1
|
Marrero-Rosado BM, Stone MF, de Araujo Furtado M, Schultz CR, Cadieux CL, Lumley LA. Novel Genetically Modified Mouse Model to Assess Soman-Induced Toxicity and Medical Countermeasure Efficacy: Human Acetylcholinesterase Knock-in Serum Carboxylesterase Knockout Mice. Int J Mol Sci 2021; 22:1893. [PMID: 33672922 PMCID: PMC7918218 DOI: 10.3390/ijms22041893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
The identification of improved medical countermeasures against exposure to chemical warfare nerve agents (CWNAs), a class of organophosphorus compounds, is dependent on the choice of animal model used in preclinical studies. CWNAs bind to acetylcholinesterase and prevent the catalysis of acetylcholine, causing a plethora of peripheral and central physiologic manifestations, including seizure. Rodents are widely used to elucidate the effects of CWNA-induced seizure, albeit with a caveat: they express carboxylesterase activity in plasma. Carboxylesterase, an enzyme involved in the detoxification of some organophosphorus compounds, plays a scavenging role and decreases CWNA availability, thus exerting a protective effect. Furthermore, species-specific amino acid differences in acetylcholinesterase confound studies that use oximes or other compounds to restore its function after inhibition by CWNA. The creation of a human acetylcholinesterase knock-in/serum carboxylesterase knockout (C57BL/6-Ces1ctm1.1LocAChEtm1.1Loc/J; a.k.a KIKO) mouse may facilitate better modeling of CWNA toxicity in a small rodent species. The current studies characterize the effects of exposure to soman, a highly toxic CWNA, and evaluate the efficacy of anti-seizure drugs in this newly developed KIKO mouse model. Data demonstrate that a combination of midazolam and ketamine reduces seizure duration and severity, eliminates the development of spontaneous recurrent seizures, and protects certain brain regions from neuronal damage in a genetically modified model with human relevance to organophosphorus compound toxicity. This new animal model and the results of this study and future studies using it will enhance medical countermeasures development for both defense and homeland security purposes.
Collapse
Affiliation(s)
- Brenda M. Marrero-Rosado
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - Michael F. Stone
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - Marcio de Araujo Furtado
- Anatomy, Physiology and Genetics Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA;
- BioSEaD, LLC, Rockville, MD 20850, USA
| | - Caroline R. Schultz
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - C. Linn Cadieux
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - Lucille A. Lumley
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| |
Collapse
|
2
|
Leeman-Markowski BA, Meador KJ, Moo LR, Cole AJ, Hoch DB, Garcia E, Schachter SC. Does memantine improve memory in subjects with focal-onset epilepsy and memory dysfunction? A randomized, double-blind, placebo-controlled trial. Epilepsy Behav 2018; 88:315-324. [PMID: 30449328 PMCID: PMC7261142 DOI: 10.1016/j.yebeh.2018.06.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 06/02/2018] [Accepted: 06/25/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Excitotoxic injury involving N-methyl-d-aspartate (NMDA) receptor hyperactivity contributes to epilepsy-related memory dysfunction (ERMD). Current treatment strategies for ERMD have limited efficacy and fail to target the underlying pathophysiology. The present pilot study evaluated the efficacy of memantine, an NMDA receptor antagonist, for the treatment of ERMD in adults with focal-onset seizures. METHODS Subjects underwent cognitive testing at baseline, after a 13-week randomized, parallel-group, double-blinded phase (of memantine titrated to 10 mg bid or placebo), and following a 13-week open-label extension phase (of memantine titrated to 10 mg bid). The selective reminding test (SRT) continuous long-term retrieval (CLTR) score and 7/24 Spatial Recall Test learning score served as the primary outcome measures. Secondary measures included tests of attention span, fluency, visual construction, and response inhibition, as well as assessments of quality of life, depression, sleepiness, and side effects. RESULTS Seventeen subjects contributed data to the blinded phase (n = 8 memantine, n = 9 placebo). No significant differences were seen between groups on the primary or secondary outcome measures. Pooled data at the end of the open-label phase from 10 subjects (initially randomized to memantine n = 3 or placebo n = 7) demonstrated statistically significant improvement from baseline in CLTR score, memory-related quality of life, spatial span, and response inhibition. No significant changes were evident in depression, sleepiness, side effects, or seizure frequency throughout the trial. SIGNIFICANCE Results demonstrated no significant effect of memantine on cognition when assessed at the end of the blinded period. Pooled data at the end of the open-label phase showed significant improvement over baseline performance in measures of verbal memory, frontal-executive function, and memory-related quality of life. These improvements, however, may be due to practice effects and should be interpreted cautiously. Findings suggest a favorable safety profile of memantine in the setting of epilepsy.
Collapse
Affiliation(s)
- Beth A. Leeman-Markowski
- Research Service, VA New York Harbor Healthcare System, New York, NY, USA,Department of Neurology, New York University Langone Medical Center, New York, NY, USA,Corresponding author at: 423 E. 23rd St., New York, NY 10010, USA. (B.A. Leeman-Markowski)
| | - Kimford J. Meador
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Lauren R. Moo
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, USA,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Andrew J. Cole
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Daniel B. Hoch
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Eduardo Garcia
- Tufts University School of Medicine, Boston, MA, USA,Newton–Wellesley Neurology Associates, PC, Newton Lower Falls, MA, USA
| | - Steven C. Schachter
- Harvard Medical School, Boston, MA, USA,Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
3
|
Kim RO, Kim BM, Jeong CB, Lee JS, Rhee JS. Effects of chlorpyrifos on life cycle parameters, cytochrome P450S expression, and antioxidant systems in the monogonont rotifer Brachionus koreanus. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2016; 35:1449-1457. [PMID: 26496856 DOI: 10.1002/etc.3288] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/25/2015] [Accepted: 10/21/2015] [Indexed: 06/05/2023]
Abstract
Chlorpyrifos is a widely used organophosphorus insecticide for controlling diverse insect pests of crops. In the monogonont rotifer Brachionus koreanus, population growth retardation with the inhibition of lifespan, fecundity, and individual body size of ovigerous females was shown over 10 d in response to chlorpyrifos exposure. At the molecular and biochemical levels, the rotifer B. koreanus defensome, composed of cytochrome P450 complements, heat shock protein 70, and antioxidant enzymatic systems (i.e., glutathione, glutathione peroxidase, glutathione reductase, and glutathione S-transferase), was significantly induced in response to different concentrations of chlorpyrifos. Thus, chlorpyrifos strongly induced a defensome system to mitigate the deleterious effects of chlorpyrifos at in vivo and in vitro levels as a trade-off in fitness costs. Environ Toxicol Chem 2016;35:1449-1457. © 2015 SETAC.
Collapse
Affiliation(s)
- Ryeo-Ok Kim
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon, South Korea
| | - Bo-Mi Kim
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon, South Korea
| | - Chang-Bum Jeong
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon, South Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, South Korea
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon, South Korea
| | - Jae-Sung Rhee
- Department of Marine Science, College of Natural Sciences, Incheon National University, Incheon, South Korea
| |
Collapse
|
4
|
Greget R, Dadak S, Barbier L, Lauga F, Linossier-Pierre S, Pernot F, Legendre A, Ambert N, Bouteiller JM, Dorandeu F, Bischoff S, Baudry M, Fagni L, Moussaoui S. Modeling and simulation of organophosphate-induced neurotoxicity: Prediction and validation by experimental studies. Neurotoxicology 2016; 54:140-152. [PMID: 27108687 DOI: 10.1016/j.neuro.2016.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/07/2016] [Accepted: 04/19/2016] [Indexed: 02/07/2023]
Abstract
Exposure to organophosphorus (OP) compounds, either pesticides or chemical warfare agents, represents a major health problem. As potent irreversible inhibitors of cholinesterase, OP may induce seizures, as in status epilepticus, and occasionally brain lesions. Although these compounds are extremely toxic agents, the search for novel antidotes remains extremely limited. In silico modeling constitutes a useful tool to identify pharmacological targets and to develop efficient therapeutic strategies. In the present work, we developed a new in silico simulator in order to predict the neurotoxicity of irreversible inhibitors of acetyl- and/or butyrylcholinesterase (ChE) as well as the potential neuroprotection provided by antagonists of cholinergic muscarinic and glutamate N-methyl-d-aspartate (NMDA) receptors. The simulator reproduced firing of CA1 hippocampal neurons triggered by exposure to paraoxon (POX), as found in patch-clamp recordings in in vitro mouse hippocampal slices. In the case of POX intoxication, it predicted a preventing action of the muscarinic receptor antagonist atropine sulfate, as well as a synergistic action with the non-competitive NMDA receptor antagonist memantine. These in silico predictions relative to beneficial effects of atropine sulfate combined with memantine were recapitulated experimentally in an in vivo model of POX in adult male Swiss mice using electroencephalic (EEG) recordings. Thus, our simulator is a new powerful tool to identify protective therapeutic strategies against OP central effects, by screening various combinations of muscarinic and NMDA receptor antagonists.
Collapse
Affiliation(s)
| | - Selma Dadak
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, INSERM, U1191, Université de Montpellier, Montpellier F-34094, France
| | - Laure Barbier
- Institut de Recherche Biomédicale des Armées (IRBA), Département de Toxicologie et Risques Chimiques, Brétigny sur Orge, France
| | - Fabien Lauga
- Institut de Recherche Biomédicale des Armées (IRBA), Département de Toxicologie et Risques Chimiques, Brétigny sur Orge, France
| | - Sandra Linossier-Pierre
- Institut de Recherche Biomédicale des Armées (IRBA), Département de Toxicologie et Risques Chimiques, Brétigny sur Orge, France
| | | | | | | | | | - Frédéric Dorandeu
- Institut de Recherche Biomédicale des Armées (IRBA), Département de Toxicologie et Risques Chimiques, Brétigny sur Orge, France; Ecole du Val-de-Grâce, Paris, France
| | | | | | - Laurent Fagni
- Institut de Génomique Fonctionnelle, CNRS, UMR-5203, INSERM, U1191, Université de Montpellier, Montpellier F-34094, France
| | | |
Collapse
|
5
|
|
6
|
Mullins RJ, Xu S, Pereira EFR, Pescrille JD, Todd SW, Mamczarz J, Albuquerque EX, Gullapalli RP. Prenatal exposure of guinea pigs to the organophosphorus pesticide chlorpyrifos disrupts the structural and functional integrity of the brain. Neurotoxicology 2015; 48:9-20. [PMID: 25704171 DOI: 10.1016/j.neuro.2015.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 02/11/2015] [Accepted: 02/11/2015] [Indexed: 11/25/2022]
Abstract
This study was designed to test the hypothesis that prenatal exposure of guinea pigs to the organophosphorus (OP) pesticide chlorpyrifos (CPF) disrupts the structural and functional integrity of the brain. Pregnant guinea pigs were injected with chlorpyrifos (25 mg/kg, s.c.) or vehicle (peanut oil) once per day for 10 consecutive days, starting approximately on the 50th day of gestation. Cognitive behavior of female offspring was examined starting at 40-45 post-natal days (PND) using the Morris water maze (MWM), and brain structural integrity was analyzed at PND 70 using magnetic resonance imaging (MRI) methods, including T2-weighted anatomical scans and diffusion kurtosis imaging (DKI). The offspring of exposed mothers had significantly decreased body weight and brain volume, particularly in the frontal regions of the brain including the striatum. Furthermore, the offspring demonstrated significant spatial learning deficits in MWM recall compared to the vehicle group. Diffusion measures revealed reduced white matter integrity within the striatum and amygdala that correlated with spatial learning performance. These findings reveal the lasting effect of prenatal exposure to CPF as well as the danger of mother to child transmission of CPF in the environment.
Collapse
Affiliation(s)
- Roger J Mullins
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Su Xu
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Core for Translational Research in Imaging, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Edna F R Pereira
- Division of Translational Toxicology, Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Joseph D Pescrille
- Division of Translational Toxicology, Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Spencer W Todd
- Division of Translational Toxicology, Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Jacek Mamczarz
- Division of Translational Toxicology, Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Edson X Albuquerque
- Division of Translational Toxicology, Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Rao P Gullapalli
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Core for Translational Research in Imaging, University of Maryland School of Medicine, Baltimore, MD 21201, United States.
| |
Collapse
|
7
|
Kaur S, Singh S, Chahal KS, Prakash A. Potential pharmacological strategies for the improved treatment of organophosphate-induced neurotoxicity. Can J Physiol Pharmacol 2014; 92:893-911. [DOI: 10.1139/cjpp-2014-0113] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Organophosphates (OP) are highly toxic compounds that cause cholinergic neuronal excitotoxicity and dysfunction by irreversible inhibition of acetylcholinesterase, resulting in delayed brain damage. This delayed secondary neuronal destruction, which arises primarily in the cholinergic areas of the brain that contain dense accumulations of cholinergic neurons and the majority of cholinergic projection, could be largely responsible for persistent profound neuropsychiatric and neurological impairments such as memory, cognitive, mental, emotional, motor, and sensory deficits in the victims of OP poisoning. The therapeutic strategies for reducing neuronal brain damage must adopt a multifunctional approach to the various steps of brain deterioration: (i) standard treatment with atropine and related anticholinergic compounds; (ii) anti-excitotoxic therapies to prevent cerebral edema, blockage of calcium influx, inhibition of apoptosis, and allow for the control of seizure; (iii) neuroprotection by aid of antioxidants and N-methyl-d-aspartate (NMDA) antagonists (multifunctional drug therapy), to inhibit/limit the secondary neuronal damage; and (iv) therapies targeting chronic neuropsychiatric and neurological symptoms. These neuroprotective strategies may prevent secondary neuronal damage in both early and late stages of OP poisoning, and thus may be a beneficial approach to treating the neuropsychological and neuronal impairments resulting from OP toxicity.
Collapse
Affiliation(s)
- Shamsherjit Kaur
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
- Punjab Technical University, Kapurthala 144601, Punjab, India
| | - Satinderpal Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Karan Singh Chahal
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Atish Prakash
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| |
Collapse
|
8
|
Dorandeu F, Dhote F, Barbier L, Baccus B, Testylier G. Treatment of status epilepticus with ketamine, are we there yet? CNS Neurosci Ther 2013; 19:411-27. [PMID: 23601960 PMCID: PMC6493567 DOI: 10.1111/cns.12096] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 02/23/2013] [Accepted: 02/23/2013] [Indexed: 12/24/2022] Open
Abstract
Status epilepticus (SE), a neurological emergency both in adults and in children, could lead to brain damage and even death if untreated. Generalized convulsive SE (GCSE) is the most common and severe form, an example of which is that induced by organophosphorus nerve agents. First- and second-line pharmacotherapies are relatively consensual, but if seizures are still not controlled, there is currently no definitive data to guide the optimal choice of therapy. The medical community seems largely reluctant to use ketamine, a noncompetitive antagonist of the N-methyl-d-aspartate glutamate receptor. However, a review of the literature clearly shows that ketamine possesses, in preclinical studies, antiepileptic properties and provides neuroprotection. Clinical evidences are scarcer and more difficult to analyze, owing to a use in situations of polytherapy. In absence of existing or planned randomized clinical trials, the medical community should make up its mind from well-conducted preclinical studies performed on appropriate models. Although potentially active, ketamine has no real place for the treatment of isolated seizures, better accepted drugs being used. Its best usage should be during GCSE, but not waiting for SE to become totally refractory. Concerns about possible developmental neurotoxicity might limit its pediatric use for refractory SE.
Collapse
Affiliation(s)
- Frederic Dorandeu
- Département de Toxicologie et risques chimiques, Institut de Recherche Biomédicale des Armées - Centre de Recherches du Service de Santé des Armées (IRBA-CRSSA), La Tronche Cedex, France.
| | | | | | | | | |
Collapse
|
9
|
Dorandeu F, Barbier L, Dhote F, Testylier G, Carpentier P. Ketamine combinations for the field treatment of soman-induced self-sustaining status epilepticus. Review of current data and perspectives. Chem Biol Interact 2013; 203:154-9. [DOI: 10.1016/j.cbi.2012.09.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 09/25/2012] [Accepted: 09/26/2012] [Indexed: 12/21/2022]
|
10
|
Multifunctional drugs as novel antidotes for organophosphates’ poisoning. Toxicology 2011; 290:149-55. [DOI: 10.1016/j.tox.2011.09.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 09/17/2011] [Accepted: 09/19/2011] [Indexed: 11/21/2022]
|
11
|
Mamczarz J, Kulkarni GS, Pereira EFR, Albuquerque EX. Galantamine counteracts development of learning impairment in guinea pigs exposed to the organophosphorus poison soman: clinical significance. Neurotoxicology 2011; 32:785-98. [PMID: 21784098 DOI: 10.1016/j.neuro.2011.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 06/19/2011] [Accepted: 07/05/2011] [Indexed: 11/15/2022]
Abstract
Galantamine, a drug used to treat Alzheimer's disease, protects guinea pigs against the acute toxicity and lethality of organophosphorus (OP) compounds, including soman. Here, we tested the hypothesis that a single exposure of guinea pigs to 1xLD50 soman triggers cognitive impairments that can be counteracted by galantamine. Thus, animals were injected intramuscularly with saline (0.5 ml/kg) or galantamine (8 mg/kg) and 30 min later injected subcutaneously with soman (26.3 μg/kg) or saline. Cognitive performance was analyzed in the Morris water maze (MWM) four days or three months after the soman challenge. Fifty percent of the saline-injected animals that were challenged with soman survived with mild-to-moderate signs of acute toxicity that subsided within a few hours. These animals showed no learning impairment and no memory retention deficit, when training in the MWM started four days post-soman challenge. In contrast, animals presented significant learning impairment when testing started three months post-challenge. Though the magnitude of the impairment correlated with the severity of the acute toxicity, animals that presented no or only mild signs of toxicity were also learning impaired. All guinea pigs that were treated with galantamine survived the soman challenge with no signs of acute toxicity and learned the MWM task as control animals, regardless of when testing began. Galantamine also prevented memory extinction in both saline- and soman-challenged animals. In conclusion, learning impairment develops months after a single exposure to 1xLD50 soman, and galantamine prevents both the acute toxicity and the delayed cognitive deficits triggered by this OP poison.
Collapse
Affiliation(s)
- Jacek Mamczarz
- Division of Translational Toxicology, Department Epidemiology and Public Health, University of Maryland School of Medicine, 10 S. Pine St. Suite 900, Baltimore, MD 21210, USA.
| | | | | | | |
Collapse
|
12
|
Lewejohann L, Pickel T, Sachser N, Kaiser S. Wild genius - domestic fool? Spatial learning abilities of wild and domestic guinea pigs. Front Zool 2010; 7:9. [PMID: 20334697 PMCID: PMC2859863 DOI: 10.1186/1742-9994-7-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 03/25/2010] [Indexed: 11/10/2022] Open
Abstract
Background Domestic animals and their wild relatives differ in a wide variety of aspects. The process of domestication of the domestic guinea pig (Cavia aperea f. porcellus), starting at least 4500 years ago, led to changes in the anatomy, physiology, and behaviour compared with their wild relative, the wild cavy, Cavia aperea. Although domestic guinea pigs are widely used as a laboratory animal, learning and memory capabilities are often disregarded as being very scarce. Even less is known about learning and memory of wild cavies. In this regard, one striking domestic trait is a reduction in relative brain size, which in the domesticated form of the guinea pig amounts to 13%. However, the common belief, that such a reduction of brain size in the course of domestication of different species is accomplished by less learning capabilities is not at all very well established in the literature. Indeed, domestic animals might also even outperform their wild conspecifics taking advantage of their adaptation to a man-made environment. In our study we compared the spatial learning abilities of wild and domestic guinea pigs. We expected that the two forms are different regarding their learning performance possibly related to the process of domestication. Therefore wild cavies as well as domestic guinea pigs of both sexes, aged 35 to 45 days, were tested in the Morris water maze to investigate their ability of spatial learning. Results Both, wild cavies and domestic guinea pigs were able to learn the task, proving the water maze to be a suitable test also for wild cavies. Regarding the speed of learning, male as well as female domestic guinea pigs outperformed their wild conspecifics significantly. Interestingly, only domestic guinea pigs showed a significant spatial association of the platform position, while other effective search strategies were used by wild cavies. Conclusion The results demonstrate that domestic guinea pigs do not at all perform worse than their wild relatives in tests of spatial learning abilities. Yet, the contrary seems to be true. Hence, artificial selection and breeding did not lead to a cognitive decline but rather to an adaptation to man-made environment that allows solving the task more efficiently.
Collapse
Affiliation(s)
- Lars Lewejohann
- Department of Behavioural Biology, University of Münster, 48149 Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Germany
| | - Thorsten Pickel
- Department of Behavioural Biology, University of Münster, 48149 Münster, Germany
| | - Norbert Sachser
- Department of Behavioural Biology, University of Münster, 48149 Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Germany
| | - Sylvia Kaiser
- Department of Behavioural Biology, University of Münster, 48149 Münster, Germany.,Otto Creutzfeldt Center for Cognitive and Behavioral Neuroscience, University of Münster, Germany
| |
Collapse
|
13
|
Agterberg MJH, van den Broek M, Philippens IHCHM. A less stressful animal model: a conditioned avoidance behaviour task for guineapigs. Lab Anim 2010; 44:206-10. [PMID: 20071411 DOI: 10.1258/la.2009.009096] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In the conventional shuttle box, animals are trained to avoid electric foot-shocks. As a consequence of these stress-inducing foot-shocks the animals become anxious and are difficult to train. The aim of the present study was to avoid the stress-inducing foot-shocks and to develop a fast and reliable conditioned avoidance behaviour task for guineapigs. We examined whether narrowband noises at four different sound levels above hearing threshold could be used as conditioned stimulus (CS). The unconditioned stimulus (UCS) was a stream of air, which was used instead of the conventionally used electric foot-shocks. The animals were initially trained with a CS of 78 dB sound pressure level (SPL). In this initial training, guineapigs learned to detect a narrowband noise of 78 dB SPL. Interestingly, during the first additional training session in which three other sound levels were applied, guineapigs did not immediately generalize the learned response at 78 dB SPL to lower sound levels of 58 and 68 dB SPL. However, in this session a noise level of 88 dB SPL led immediately to a high level of responses. The response latency decreased with increasing sound level, from approximately 7 s at 58 dB SPL to approximately 3 s at 88 dB SPL. The escape latency during the UCS was approximately 0.6 s. The present results demonstrate that after reducing the level of stress guineapigs can acquire a response in only a few sessions and furthermore, although the guineapigs were less anxious, training at sound levels of 78 and 88 dB SPL was influenced by an aversive reaction by the guineapig. The results indicate that this aversive reaction of the guineapig is crucial for the training.
Collapse
Affiliation(s)
- Martijn J H Agterberg
- Department of Otorhinolaryngology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | |
Collapse
|
14
|
Zaja-Milatovic S, Gupta RC, Aschner M, Milatovic D. Protection of DFP-induced oxidative damage and neurodegeneration by antioxidants and NMDA receptor antagonist. Toxicol Appl Pharmacol 2009; 240:124-31. [PMID: 19615394 DOI: 10.1016/j.taap.2009.07.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 07/02/2009] [Accepted: 07/06/2009] [Indexed: 12/20/2022]
Abstract
Prophylactic agents acutely administered in response to anticholinesterases intoxication can prevent toxic symptoms, including fasciculations, seizures, convulsions and death. However, anticholinesterases also have long-term unknown pathophysiological effects, making rational prophylaxis/treatment problematic. Increasing evidence suggests that in addition to excessive cholinergic stimulation, organophosphate compounds such as diisopropylphosphorofluoridate (DFP) induce activation of glutamatergic neurons, generation of reactive oxygen (ROS) and nitrogen species (RNS), leading to neurodegeneration. The present study investigated multiple affectors of DFP exposure critical to cerebral oxidative damage and whether antioxidants and NMDA receptor antagonist memantine provide neuroprotection by preventing DFP-induced biochemical and morphometric changes in rat brain. Rats treated acutely with DFP (1.25 mg/kg, s.c.) developed onset of toxicity signs within 7-15 min that progressed to maximal severity of seizures and fasciculations within 60 min. At this time point, DFP caused significant (p<0.01) increases in biomarkers of ROS (F2-isoprostanes, F2-IsoPs; and F4-neuroprostanes, F4-NeuroPs), RNS (citrulline), and declines in high-energy phosphates (HEP) in rat cerebrum. At the same time, quantitative morphometric analysis of pyramidal neurons of the hippocampal CA1 region revealed significant (p<0.01) reductions in dendritic lengths and spine density. When rats were pretreated with the antioxidants N-tert-butyl-alpha-phenylnitrone (PBN, 200 mg/kg, i.p.), or vitamin E (100 mg/kg, i.p./day for 3 days), or memantine (18 mg/kg, i.p.), significant attenuations in DFP-induced increases in F2-IsoPs, F4-NeuroPs, citrulline, and depletion of HEP were noted. Furthermore, attenuation in oxidative damage following antioxidants or memantine pretreatment was accompanied by rescue from dendritic degeneration of pyramidal neurons in the CA1 hippocampal area. These findings closely associated DFP-induced lipid peroxidation with dendritic degeneration of pyramidal neurons in the CA1 hippocampal area and point to possible interventions to limit oxidative injury and dendritic degeneration induced by anticholinesterase neurotoxicity.
Collapse
Affiliation(s)
- Snjezana Zaja-Milatovic
- Vanderbilt University School of Medicine, Department of Pediatrics/Pediatric Toxicology, Nashville, TN 37232-0414, USA
| | | | | | | |
Collapse
|
15
|
Weissman BA, Raveh L. Therapy against organophosphate poisoning: The importance of anticholinergic drugs with antiglutamatergic properties. Toxicol Appl Pharmacol 2008; 232:351-8. [DOI: 10.1016/j.taap.2008.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 06/23/2008] [Accepted: 07/07/2008] [Indexed: 10/21/2022]
|
16
|
Johnson EA, Daugherty KS, Gallagher SJ, Moran AV, DeFord SM. Glutamate receptor pathology is present in the hippocampus following repeated sub-lethal soman exposure in the absence of spatial memory deficits. Neurotoxicology 2007; 29:73-80. [PMID: 17942156 DOI: 10.1016/j.neuro.2007.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 09/05/2007] [Accepted: 09/06/2007] [Indexed: 11/25/2022]
Abstract
Much is still unknown about the long-term effects of repeated, sub-lethal exposure to organophosphorus (OP) nerve agents, such as soman (GD), on learning and memory tasks and related protein expression in the hippocampus. In the present study, guinea pigs were exposed to sub-lethal doses of GD for 10 days and cognitive performance assessed using the Morris water maze (MWM) up to 88 days post-exposure to investigate spatial learning. Additionally, hippocampal lysates were probed for cytoskeletal, synaptic and glutamate receptor proteins using Western blot analyses. No significant difference in MWM performance was observed between repeated sub-lethal GD exposed and saline control groups. However, Western blot analyses revealed significant changes in glutamate receptor protein immunoreactivity for subunits GluR2, NMDAR1, NMDAR2a and NMDAR2b in the hippocampi of GD-exposed guinea pigs. Levels of GluR2, NMDAR2a and NMDAR2b increased by 3 months post-initial exposure and returned to control levels by 6 months while NMDAR1 decreased by 6 months. No significant differences in neurofilament medium (NFM), neurofilament light (NFL) or synaptophysin densitometry were detected and alpha-II-spectrin proteolytic breakdown was also absent. These results reveal that repeated, sub-lethal exposure to GD affects glutamate receptor subunit expression but does not affect cytoskeletal protein immunoreactivity or the proteolytic state in the hippocampus. Though these changes do not affect spatial memory, they may contribute to other cognitive deficits previously observed following sub-lethal OP exposure.
Collapse
Affiliation(s)
- Erik A Johnson
- US Army Medical Research Institute of Chemical Defense, 3100 Ricketts Point Road, Comparative Medicine Division, Comparative Pathology Branch, Aberdeen Proving Ground, MD 21010-5400, USA.
| | | | | | | | | |
Collapse
|
17
|
Hamilton MG, Lundy PM. Medical countermeasures to WMDs: Defence research for civilian and military use. Toxicology 2007; 233:8-12. [PMID: 17029736 DOI: 10.1016/j.tox.2006.08.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 08/16/2006] [Accepted: 08/26/2006] [Indexed: 10/24/2022]
Abstract
This document will address the contributions of defence research to both military and civilian applications. Compared to civilian research capabilities, particularly in terms of personnel, defence research resources are limited. Additionally, many of the research targets are either classified or involve applications that have (or had) limited civilian use. Recently, however, spurred by counter-terrorism preparedness particularly in North America, many previously 'orphaned' research projects have assumed much greater prominence. This has occurred in all areas of militarily significant research, but this paper will focus on medical countermeasures to weapons of mass destruction and to a lesser extent on detection/identification issues. In the area of countermeasures to chemical weapons, most of the defence research has been devoted to "classical" CWAs such as nerve agents, vesicants, choking and blood agents, with considerable success in some applications. Similarly vaccination programs for the biological weapons have also been quite successful. And recent attention has been directed toward the "emerging" threats such as some of the exquisitely lethal toxins. The difference now is that all of these research programs have the objective of moving from research to development of not only military but also civilian use very much sooner than later.
Collapse
Affiliation(s)
- Murray G Hamilton
- Rocky Mountain Center for Homeland Defense, University of Denver, Denver, CO, USA
| | | |
Collapse
|
18
|
Tetz LM, Rezk PE, Ratcliffe RH, Gordon RK, Steele KE, Nambiar MP. Development of a rat pilocarpine model of seizure/status epilepticus that mimics chemical warfare nerve agent exposure. Toxicol Ind Health 2006; 22:255-66. [PMID: 16924957 DOI: 10.1191/0748233706th268oa] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We developed a rat pilocarpine seizure/status epilepticus (SE) model, which closely resembles 1.6-2.0 x LD50 soman exposure, to analyse the molecular mechanism of neuronal damage and to screen effective neuroprotectants against cholinergic agonist and chemical warfare nerve agent (CWNA) exposure. Rats implanted with radiotelemetry probes capable of recording electroencephalogram (EEG), electrocardiogram (ECG), temperature, and physical activity were treated with lithium chloride (5 mEq/kg, im), followed 24 h later by (ip) doses of pilocarpine hydrochloride. Based on radiotelemetry analysis, a dose of 240 mg/kg (ip) pilocarpine generated seizure/SE analogous to 1.6-2.0 x LD50 of soman. The model was refined by reducing the peripheral convulsions without affecting the central nervous system (CNS) by administering methylscopolamine bromide (1 mg/kg, ip), an anti-cholinergic that does not cross the blood-brain barrier. However, when methylscopolamine bromide was administered, a higher dose of pilocarpine (320 mg/kg, ip) was required to generate the equivalent seizure/SE. Histopathology data indicated that pilocarpine induces significant damage to the hippocampal region of the brain, with similar neuropathology to that of 1.6-2.0 x LD50 soman exposure. There was a reduction in body temperature after the administration of pilocarpine, as observed in organophosphate (OP) nerve agents exposure. The heart-rate of pilocarpine-treated animals increased compared to the normal range. The pilocarpine seizure/SE model was also reproducible in the absence of lithium chloride. These results support that pilocarpine seizure/SE model is useful in studying the molecular mechanisms of neuropathology and screening neuroprotectants following cholinergic agonist and CWNA exposure.
Collapse
Affiliation(s)
- Lauren M Tetz
- Department of Biochemical Pharmacology/Division of Biochemistry, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910-7500, USA
| | | | | | | | | | | |
Collapse
|
19
|
Damodaran TV, Greenfield ST, Patel AG, Dressman HK, Lin SK, Abou-Donia MB. Toxicogenomic studies of the rat brain at an early time point following acute sarin exposure. Neurochem Res 2006; 31:367-81. [PMID: 16733813 DOI: 10.1007/s11064-005-9023-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2005] [Indexed: 10/24/2022]
Abstract
We have studied sarin-induced global gene expression patterns at an early time point (2 h: 0.5 x LD50) using Affymetrix Rat Neurobiology U34 chips and male Sprague-Dawley rats. A total of 46 genes showed statistically significant alterations from control levels. Three gene categories contained more of the altered genes than any other groups: ion channel (8 genes) and calcium channel and binding proteins (6 genes). Alterations were also found in the following gene groups: ATPases and ATP-based transporters (4), growth factors (4), G-protein-coupled receptor pathway-related molecules (3), neurotransmission and neurotransmitter transporters (3), cytoskeletal and cell adhesion molecules (2), hormones (2), mitochondria-associated proteins (2), myelin proteins (2), stress-activated molecules (2), cytokine (1), caspase (1), GABAnergic (1), glutamergic (1), immediate early gene (1), prostaglandin (1), transcription factor (1), and tyrosine phosphorylation molecule (1). Persistent alteration of the following genes also were noted: Arrb1, CaMKIIa, CaMKIId, Clcn5, IL-10, c-Kit, and Plp1, suggesting altered GPCR, kinase, channel, and cytokine pathways. Selected genes from the microarray data were further validated using relative RT-PCR. Some of those genes (GFAP, NF-H, CaMKIIa, Calm, and MBP) have been shown by other laboratories and ours, to be involved in the pathogenesis of sarin-induced pathology and organophosphate-induced delayed neurotoxicity (OPIDN). Induction of both proapoptotic (Bcl2l11, Casp6) and antiapoptotic (Bcl-X) genes, besides suppression of p21, suggest complex cell death/protection-related mechanisms operating early on. Principal component analysis (PCA) of the expression data confirmed that the changes in gene expression are a function of sarin exposure, since the control and treatment groups separated clearly. Our model (based on current and previous studies) indicates that both degenerative and regenerative pathways are activated early and contribute to the level of neurodegeneration at a later time, leading to neuro-pathological alterations.
Collapse
Affiliation(s)
- Tirupapuliyur V Damodaran
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, P.O. Box 3813, Durham, NC, 27710, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Langston JL, Adkins AL, Moran AV, Rockwood GA, Deford MS. Effects of sarin on the operant behavior of guinea pigs. Neurotoxicol Teratol 2005; 27:841-53. [PMID: 16046097 DOI: 10.1016/j.ntt.2005.06.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Revised: 06/20/2005] [Accepted: 06/21/2005] [Indexed: 11/22/2022]
Abstract
The present study evaluated the dose-response effects of subacute exposure to sublethal doses of the organophosphorus (OP) chemical warfare nerve agent (CWNA) sarin (GB) on the operant behavior of guinea pigs. Dietary restricted guinea pigs, trained to respond for food under a progressive ratio (PR) schedule of reinforcement, were injected five times per week (Monday-Friday) for 2 weeks with fractions (0.1, 0.2, and 0.4) of the established LD(50) of GB (42 microg/kg). Changes in body weight, whole blood (WB) acetylcholinesterase (AChE) levels, and operant performances were monitored over the 2 weeks of GB exposure and for an additional 2 weeks following the termination of exposures. There were dose-related changes in body weight and WB AChE levels throughout the exposure and post-exposure periods. Several parameters of PR performance were disrupted during exposure to 0.4 LD50 GB, however, concurrent weight loss indicated the presence of overt toxicity. PR performance recovered following the termination of exposures. Lower doses (0.1 and 0.2 LD50) of GB failed to produce reliable effects on operant performance during the exposure period. Overall responding decreased during exposure to 0.4 LD50 GB, resulting in reduced response rates and break points. The decrease in overall response rates was attributed to an increase in pausing since there was no decrease in running rate. Motor effects of 0.4 LD50 GB were evident as an increase in the proportion of lever press durations > or = 1.0 s. In the present study, doses of GB lower than 0.4 LD50 produced no marked alteration of operant performance in guinea pigs, although WB AChE levels were maximally inhibited to 20% of control.
Collapse
Affiliation(s)
- Jeffrey L Langston
- Drug Assessment Division, US Army Medical Research Institute of Chemical Defense, MCMR-UV-DA, Aberdeen Proving Ground, MD 21010-5400, USA.
| | | | | | | | | |
Collapse
|
21
|
Wang YA, Zhou WX, Li JX, Liu YQ, Yue YJ, Zheng JQ, Liu KL, Ruan JX. Anticonvulsant effects of phencynonate hydrochloride and other anticholinergic drugs in soman poisoning: neurochemical mechanisms. Life Sci 2005; 78:210-23. [PMID: 16154160 DOI: 10.1016/j.lfs.2005.04.071] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 04/19/2005] [Indexed: 11/19/2022]
Abstract
Previous studies have paid little attention to the anticonvulsant effect of anticholinergic drugs that act on both muscarinic (M) and nicotinic (N) receptors during soman-induced seizures. Therefore, with the establishment of a soman-induced seizures model in rats, this study evaluated the efficacy in preventing soman-induced convulsions of two antagonists of both the M and N receptors, phencynonate hydrochloride (PCH) and penehyclidine hydrochloride (8018), which were synthesized by our institute, and of other anticholinergic drugs, and investigated the mechanisms of their antiseizures responses. Male rats, previously prepared with electrodes to record electroencephalographic (EEG) activity, were pretreated with the oxime HI-6 (125 mg kg-1, i.p.) 30 min before they were administered soman (180 microg kg-1, s.c.). All animals developed seizures subsequent to this treatment. Different drugs were given at different times (5, 20 and 40 min after seizures onset) and their anticonvulsant effects were monitored and compared using the two variables, i.e. the dose that could totally control the ongoing seizures, as well as the speed of seizures control. The anticonvulsant effects of atropine, scopolamine and 8018 decreased with the progression of the seizures, and they eventually lost their anticonvulsant activity when the seizures had progressed for 40 min. In contrast, PCH showed good anticonvulsant effectiveness at 5 and 20 min, and especially at 40 min after seizures onset. Of the anticholinergic drugs tested, atropine, scopolamine, and 8018 showed no obvious protection against pentylenetetrazol (PTZ)-induced convulsions or N-methyl-D-aspartate (NMDA)-induced lethality in mice. However, PCH antagonized the PTZ-induced convulsions in a dose-dependant manner with an ED50 of 10.8 mg kg-1, i.p. (range of 7.1-15.2 mg kg-1) and partly blocked the lethal effects of NMDA in mice. PCH also dose-dependently inhibited NMDA-induced injury in rat primary hippocampal neuronal cultures, suggesting a possible neuroprotective action in vivo. In conclusion, our study suggests that the mechanisms of PCH action against soman-induced seizures might differ from those of the M receptor antagonists atropine and scopolamine, and that of the antagonist of both the M and N receptors, 8018. The pharmacological profile of PCH might include anticholinergic and anti-NMDA properties. Compared with the currently recommended anticonvulsant drug diazepam, with known NMDA receptor antagonists such as MK-801 and with conventional anticholinergics such as scopolamine and atropine, the potent anticonvulsant effects of PCH during the entire initial 40 min period of soman poisoning, and its fewer adverse effects, all suggest that PCH might serve as a new type of anticonvulsant for the treatment of seizures induced by soman.
Collapse
Affiliation(s)
- Yong-An Wang
- Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Abstract
Organophosphorus compounds are potent neurotoxic chemicals that are widely used in medicine, industry, and agriculture. The neurotoxicity of these chemicals has been documented in accidental human poisoning, epidemiological studies, and animal models. Organophosphorus compounds have 3 distinct neurotoxic actions. The primary action is the irreversible inhibition of acetylcholinesterase, resulting in the accumulation of acetylcholine and subsequent overstimulation of the nicotinic and muscarinic acetylcholine receptors, resulting in cholinergic effects. Another action of some of these compounds, arising from single or repeated exposure, is a delayed onset of ataxia, accompanied by a Wallerian-type degeneration of the axon and myelin in the most distal portion of the longest tracts in both the central and peripheral nervous systems, and is known as organophosphorus ester-induced delayed neurotoxicity (OPIDN). In addition, since the introduction and extensive use of synthetic organophosphorus compounds in agriculture and industry half a century ago, many studies have reported long-term, persistent, chronic neurotoxicity symptoms in individuals as a result of acute exposure to high doses that cause acute cholinergic toxicity, or from long-term, low-level, subclinical doses of these chemicals. The author attempts to define the neuronal disorder that results from organophosphorus ester-induced chronic neurotoxicity (OPICN), which leads to long-term neurological and neurobehavioral deficits. Although the mechanisms of this neurodegenerative disorder have yet to be established, the sparse available data suggest that large toxic doses of organophosphorus compounds cause acute necrotic neuronal cell death in the brain, whereas sublethal or subclinical doses produce apoptotic neuronal cell death and involve oxidative stress.
Collapse
Affiliation(s)
- Mohamed B Abou-Donia
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
24
|
Munirathinam S, Bahr BA. Repeated contact with subtoxic soman leads to synaptic vulnerability in hippocampus. J Neurosci Res 2004; 77:739-46. [PMID: 15352221 DOI: 10.1002/jnr.20209] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Soman, an anticholinesterase and dangerous nerve agent, produces convulsions, memory impairment, and cell loss in the brain, especially in the hippocampus. Soman-induced accumulation of acetylcholine initiates mechanisms responsible for the development of incapacitating seizures. The prolonged epileptiform nature of these seizures causes the release of another excitatory neurotransmitter, glutamate, which has been linked to the toxic action of the nerve agent. Here, we tested whether subtoxic soman exposures influence the brain's sensitivity to glutamate-based excitotoxicity. Over a 1-week period, hippocampal slice cultures were exposed daily to a transient level of soman that produced no evidence of synaptic deterioration. After the subtoxic soman treatments, however, the tissue became vulnerable to a brief episode of glutamate receptor overstimulation that normally resulted in little or no excitotoxic damage. In those slice cultures treated with subtoxic soman, a decline in synaptic markers as well as an increase in spectrin breakdown occurred 24 hr after the mild excitotoxic event. Exposure to high soman concentrations alone produced similar synaptic degeneration, but without evident cell death, suggesting that synaptic decline is an early neurotoxicological response to the nerve agent. Interestingly, enhanced excitotoxic sensitivity caused the brain tissue to become susceptible to disparate insults initiated before or after the soman contact. These findings indicate that seemingly innocuous soman exposures leave the hippocampus sensitive to the types of insults implicated in traumatic brain injury and stroke. They also warn that asymptomatic contact with soman may lead to progressive synaptopathogenesis and that early indicators of soman exposure are critical to prevent potential brain injury.
Collapse
Affiliation(s)
- Subramani Munirathinam
- Department of Pharmaceutical Sciences, Center for Drug Discovery, University of Connecticut, Storrs, Connecticut 06269, USA
| | | |
Collapse
|