment either as a standalone treatment, or as an adjunct to delayed treatment with Standard of Care (SOC), containing atropine, 2-PAM, and midazolam. The study provides a framework for a small animal model for evaluating the efficacy of treatment administered at a specific stage of the toxidrome, when immediate treatment is absent. As an adjunct, scopolamine treatment did not result in improved survival, but did show a beneficial effect on recovery, in terms of general posture. As a standalone treatment, scopolamine showed a significant, dose-responsive, beneficial effect on survival and recovery. These promising results warrant additional studies to investigate which observed physiological improvements are relevant for the recovery process and residual injury.
Collapse
Affiliation(s)
- Alex S Cornelissen
- TNO Defence, Safety and Security, CBRN Protection, Rijswijk, the Netherlands.
| | - Efrain E Garcia
- Biomedical Advanced Research and Development Authority (BARDA), Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS), United States
| | - Robert E Raulli
- Biomedical Advanced Research and Development Authority (BARDA), Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS), United States
| | - Judith Laney
- Biomedical Advanced Research and Development Authority (BARDA), Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS), United States
| | - Marloes J A Joosen
- TNO Defence, Safety and Security, CBRN Protection, Rijswijk, the Netherlands
| |
Collapse
|
17
|
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: 19] [Impact Index Per Article: 6.3] [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
|
18
|
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: 4.5] [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
|
19
|
Acute administration of diazepam or midazolam minimally alters long-term neuropathological effects in the rat brain following acute intoxication with diisopropylfluorophosphate. Eur J Pharmacol 2020; 886:173538. [PMID: 32898549 DOI: 10.1016/j.ejphar.2020.173538] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/30/2022]
Abstract
Acute intoxication with organophosphorus cholinesterase inhibitors (OPs) can trigger seizures that rapidly progress to life-threatening status epilepticus. Diazepam, long considered the standard of care for treating OP-induced seizures, is being replaced by midazolam. Whether midazolam is more effective than diazepam in mitigating the persistent effects of acute OP intoxication has not been rigorously evaluated. We compared the efficacy of diazepam vs. midazolam in preventing persistent neuropathology in adult male Sprague-Dawley rats acutely intoxicated with the OP diisopropylfluorophosphate (DFP). Subjects were administered pyridostigmine bromide (0.1 mg/kg, i.p.) 30 min prior to injection with DFP (4 mg/kg, s.c.) or vehicle (saline) followed 1 min later by atropine sulfate (2 mg/kg, i.m.) and pralidoxime (25 mg/kg, i.m.), and 40 min later by diazepam (5 mg/kg, i.p.), midazolam (0.73 mg/kg, i.m.), or vehicle. At 3 and 6 months post-exposure, neurodegeneration, reactive astrogliosis, microglial activation, and oxidative stress were assessed in multiple brain regions using quantitative immunohistochemistry. Brain mineralization was evaluated by in vivo micro-computed tomography (micro-CT). Acute DFP intoxication caused persistent neurodegeneration, neuroinflammation, and brain mineralization. Midazolam transiently mitigated neurodegeneration, and both benzodiazepines partially protected against reactive astrogliosis in a brain region-specific manner. Neither benzodiazepine attenuated microglial activation or brain mineralization. These findings indicate that neither benzodiazepine effectively protects against persistent neuropathological changes, and suggest that midazolam is not significantly better than diazepam. Overall, this study highlights the need for improved neuroprotective strategies for treating humans in the event of a chemical emergency involving OPs.
Collapse
|
20
|
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: 1.8] [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
|
21
|
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
|
22
|
Mishra R, Geiling J. Chemical Agents in Disaster: Care and Management in the Intensive Care Unit. Crit Care Clin 2019; 35:633-645. [PMID: 31445610 DOI: 10.1016/j.ccc.2019.05.002] [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] [Indexed: 10/26/2022]
Abstract
Chemical agents of warfare are divided into lung agents, blood agents, vesicants, and nerve agents. Intensivists must familiarize themselves with the clinical presentation and management principles in the event of a chemical attack. Key principles in management include aggressive supportive care and early administration of specific antidotes, if available. Management includes proper personal protection for critical care providers. Patients may make complete recovery with aggressive supportive care, even if they appear to have a poor prognosis. Hospitals must have an emergency response disaster plan in place to deal with all potential causes of disasters, including illnesses resulting from chemical agents.
Collapse
Affiliation(s)
- Rashmi Mishra
- The Lung Center, Penn Highlands Healthcare, 100 Hospital Avenue, DuBois, PA 15801, USA.
| | - James Geiling
- Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA; Medical Service, VA Medical Center, White River Junction, VT 05009, USA
| |
Collapse
|
23
|
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: 2.0] [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
|
24
|
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: 1.2] [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
|
25
|
Time dependent dual effect of anti-inflammatory treatments on sarin-induced brain inflammation: Suggested role of prostaglandins. Neurotoxicology 2019; 74:19-27. [PMID: 31095963 DOI: 10.1016/j.neuro.2019.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/12/2019] [Accepted: 05/12/2019] [Indexed: 11/21/2022]
Abstract
A common consequence of exposure to organophosphate nerve agents is the centrally mediated seizure activity that appears even after conventional treatment with atropine and oximes. We have previously demonstrated a major inflammatory response with subsequent brain damage which was correlated with the duration of the sarin-induced seizures (Chapman et al., 2006). In the present work seizures were induced by the nerve agent sarin (1.2 LD50) insufficiently treated 1 min later by atropine and trimedoxime bromide (TA), with additional midazolam treatment either 5 or 30 min after continuous seizure activity. The efficacy of both steroidal and nonsteroidal anti-inflammatory drugs (NSAIDs), as well as other drugs that were reported as beneficial in neuroprotection, were evaluated for their contribution as adjunct treatment against sarin induced seizures and the ensuing inflammatory brain damage. Results show that both steroids and NSAIDs were harmful when administered during convulsions, and steroids were at best ineffective if administered at their termination. However, if administered at termination of convulsions, the NSAID ibuprofen, the selective COX 2 inhibitor nimesulide and the PLA2 inhibitor quinacrine were partially effective in reducing brain inflammatory markers. Administration of exogenous analogs of prostaglandins (PGE2) immediately following sarin-induced convulsions was found to have a beneficial effect in reducing brain inflammatory markers measured at 24 h and one week post sarin exposure. These findings support the hypothesis that elevated levels of PGE2 have a beneficial role immediately following sarin induced seizures, and that early inhibition of PGE2 production by both steroids and NSAID is contraindicative.
Collapse
|
26
|
Ríos C, Farfán-Briseño AC, Manjarrez-Marmolejo J, Franco-Pérez J, Méndez-Armenta M, Nava-Ruiz C, Caballero-Chacón S, Ruiz-Diaz A, Baron-Flores V, Díaz-Ruiz A. Efficacy of dapsone administered alone or in combination with diazepam to inhibit status epilepticus in rats. Brain Res 2019; 1708:181-187. [DOI: 10.1016/j.brainres.2018.12.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/13/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022]
|
27
|
Contingency Medical Countermeasures for Mass Nerve-Agent Exposure: Use of Pharmaceutical Alternatives to Community Stockpiled Antidotes. Disaster Med Public Health Prep 2018; 13:605-612. [PMID: 30319091 DOI: 10.1017/dmp.2018.99] [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: 11/07/2022]
Abstract
Having sufficient medical countermeasures (MCMs) available for the treatment of acetylcholinesterase-inhibiting nerve agent poisoned patients following a mass chemical exposure is a challenge for communities. After stockpiles containing auto-injectors are exhausted, communities need to be aware of alternative pharmaceutical options. The Department of Homeland Security Chemical Defense Program convened a federal interagency working group consisting of first responders, clinicians, and experts from the fields of medical toxicology, pharmacology, and emergency management. A literature review of pharmaceutical alternatives for treating nerve agent toxicity was performed. Pharmaceuticals that met the federal Public Health Emergency Medical Countermeasures Enterprise Product Specific Requirements were prioritized. Food and Drug Administration approval for one indication, market availability, and alignment to government procurement strategy were considered. This article summarizes the literature on comparative pharmacokinetics and efficacy against nerve agents (where available) of Food and Drug Administration approved drugs with muscarinic acetylcholine receptor antagonist and gamma-aminobutyric acid receptor agonist effects. This work is intended to serve as a resource of pharmaceutical options that may be available to communities (ie, emergency managers, planners, clinicians, and poison centers) when faced with a mass human exposure to a nerve agent and inadequate supplies of MCMs. (Disaster Med Public Health Preparedness. 2019;13:605-612).
Collapse
|
28
|
Bajgar J. Complex View on Poisoning with Nerve Agents and Organophosphates. ACTA MEDICA (HRADEC KRÁLOVÉ) 2018. [DOI: 10.14712/18059694.2018.23] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
OP/nerve agents are still considered as important chemicals acting on living organisms and widely used in human practice. Nerve agents are the most lethal chemical warfare agents. They are characterized according to their action as compounds influencing cholinergic nerve transmission via inhibition of AChE. The symptoms of intoxication comprise nicotinic, muscarinic and central symptoms, for some OP/nerve agents, a delayed neurotoxicity is observed. Cholinesterases (AChE and BuChE) are characterized as the main enzymes involved in the toxic effect of these compounds including their molecular forms. The activity of both enzymes (and molecular forms) is influenced by inhibitors and other factors such as pathological states. There are different methods for cholinesterase determination, however, the most frequent is the method based on the hydrolysis of thiocholine esters and following detection of free SH-group of the released thiocholine. The diagnosis of OP/nerve agents poisoning is based on anamnesis, the clinical status of the intoxicated organism and on cholinesterase determination in the blood. Some principles of prophylaxis against OP/nerve agents poisoning comprising the administration of reversible cholinesterase inhibitors such as pyridostigmine (alone or in combination with other drugs), scavengers such as preparations of cholinesterases, some therapeutic drugs and possible combinations are given. Basic principles of the treatment of nerve agents/OP poisoning are described. New drugs for the treatment are under experimental study based on new approaches to the mechanism of action.
Collapse
|
29
|
Drexler B, Seeger T, Worek F, Thiermann H, Antkowiak B, Grasshoff C. Midazolam is effective to reduce cortical network activity in organotypic cultures during severe cholinergic overstimulation with soman. Toxicol Lett 2018; 297:19-23. [PMID: 30165091 DOI: 10.1016/j.toxlet.2018.08.016] [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: 06/04/2018] [Revised: 08/14/2018] [Accepted: 08/25/2018] [Indexed: 12/13/2022]
Abstract
Intoxication with organophosphorus compounds can result in life-threatening organ dysfunction and refractory seizures. Sedation or hypnosis is essential to facilitate mechanical ventilation and control seizure activity. The range of indications for midazolam includes both hypnosis and seizure control. Since benzodiazepines cause sedation and hypnosis by dampening neuronal activity of the cerebral cortex, we investigated the drug's effect on action potential firing of cortical neurons in brain slices. Extensive cholinergic overstimulation was induced by increasing acetylcholine levels and simultaneously treating the slices with soman to block acetylcholinesterase activity. At control conditions midazolam reduced discharge rates (median/95% confidence interval) from 8.8 (7.0-10.5) Hz (in the absence of midazolam) to 2.2 (1.4-2.9) Hz (10 μM midazolam) and 1.6 (0.9-2.2) Hz (20 μM midazolam). Without midazolam, cholinergic overstimulation significantly enhanced neuronal activity to 13.1 (11.0-15.2) Hz. Midazolam attenuated firing rates during cholinergic overstimulation to 6.5 (4.8-8.2) Hz (10 μM midazolam) and 4.1 (3.3-6.0) Hz (20 μM midazolam), respectively. Thus, high cholinergic tone attenuated the drug's efficacy only moderately. These results suggest that midazolam is worth being tested as a promising drug to induce sedation and hypnosis in patients suffering from severe organophosphorous intoxication.
Collapse
Affiliation(s)
- Berthold Drexler
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072 Tuebingen, Germany.
| | - Thomas Seeger
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany.
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany.
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany.
| | - Bernd Antkowiak
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937 Munich, Germany; Werner Reichardt Center for Integrative Neuroscience, Eberhard-Karls-University, Tuebingen, Germany.
| | - Christian Grasshoff
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072 Tuebingen, Germany.
| |
Collapse
|
30
|
Figueiredo TH, Apland JP, Braga MFM, Marini AM. Acute and long-term consequences of exposure to organophosphate nerve agents in humans. Epilepsia 2018; 59 Suppl 2:92-99. [PMID: 30159887 DOI: 10.1111/epi.14500] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2018] [Indexed: 12/20/2022]
Abstract
Nerve agents are organophosphate (OP) compounds and among the most powerful poisons known to man. A terrorist attack on civilian or military populations causing mass casualties is a real threat. The OP nerve agents include soman, sarin, cyclosarin, tabun, and VX. The major mechanism of acute toxicity is the irreversible inhibition of acetylcholinesterase. Acetylcholinesterase inhibition results in the accumulation of excessive acetylcholine levels in synapses, leading to progression of toxic signs including hypersecretions, tremors, status epilepticus, respiratory distress, and death. Miosis and rhinorrhea are the most common clinical findings in those individuals acutely exposed to OP nerve agents. Prolonged seizures are responsible for the neuropathology. The brain region that shows the most severe damage is the amygdala, followed by the piriform cortex, hippocampus, cortex, thalamus, and caudate/putamen. Current medical countermeasures are only modestly effective in attenuating the seizures and neuropathology. Anticonvulsants such as benzodiazepines decrease seizure activity and improve outcome, but their efficacy depends upon the administration time after exposure to the nerve agent. Administration of benzodiazepines may increase the risk for seizure recurrence. Recent studies document long-term neurologic and behavior deficits, and technological advances demonstrate structural brain changes on magnetic resonance imaging.
Collapse
Affiliation(s)
- Taiza H Figueiredo
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - James P Apland
- Neuroscience Program, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ann M Marini
- Department of Neurology and Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
31
|
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: 62] [Impact Index Per Article: 10.3] [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
|
32
|
Niquet J, Baldwin R, Norman K, Suchomelova L, Lumley L, Wasterlain CG. Midazolam-ketamine dual therapy stops cholinergic status epilepticus and reduces Morris water maze deficits. Epilepsia 2016; 57:1406-15. [PMID: 27500978 DOI: 10.1111/epi.13480] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Pharmacoresistance remains an unsolved therapeutic challenge in status epilepticus (SE) and in cholinergic SE induced by nerve agent intoxication. SE triggers a rapid internalization of synaptic γ-aminobutyric acid A (GABAA ) receptors and externalization of N-methyl-d-aspartate (NMDA) receptors that may explain the loss of potency of standard antiepileptic drugs (AEDs). We hypothesized that a drug combination aimed at correcting the consequences of receptor trafficking would reduce SE severity and its long-term consequences. METHODS A severe model of SE was induced in adult Sprague-Dawley rats with a high dose of lithium and pilocarpine. The GABAA receptor agonist midazolam, the NMDA receptor antagonist ketamine, and/or the AED valproate were injected 40 min after SE onset in combination or as monotherapy. Measures of SE severity were the primary outcome. Secondary outcomes were acute neuronal injury, spontaneous recurrent seizures (SRS), and Morris water maze (MWM) deficits. RESULTS Midazolam-ketamine dual therapy was more efficient than double-dose midazolam or ketamine monotherapy or than valproate-midazolam or valproate-ketamine dual therapy in reducing several parameters of SE severity, suggesting a synergistic mechanism. In addition, midazolam-ketamine dual therapy reduced SE-induced acute neuronal injury, epileptogenesis, and MWM deficits. SIGNIFICANCE This study showed that a treatment aimed at correcting maladaptive GABAA receptor and NMDA receptor trafficking can stop SE and reduce its long-term consequences. Early midazolam-ketamine dual therapy may be superior to monotherapy in the treatment of benzodiazepine-refractory SE.
Collapse
Affiliation(s)
- Jerome Niquet
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Roger Baldwin
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Keith Norman
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Lucie Suchomelova
- Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A
| | - Lucille Lumley
- US Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen Proving Ground, Maryland, U.S.A
| | - Claude G Wasterlain
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A.,Epilepsy Research Laboratory (151), Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, U.S.A.,Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, U.S.A
| |
Collapse
|
33
|
Betten DP, Vohra RB, Cook MD, Matteucci MJ, Clark RF. Antidote Use in the Critically Ill Poisoned Patient. J Intensive Care Med 2016; 21:255-77. [PMID: 16946442 DOI: 10.1177/0885066606290386] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The proper use of antidotes in the intensive care setting when combined with appropriate general supportive care may reduce the morbidity and mortality associated with severe poisonings. The more commonly used antidotes that may be encountered in the intensive care unit ( N-acetylcysteine, ethanol, fomepizole, physostigmine, naloxone, flumazenil, sodium bicarbonate, octreotide, pyridoxine, cyanide antidote kit, pralidoxime, atropine, digoxin immune Fab, glucagon, calcium gluconate and chloride, deferoxamine, phytonadione, botulism antitoxin, methylene blue, and Crotaline snake antivenom) are reviewed. Proper indications for their use and knowledge of the possible adverse effects accompanying antidotal therapy will allow the physician to appropriately manage the severely poisoned patient.
Collapse
Affiliation(s)
- David P Betten
- Department of Emergency Medicine, Sparrow Health System, Michigan State University College of Human Medicine, Lansing, Michigan 48912-1811, USA.
| | | | | | | | | |
Collapse
|
34
|
Acon-Chen C, Koenig JA, Smith GR, Truitt AR, Thomas TP, Shih TM. Evaluation of acetylcholine, seizure activity and neuropathology following high-dose nerve agent exposure and delayed neuroprotective treatment drugs in freely moving rats. Toxicol Mech Methods 2016; 26:378-88. [PMID: 27329284 DOI: 10.1080/15376516.2016.1197992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Organophosphorus nerve agents such as soman (GD) inhibit acetylcholinesterase, producing an excess of acetylcholine (ACh), which results in respiratory distress, convulsions and status epilepticus that leads to neuropathology. Several drugs (topiramate, clobazam, pregnanolone, allopregnanolone, UBP 302, cyclopentyladenosine [CPA], ketamine, midazolam and scopolamine) have been identified as potential neuroprotectants that may terminate seizures and reduce brain damage. To systematically evaluate their efficacy, this study employed in vivo striatal microdialysis and liquid chromatography to respectively collect and analyze extracellular ACh in freely moving rats treated with these drugs 20 min after seizure onset induced by a high dose of GD. Along with microdialysis, EEG activity was recorded and neuropathology assessed at 24 h. GD induced a marked increase of ACh, which peaked at 30 min post-exposure to 800% of control levels and then steadily decreased toward baseline levels. Approximately 40 min after treatment, only midazolam (10 mg/kg) and CPA (60 mg/kg) caused a significant reduction of ACh levels, with CPA reducing ACh levels more rapidly than midazolam. Both drugs facilitated a return to baseline levels at least 55 min after treatment. At 24 h, only animals treated with CPA (67%), midazolam (18%) and scopolamine (27%) exhibited seizure termination. While all treatments except for topiramate reduced neuropathology, CPA, midazolam and scopolamine showed the greatest reduction in pathology. Our results suggest that delayed treatment with CPA, midazolam, or scopolamine is effective at reducing GD-induced seizure activity and neuropathology, with CPA and midazolam capable of facilitating a reduction in GD-induced ACh elevation.
Collapse
Affiliation(s)
- Cindy Acon-Chen
- a Pharmacology Branch, Research Division , US Army Medical Research Institute of Chemical Defense , MD , USA
| | - Jeffrey A Koenig
- a Pharmacology Branch, Research Division , US Army Medical Research Institute of Chemical Defense , MD , USA
| | - Garrett R Smith
- a Pharmacology Branch, Research Division , US Army Medical Research Institute of Chemical Defense , MD , USA
| | - Amber R Truitt
- a Pharmacology Branch, Research Division , US Army Medical Research Institute of Chemical Defense , MD , USA
| | - Thaddeus P Thomas
- a Pharmacology Branch, Research Division , US Army Medical Research Institute of Chemical Defense , MD , USA
| | - Tsung-Ming Shih
- a Pharmacology Branch, Research Division , US Army Medical Research Institute of Chemical Defense , MD , USA
| |
Collapse
|
35
|
McCarren HS, McDonough JH. Anticonvulsant discovery through animal models of status epilepticus induced by organophosphorus nerve agents and pesticides. Ann N Y Acad Sci 2016; 1374:144-50. [DOI: 10.1111/nyas.13092] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hilary S. McCarren
- Pharmacology Department U.S. Army Medical Research Institute of Chemical Defense Aberdeen Proving Ground Maryland
| | - John H. McDonough
- Pharmacology Department U.S. Army Medical Research Institute of Chemical Defense Aberdeen Proving Ground Maryland
| |
Collapse
|
36
|
Piermartiri T, Pan H, Figueiredo TH, Marini AM. α-Linolenic Acid, A Nutraceutical with Pleiotropic Properties That Targets Endogenous Neuroprotective Pathways to Protect against Organophosphate Nerve Agent-Induced Neuropathology. Molecules 2015; 20:20355-80. [PMID: 26569216 PMCID: PMC6332275 DOI: 10.3390/molecules201119698] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 10/29/2015] [Accepted: 11/03/2015] [Indexed: 01/23/2023] Open
Abstract
α-Linolenic acid (ALA) is a nutraceutical found in vegetable products such as flax and walnuts. The pleiotropic properties of ALA target endogenous neuroprotective and neurorestorative pathways in brain and involve the transcription factor nuclear factor kappa B (NF-κB), brain-derived neurotrophic factor (BDNF), a major neuroprotective protein in brain, and downstream signaling pathways likely mediated via activation of TrkB, the cognate receptor of BDNF. In this review, we discuss possible mechanisms of ALA efficacy against the highly toxic OP nerve agent soman. Organophosphate (OP) nerve agents are highly toxic chemical warfare agents and a threat to military and civilian populations. Once considered only for battlefield use, these agents are now used by terrorists to inflict mass casualties. OP nerve agents inhibit the critical enzyme acetylcholinesterase (AChE) that rapidly leads to a cholinergic crisis involving multiple organs. Status epilepticus results from the excessive accumulation of synaptic acetylcholine which in turn leads to the overactivation of muscarinic receptors; prolonged seizures cause the neuropathology and long-term consequences in survivors. Current countermeasures mitigate symptoms and signs as well as reduce brain damage, but must be given within minutes after exposure to OP nerve agents supporting interest in newer and more effective therapies. The pleiotropic properties of ALA result in a coordinated molecular and cellular program to restore neuronal networks and improve cognitive function in soman-exposed animals. Collectively, ALA should be brought to the clinic to treat the long-term consequences of nerve agents in survivors. ALA may be an effective therapy for other acute and chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- Tetsade Piermartiri
- Molecular and Cellular Biology Graduate School Program, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - Hongna Pan
- Department of Neurology and Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - Taiza H Figueiredo
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| | - Ann M Marini
- Department of Neurology and Program in Neuroscience, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| |
Collapse
|
37
|
Reddy SD, Reddy DS. Midazolam as an anticonvulsant antidote for organophosphate intoxication--A pharmacotherapeutic appraisal. Epilepsia 2015; 56:813-21. [PMID: 26032507 DOI: 10.1111/epi.12989] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE This review summarizes the therapeutic potential of midazolam as an anticonvulsant antidote for organophosphate (OP) intoxication. METHODS Benzodiazepines are widely used to treat acute seizures and status epilepticus (SE), a neurologic emergency of persistent seizures that can lead to severe neuronal damage or death. Midazolam is a benzodiazepine hypnotic with a rapid onset and short duration of action. RESULTS Midazolam is considered the new drug of choice for persistent acute seizures and SE, including those caused by neurotoxic OPs and nerve agents. Midazolam is a positive allosteric modulator of synaptic γ-aminobutyric acid (GABA)A receptors in the brain. It potentiates GABAergic inhibition and thereby controls hyperexcitability and seizures. Midazolam is administered intravenously or intramuscularly to control acute seizures and SE. Due to its favorable pharmacokinetic features, midazolam is being considered as a replacement anticonvulsant for diazepam in the antidote kit for nerve agents. Clinical studies such as the recent Rapid Anticonvulsant Medication Prior to Arrival Trial (RAMPART) trial have confirmed the anticonvulsant efficacy of midazolam in SE in prehospital settings. SIGNIFICANCE In experimental models, midazolam is effective when given at the onset of seizures caused by nerve agents. However, benzodiazepines are less effective at terminating seizures when given 30 min or later after OP exposure or seizure onset, likely because of internalization or downregulation of synaptic, but not extrasynaptic, GABAA receptors, which can lead to diminished potency and seizure recurrence.
Collapse
Affiliation(s)
- Sandesh D Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, U.S.A
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, U.S.A
| |
Collapse
|
38
|
Chapman S, Yaakov G, Egoz I, Rabinovitz I, Raveh L, Kadar T, Gilat E, Grauer E. Sarin-induced brain damage in rats is attenuated by delayed administration of midazolam. Neurotoxicology 2015; 49:132-8. [PMID: 25976749 DOI: 10.1016/j.neuro.2015.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/28/2015] [Accepted: 05/02/2015] [Indexed: 01/27/2023]
Abstract
Sarin poisoned rats display a hyper-cholinergic activity including hypersalivation, tremors, seizures and death. Here we studied the time and dose effects of midazolam treatment following nerve agent exposure. Rats were exposed to sarin (1.2 LD50, 108 μg/kg, im), and treated 1 min later with TMB4 and atropine (TA 7.5 and 5 mg/kg, im, respectively). Midazolam was injected either at 1 min (1 mg/kg, im), or 1 h later (1 or 5 mg/kg i.m.). Cortical seizures were monitored by electrocorticogram (ECoG). At 5 weeks, rats were assessed in a water maze task, and then their brains were extracted for biochemical analysis and histological evaluation. Results revealed a time and dose dependent effects of midazolam treatment. Rats treated with TA only displayed acute signs of sarin intoxication, 29% died within 24h and the ECoG showed seizures for several hours. Animals that received midazolam within 1 min survived with only minor clinical signs but with no biochemical, behavioral, or histological sequel. Animals that lived to receive midazolam at 1h (87%) survived and the effects of the delayed administration were dose dependent. Midazolam 5 mg/kg significantly counteracted the acute signs of intoxication and the impaired behavioral performance, attenuated some of the inflammatory response with no effect on morphological damage. Midazolam 1mg/kg showed only a slight tendency to modulate the cognitive function. In addition, the delayed administration of both midazolam doses significantly attenuated ECoG compared to TA treatment only. These results suggest that following prolonged seizure, high dose midazolam is beneficial in counteracting adverse effects of sarin poisoning.
Collapse
Affiliation(s)
- Shira Chapman
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Guy Yaakov
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Inbal Egoz
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ishai Rabinovitz
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Lily Raveh
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Tamar Kadar
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Eran Gilat
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ettie Grauer
- Department of Pharmacology, Israel Institute for Biological Research, Ness Ziona, Israel.
| |
Collapse
|
39
|
Pereira EFR, Aracava Y, DeTolla LJ, Beecham EJ, Basinger GW, Wakayama EJ, Albuquerque EX. Animal models that best reproduce the clinical manifestations of human intoxication with organophosphorus compounds. J Pharmacol Exp Ther 2014; 350:313-21. [PMID: 24907067 DOI: 10.1124/jpet.114.214932] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The translational capacity of data generated in preclinical toxicological studies is contingent upon several factors, including the appropriateness of the animal model. The primary objectives of this article are: 1) to analyze the natural history of acute and delayed signs and symptoms that develop following an acute exposure of humans to organophosphorus (OP) compounds, with an emphasis on nerve agents; 2) to identify animal models of the clinical manifestations of human exposure to OPs; and 3) to review the mechanisms that contribute to the immediate and delayed OP neurotoxicity. As discussed in this study, clinical manifestations of an acute exposure of humans to OP compounds can be faithfully reproduced in rodents and nonhuman primates. These manifestations include an acute cholinergic crisis in addition to signs of neurotoxicity that develop long after the OP exposure, particularly chronic neurologic deficits consisting of anxiety-related behavior and cognitive deficits, structural brain damage, and increased slow electroencephalographic frequencies. Because guinea pigs and nonhuman primates, like humans, have low levels of circulating carboxylesterases-the enzymes that metabolize and inactivate OP compounds-they stand out as appropriate animal models for studies of OP intoxication. These are critical points for the development of safe and effective therapeutic interventions against OP poisoning because approval of such therapies by the Food and Drug Administration is likely to rely on the Animal Efficacy Rule, which allows exclusive use of animal data as evidence of the effectiveness of a drug against pathologic conditions that cannot be ethically or feasibly tested in humans.
Collapse
Affiliation(s)
- Edna F R Pereira
- Division of Translational Toxicology, Department of Epidemiology and Public Health (E.F.R.P., Y.A., E.X.A.), and Program of Comparative Medicine and Departments of Pathology, Medicine, and Epidemiology and Public Health (L.J.D.), University of Maryland School of Medicine, Baltimore, Maryland; Countervail Corporation, Charlotte, North Carolina (E.J.B., G.W.B.); and Biomedical Advanced Research and Development Authority and Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington, DC (E.J.W.)
| | - Yasco Aracava
- Division of Translational Toxicology, Department of Epidemiology and Public Health (E.F.R.P., Y.A., E.X.A.), and Program of Comparative Medicine and Departments of Pathology, Medicine, and Epidemiology and Public Health (L.J.D.), University of Maryland School of Medicine, Baltimore, Maryland; Countervail Corporation, Charlotte, North Carolina (E.J.B., G.W.B.); and Biomedical Advanced Research and Development Authority and Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington, DC (E.J.W.)
| | - Louis J DeTolla
- Division of Translational Toxicology, Department of Epidemiology and Public Health (E.F.R.P., Y.A., E.X.A.), and Program of Comparative Medicine and Departments of Pathology, Medicine, and Epidemiology and Public Health (L.J.D.), University of Maryland School of Medicine, Baltimore, Maryland; Countervail Corporation, Charlotte, North Carolina (E.J.B., G.W.B.); and Biomedical Advanced Research and Development Authority and Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington, DC (E.J.W.)
| | - E Jeffrey Beecham
- Division of Translational Toxicology, Department of Epidemiology and Public Health (E.F.R.P., Y.A., E.X.A.), and Program of Comparative Medicine and Departments of Pathology, Medicine, and Epidemiology and Public Health (L.J.D.), University of Maryland School of Medicine, Baltimore, Maryland; Countervail Corporation, Charlotte, North Carolina (E.J.B., G.W.B.); and Biomedical Advanced Research and Development Authority and Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington, DC (E.J.W.)
| | - G William Basinger
- Division of Translational Toxicology, Department of Epidemiology and Public Health (E.F.R.P., Y.A., E.X.A.), and Program of Comparative Medicine and Departments of Pathology, Medicine, and Epidemiology and Public Health (L.J.D.), University of Maryland School of Medicine, Baltimore, Maryland; Countervail Corporation, Charlotte, North Carolina (E.J.B., G.W.B.); and Biomedical Advanced Research and Development Authority and Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington, DC (E.J.W.)
| | - Edgar J Wakayama
- Division of Translational Toxicology, Department of Epidemiology and Public Health (E.F.R.P., Y.A., E.X.A.), and Program of Comparative Medicine and Departments of Pathology, Medicine, and Epidemiology and Public Health (L.J.D.), University of Maryland School of Medicine, Baltimore, Maryland; Countervail Corporation, Charlotte, North Carolina (E.J.B., G.W.B.); and Biomedical Advanced Research and Development Authority and Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington, DC (E.J.W.)
| | - Edson X Albuquerque
- Division of Translational Toxicology, Department of Epidemiology and Public Health (E.F.R.P., Y.A., E.X.A.), and Program of Comparative Medicine and Departments of Pathology, Medicine, and Epidemiology and Public Health (L.J.D.), University of Maryland School of Medicine, Baltimore, Maryland; Countervail Corporation, Charlotte, North Carolina (E.J.B., G.W.B.); and Biomedical Advanced Research and Development Authority and Office of the Assistant Secretary for Preparedness and Response, Department of Health and Human Services, Washington, DC (E.J.W.)
| |
Collapse
|
40
|
Lehmann C, Wannarka GL. Bioavailability and Dose Proportionality of Intramuscular Diazepam Administered by Autoinjector. J Clin Pharmacol 2013; 48:436-44. [DOI: 10.1177/0091270007311572] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
41
|
Moshiri M, Darchini-Maragheh E, Balali-Mood M. Advances in toxicology and medical treatment of chemical warfare nerve agents. ACTA ACUST UNITED AC 2012; 20:81. [PMID: 23351280 PMCID: PMC3556041 DOI: 10.1186/2008-2231-20-81] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 11/17/2012] [Indexed: 11/10/2022]
Abstract
Organophosphorous (OP) Nerve agents (NAs) are known as the deadliest chemical warfare agents. They are divided into two classes of G and V agents. Most of them are liquid at room temperature. NAs chemical structures and mechanisms of actions are similar to OP pesticides, but their toxicities are higher than these compounds. The main mechanism of action is irreversible inhibition of Acetyl Choline Esterase (AChE) resulting in accumulation of toxic levels of acetylcholine (ACh) at the synaptic junctions and thus induces muscarinic and nicotinic receptors stimulation. However, other mechanisms have recently been described. Central nervous system (CNS) depression particularly on respiratory and vasomotor centers may induce respiratory failure and cardiac arrest. Intermediate syndrome after NAs exposure is less common than OP pesticides poisoning. There are four approaches to detect exposure to NAs in biological samples: (I) AChE activity measurement, (II) Determination of hydrolysis products in plasma and urine, (III) Fluoride reactivation of phosphylated binding sites and (IV) Mass spectrometric determination of cholinesterase adducts. The clinical manifestations are similar to OP pesticides poisoning, but with more severity and fatalities. The management should be started as soon as possible. The victims should immediately be removed from the field and treatment is commenced with auto-injector antidotes (atropine and oximes) such as MARK I kit. A 0.5% hypochlorite solution as well as novel products like M291 Resin kit, G117H and Phosphotriesterase isolated from soil bacterias, are now available for decontamination of NAs. Atropine and oximes are the well known antidotes that should be infused as clinically indicated. However, some new adjuvant and additional treatment such as magnesium sulfate, sodium bicarbonate, gacyclidine, benactyzine, tezampanel, hemoperfusion, antioxidants and bioscavengers have recently been used for OP NAs poisoning.
Collapse
Affiliation(s)
- Mohammd Moshiri
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | |
Collapse
|
42
|
Pouliot W, Bialer M, Hen N, Shekh-Ahmad T, Kaufmann D, Yagen B, Ricks K, Roach B, Nelson C, Dudek FE. A comparative electrographic analysis of the effect of sec-butyl-propylacetamide on pharmacoresistant status epilepticus. Neuroscience 2012; 231:145-56. [PMID: 23159312 DOI: 10.1016/j.neuroscience.2012.11.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 11/03/2012] [Indexed: 01/10/2023]
Abstract
Better treatment of status epilepticus (SE), which typically becomes refractory after about 30 min, will require new pharmacotherapies. The effect of sec-butyl-propylacetamide (SPD), an amide derivative of valproic acid (VPA), on electrographic status epilepticus (ESE) was compared quantitatively to other standard-of-care compounds. Cortical electroencephalograms (EEGs) were recorded from rats during ESE induced with lithium-pilocarpine. Using a previously-published algorithm, the effects of SPD on ESE were compared quantitatively to other relevant compounds. To confirm benzodiazepine resistance, diazepam (DZP) was shown to suppress ESE when administered 15 min after the first motor seizure, but not after 30 min (100mg/kg). VPA (300 mg/kg) also lacked efficacy at 30 min. SPD (130 mg/kg) strongly suppressed ESE at 30 min, less after 45 min, and not at 60 min. At a higher dose (180 mg/kg), SPD profoundly suppressed ESE at 60 min, similar to propofol (100mg/kg) and pentobarbital (30 mg/kg). After 4-6h of SPD-induced suppression, EEG activity often overshot control levels at 7-12h. Valnoctamide (VCD, 180 mg/kg), an SPD homolog, was also efficacious at 30 min. SPD blocks pilocarpine-induced electrographic seizures when administered at 1h after the first motor seizure. SPD has a faster onset and greater efficacy than DZP and VPA, and is similar to propofol and pentobarbital. SPD and structurally similar compounds may be useful for the treatment of refractory ESE. Further development and use of automated analyses of ESE may facilitate drug discovery for refractory SE.
Collapse
Affiliation(s)
- W Pouliot
- Department of Physiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
de Araujo Furtado M, Rossetti F, Chanda S, Yourick D. Exposure to nerve agents: from status epilepticus to neuroinflammation, brain damage, neurogenesis and epilepsy. Neurotoxicology 2012; 33:1476-1490. [PMID: 23000013 DOI: 10.1016/j.neuro.2012.09.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 07/25/2012] [Accepted: 09/03/2012] [Indexed: 12/17/2022]
Abstract
Epilepsy is a common neurological disorder characterized by an initial injury due to stroke, traumatic brain injury, brain infection, or febrile seizures causing status epilepticus (SE). This phenomenon precedes recurrent (secondary) seizures, the latent period (period without seizures) and downstream appearance of spontaneous recurrent seizures (SRS). Epilepsy inducers include the organophosphorous (OP) compounds modified as chemical warfare nerve agents, such as soman. SE induced by soman is a result of cholinergic system hyperactivity caused by the irreversible inhibition of acetylcholinesterase, and the subsequent increase in the amount of the neurotransmitter acetylcholine at central and peripheral sites. SE leads to profound, permanent, complex and widespread brain damage and associated cognitive and behavioral deficits, accompanied by impaired neurogenesis. Several anticonvulsant and neuroprotective strategies have been studied in order to avoid the epileptogenesis which occurs after SE caused by soman exposure. In recent studies, we showed that SRS occur post-soman exposure and neuropathology can be reduced with diazepam (DZP) and valproic acid (VPA) when administered in combination treatment. These effects are accompanied by neurogenesis seen 15 days post-exposure in the hippocampal dentate gyrus (DG). This review discusses several findings about epilepsy induced by soman exposure such as behavioral changes, EEG anomalies, neuropathology, neuroinflammation, neurogenesis, possible circuitry changes and current strategies for treatment. The soman seizure model is an important model of temporal lobe epilepsy (TLE) and comparable in certain respects with well studied models in the literature such as pilocarpine and kainic acid. All these models together allow for a greater understanding of the different mechanisms of seizure induction, propagation and options for treatment. These studies are very necessary for current military and civilian treatment regimens, against OP nerve agent exposure, which fail to prevent SE resulting in severe neuropathology and epilepsy.
Collapse
Affiliation(s)
- Marcio de Araujo Furtado
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Franco Rossetti
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Soma Chanda
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Debra Yourick
- Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States.
| |
Collapse
|
44
|
Fauvelle F, Carpentier P, Dorandeu F, Foquin A, Testylier G. Prediction of Neuroprotective Treatment Efficiency Using a HRMAS NMR-Based Statistical Model of Refractory Status Epilepticus on Mouse: A Metabolomic Approach Supported by Histology. J Proteome Res 2012; 11:3782-95. [DOI: 10.1021/pr300291d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Florence Fauvelle
- Département Effets Biologiques
des Rayonnements, IRBA-CRSSA, La Tronche,
France
| | - Pierre Carpentier
- Département
de Toxicologie
et Risques Chimiques, IRBA-CRSSA, La Tronche,
France
| | - Frederic Dorandeu
- Département
de Toxicologie
et Risques Chimiques, IRBA-CRSSA, La Tronche,
France
- Ecole du Val-de-Grâce, Paris, France
| | - Annie Foquin
- Département
de Toxicologie
et Risques Chimiques, IRBA-CRSSA, La Tronche,
France
| | - Guy Testylier
- Département
de Toxicologie
et Risques Chimiques, IRBA-CRSSA, La Tronche,
France
| |
Collapse
|
45
|
Wang Y, Oguntayo S, Wei Y, Wood E, Brown A, Jensen N, Auta J, Guiodotti A, Doctor BP, Nambiar MP. Neuroprotective effects of imidazenil against chemical warfare nerve agent soman toxicity in guinea pigs. Neurotoxicology 2012; 33:169-77. [DOI: 10.1016/j.neuro.2011.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/11/2011] [Accepted: 12/29/2011] [Indexed: 01/01/2023]
|
46
|
Rossetti F, de Araujo Furtado M, Pak T, Bailey K, Shields M, Chanda S, Addis M, Robertson BD, Moffett M, Lumley LA, Yourick DL. Combined diazepam and HDAC inhibitor treatment protects against seizures and neuronal damage caused by soman exposure. Neurotoxicology 2012; 33:500-11. [PMID: 22387230 DOI: 10.1016/j.neuro.2012.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 01/23/2012] [Accepted: 02/15/2012] [Indexed: 01/31/2023]
Abstract
The occurrence of status epilepticus (SE) is considered the main cause of brain lesions and morphological alterations, such as hippocampal neuron loss, that result in chronic epilepsy. Previous work demonstrated the convulsive and widespread neuropathological effects of soman, an organophosphorus compound that causes SE and severe recurrent seizures as a result of exposure. Seizures begin rapidly after exposure, can continue for hours, and contribute to prolonged physical incapacitation of the victim. This study attempts to identify anticonvulsive and neuroprotective drugs against soman exposure. Male Sprague-Dawley rats were exposed to 1.0 LD(50) soman. EEGraphical and neuropathological (Fluoro-Jade B staining) effects were analyzed at 72 h post-exposure to soman and subsequent treatments with diazepam (DZP) alone or in combination with histone deacetylase inhibitors, suberoylanilide hydroxamic acid (SAHA) or valproic acid (VPA). The extent of brain damage was dependent on the length of SE and not on the number of recurrent seizures. DZP treatment alone decreased SE time and damage in hippocampus, amygdala, thalamus and cortex, but not in piriform nuclei. The combination of DZP and VPA 100 mg/kg showed more anticonvulsive effects, decreased SE time, and afforded more neuroprotection in the hippocampus, mainly the ventral portion. The combination DZP and SAHA 25 mg/kg was more neuroprotective, but not more anticonvulsant than DZP alone. The DZP combination with VPA HDAC inhibitor proved to be a good treatment for SE and neuronal damage caused by soman exposure.
Collapse
Affiliation(s)
- Franco Rossetti
- Walter Reed Army Institute of Research, Silver Spring, MD 20910-7500, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cho Y, Kim WS, Hur GH, Ha YC. Minimum effective drug concentrations of a transdermal patch system containing procyclidine and physostigmine for prophylaxis against soman poisoning in rhesus monkeys. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 33:1-8. [PMID: 22051905 DOI: 10.1016/j.etap.2011.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 08/25/2011] [Accepted: 10/07/2011] [Indexed: 05/31/2023]
Abstract
A transdermal patch system containing procyclidine, an N-methyl-d-aspartate receptor antagonist possessing anticholinergic action, and physostigmine, a reversible cholinesterase inhibitor, was developed, and its prophylactic efficacy against soman intoxication was investigated. Male rhesus monkeys were shaved on the dorsal area, attached with a matrix-type patch with various sizes (2×2 to 7×7 cm) for 24 or 72 h, and challenged with 2×LD₅₀ doses (13μg/kg) of soman. The smallest patch size for the protection against lethality induced by soman intoxication was 3×3cm, resulting in blood procyclidine concentration of 10.8 ng/ml, blood physostigmine concentration of 0.54 ng/ml, which are much lower concentrations than maximum sign-free doses, and blood cholinesterase inhibition of 42%. The drug concentrations and enzyme inhibition rate corresponding to a diverging point of survivability were presumably estimated to be around 7 ng/ml for procyclidine, 0.35 ng/ml for physostigmine, and 37% of enzyme inhibition. Separately, in combination with the patch treatment, the post treatment consisting of atropine (0.5 mg/kg) plus 1-[([4-(aminocarbonyl)pyridinio]methoxy)methyl]-2-[(hydroxyimino)methyl]pyridinium (HI-6, 50 mg/kg) exerted protection against 5×LD₅₀ challenge of soman, which means the posttreatment remarkably augmented the efficacy of the patch. Additionally, it was found that brain injuries induced by soman toxicity were effectively prevented by the patch treatment according to histopathological examinations. These results suggest that the patch system could be an effective alternative for diazepam, an anticonvulsant, and the current pyridostigmine pretreatment, and especially in combination with atropine plus HI-6, could be a choice for quality survival from nerve-agent poisoning.
Collapse
Affiliation(s)
- Young Cho
- Chemical Biological Defense Research Division, 5th R&D Center (5-4), Agency for Defense Development, Yuseong P.O. Box 35, Taejon 305-600, Republic of Korea.
| | | | | | | |
Collapse
|
48
|
White HS, Alex AB, Pollock A, Hen N, Shekh-Ahmad T, Wilcox KS, McDonough JH, Stables JP, Kaufmann D, Yagen B, Bialer M. A new derivative of valproic acid amide possesses a broad-spectrum antiseizure profile and unique activity against status epilepticus and organophosphate neuronal damage. Epilepsia 2011; 53:134-46. [PMID: 22150444 DOI: 10.1111/j.1528-1167.2011.03338.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE sec-Butyl-propylacetamide (SPD) is a one-carbon homolog of valnoctamide (VCD), a central nervous system (CNS)-active amide derivative of valproic acid (VPA) currently in phase II clinical trials. The study reported herein evaluated the anticonvulsant activity of SPD in a battery of rodent seizure and epilepsy models and assessed its efficacy in rat and guinea pig models of status epilepticus (SE) and neuroprotection in an organotypic hippocampal slice model of excitotoxic cell death. METHODS The anticonvulsant activity of SPD was evaluated in several rodent seizure and epilepsy models, including maximal electroshock (MES), 6-Hz psychomotor; subcutaneous (s.c.) metrazol-, s.c. picrotoxin, s.c. bicuculline, and audiogenic, corneal, and hippocampal kindled seizures following intraperitoneal administration. Results obtained with SPD are discussed in relationship to those obtained with VPA and VCD. SPD was also evaluated for its ability to block benzodiazepine-resistant SE induced by pilocarpine (rats) and soman (rats and guinea pigs) following intraperitoneal administration. SPD was tested for its ability to block excitotoxic cell death induced by the glutamate agonists N-methyl-D-aspartate (NMDA) and kainic acid (KA) using organotypic hippocampal slices and SE-induced hippocampal cell death using FluoroJade B staining. The cognitive function of SPD-treated rats that were protected against pilocarpine-induced convulsive SE was examined 10-14 days post-SE using the Morris water maze (MWM). The relationship between the pharmacokinetic profile of SPD and its efficacy against soman-induced SE was evaluated in two parallel studies following SPD (60 mg/kg, i.p.) administration in the soman SE rat model. KEY FINDINGS SPD was highly effective and displayed a wide protective index (PI = median neurotoxic dose/median effective dose [TD(50)/ED(50)]) in the standardized seizure and epilepsy models employed. The wide PI values of SPD demonstrate that it is effective at doses well below those that produce behavioral impairment. Unlike VCD, SPD also displayed anticonvulsant activity in the rat pilocarpine model of SE. Thirty minutes after the induction of SE, the calculated rat ED(50) for SPD against convulsive SE in this model was 84 mg/kg. SPD was not neuroprotective in the organotypic hippocampal slice preparation; however, it did display hippocampal neuroprotection in both SE models and cognitive sparing in the MWM, which was associated with its antiseizure effect against pilocarpine-induced SE. When administered 20 and 40 min after SE onset, SPD (100-174 mg/kg) produced long-lasting efficacy (e.g., 4-8 h) against soman-induced convulsive and electrographic SE in both rats and guinea pigs. SPD ED(50) values in guinea pigs were 67 and 92 mg/kg when administered at SE onset or 40 min after SE onset, respectively. Assuming linear pharmacokinetics (PK), the PK-PD (pharmacodynamic) results (rats) suggests that effective SPD plasma levels ranged between 8 and 40 mg/L (20 min after the onset of soman-induced seizures) and 12-50 mg/L (40 min after the onset of soman-induced seizures). The time to peak (t(max)) pharmacodynamic effect (PD-t(max)) occurred after the PK-t(max), suggesting that SPD undergoes slow distribution to extraplasmatic sites, which is likely responsible for antiseizure activity of SPD. SIGNIFICANCE The results demonstrate that SPD is a broad-spectrum antiseizure compound that blocks SE induced by pilocarpine and soman and affords in vivo neuroprotection that is associated with cognitive sparing. Its activity against SE is superior to that of diazepam in terms of rapid onset, potency, and its effect on animal mortality and functional improvement.
Collapse
Affiliation(s)
- H Steve White
- Anticonvulsant Drug Development Program, Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Che MM, Chanda S, Song J, Doctor BP, Rezk PE, Sabnekar P, Perkins MW, Sciuto AM, Nambiar MP. Aerosolized scopolamine protects against microinstillation inhalation toxicity to sarin in guinea pigs. Toxicol Mech Methods 2011; 21:463-72. [DOI: 10.3109/15376516.2011.562258] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Shih TM, Guarisco JA, Myers TM, Kan RK, McDonough JH. The oxime pro-2-PAM provides minimal protection against the CNS effects of the nerve agents sarin, cyclosarin, and VX in guinea pigs. Toxicol Mech Methods 2010; 21:53-62. [DOI: 10.3109/15376516.2010.529190] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|