Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2023

Open Access 01-12-2023 | Status Epilepticus | Research

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

Authors: Suraj S. Vasanthi, Nikhil S. Rao, Manikandan Samidurai, Nyzil Massey, Christina Meyer, Meghan Gage, Mihir Kharate, Aida Almanza, Logan Wachter, Candide Mafuta, Lily Trevino, Adriana M. Carlo, Elijah Bryant, Brooke E. Corson, Morgan Wohlgemuth, Morgan Ostrander, Lucas Showman, Chong Wang, Thimmasettappa Thippeswamy

Published in: Journal of Neuroinflammation | Issue 1/2023

Login to get access

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.
Appendix
Available only for authorised users
Literature
1.
go back to reference Gage M, Rao NS, Samidurai M, Putra M, Vasanthi SS, Meyer C, Wang C, Thippeswamy T. Soman (GD) rat model to mimic civilian exposure to nerve agent: mortality, video-EEG based status epilepticus severity, sex differences, spontaneously recurring seizures, and brain pathology. Front Cell Neurosci. 2021;15: 798247.PubMedCrossRef Gage M, Rao NS, Samidurai M, Putra M, Vasanthi SS, Meyer C, Wang C, Thippeswamy T. Soman (GD) rat model to mimic civilian exposure to nerve agent: mortality, video-EEG based status epilepticus severity, sex differences, spontaneously recurring seizures, and brain pathology. Front Cell Neurosci. 2021;15: 798247.PubMedCrossRef
2.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
3.
go back to reference Hulse EJ, Haslam JD, Emmett SR, Woolley T. Organophosphorus nerve agent poisoning: managing the poisoned patient. Br J Anaesth. 2019;123:457–63.PubMedCrossRef Hulse EJ, Haslam JD, Emmett SR, Woolley T. Organophosphorus nerve agent poisoning: managing the poisoned patient. Br J Anaesth. 2019;123:457–63.PubMedCrossRef
4.
go back to reference Nallapaneni A, Liu J, Karanth S, Pope C. Modulation of paraoxon toxicity by the cannabinoid receptor agonist WIN 55,212–2. Toxicology. 2006;227:173–83.PubMedCrossRef Nallapaneni A, Liu J, Karanth S, Pope C. Modulation of paraoxon toxicity by the cannabinoid receptor agonist WIN 55,212–2. Toxicology. 2006;227:173–83.PubMedCrossRef
5.
go back to reference Jackson C, Ardinger C, Winter KM, McDonough JH, McCarren HS. Validating a model of benzodiazepine refractory nerve agent-induced status epilepticus by evaluating the anticonvulsant and neuroprotective effects of scopolamine, memantine, and phenobarbital. J Pharmacol Toxicol Methods. 2019;97:1–12.PubMedPubMedCentralCrossRef Jackson C, Ardinger C, Winter KM, McDonough JH, McCarren HS. Validating a model of benzodiazepine refractory nerve agent-induced status epilepticus by evaluating the anticonvulsant and neuroprotective effects of scopolamine, memantine, and phenobarbital. J Pharmacol Toxicol Methods. 2019;97:1–12.PubMedPubMedCentralCrossRef
6.
go back to reference McDonough JH, McMonagle J, Copeland T, Zoeffel D, Shih TM. Comparative evaluation of benzodiazepines for control of soman-induced seizures. Arch Toxicol. 1999;73:473–8.PubMedCrossRef McDonough JH, McMonagle J, Copeland T, Zoeffel D, Shih TM. Comparative evaluation of benzodiazepines for control of soman-induced seizures. Arch Toxicol. 1999;73:473–8.PubMedCrossRef
7.
go back to reference Reddy SD, Reddy DS. Midazolam as an anticonvulsant antidote for organophosphate intoxication–a pharmacotherapeutic appraisal. Epilepsia. 2015;56:813–21.PubMedPubMedCentralCrossRef Reddy SD, Reddy DS. Midazolam as an anticonvulsant antidote for organophosphate intoxication–a pharmacotherapeutic appraisal. Epilepsia. 2015;56:813–21.PubMedPubMedCentralCrossRef
8.
go back to reference Shih T-M, McDonough JH, Koplovitz I. Anticonvulsants for soman-induced seizure activity. J Biomed Sci. 1999;6:86–96.PubMed Shih T-M, McDonough JH, Koplovitz I. Anticonvulsants for soman-induced seizure activity. J Biomed Sci. 1999;6:86–96.PubMed
9.
go back to reference Shih TM, McDonough JH. Organophosphorus nerve agents-induced seizures and efficacy of atropine sulfate as anticonvulsant treatment. Pharmacol Biochem Behav. 1999;64:147–53.PubMedCrossRef Shih TM, McDonough JH. Organophosphorus nerve agents-induced seizures and efficacy of atropine sulfate as anticonvulsant treatment. Pharmacol Biochem Behav. 1999;64:147–53.PubMedCrossRef
10.
go back to reference Apland JP, Aroniadou-Anderjaska V, Figueiredo TH, Rossetti F, Miller SL, Braga MFM. The limitations of diazepam as a treatment for nerve agent-induced seizures and neuropathology in rats: comparison with UBP302. J Pharmacol Exp Ther. 2014;351:359–72.PubMedPubMedCentralCrossRef Apland JP, Aroniadou-Anderjaska V, Figueiredo TH, Rossetti F, Miller SL, Braga MFM. The limitations of diazepam as a treatment for nerve agent-induced seizures and neuropathology in rats: comparison with UBP302. J Pharmacol Exp Ther. 2014;351:359–72.PubMedPubMedCentralCrossRef
12.
go back to reference Figueiredo TH, Apland JP, Braga MFM, Marini AM. Acute and long-term consequences of exposure to organophosphate nerve agents in humans. Epilepsia. 2018;59:92–9.PubMedPubMedCentralCrossRef Figueiredo TH, Apland JP, Braga MFM, Marini AM. Acute and long-term consequences of exposure to organophosphate nerve agents in humans. Epilepsia. 2018;59:92–9.PubMedPubMedCentralCrossRef
13.
go back to reference 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;367:302–21.PubMedPubMedCentralCrossRef 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;367:302–21.PubMedPubMedCentralCrossRef
14.
go back to reference Putra M, Sharma S, Gage M, et al. Inducible nitric oxide synthase inhibitor, 1400W, mitigates DFP-induced long-term neurotoxicity in the rat model. Neurobiol Dis. 2020;133: 104443.PubMedCrossRef Putra M, Sharma S, Gage M, et al. Inducible nitric oxide synthase inhibitor, 1400W, mitigates DFP-induced long-term neurotoxicity in the rat model. Neurobiol Dis. 2020;133: 104443.PubMedCrossRef
15.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
16.
go back to reference Beamer E, Otahal J, Sills GJ, Thippeswamy T. N w -Propyl- l -arginine (L-NPA) reduces status epilepticus and early epileptogenic events in a mouse model of epilepsy: behavioural, EEG and immunohistochemical analyses: L-NPA modulates SE and epileptogenesis. Eur J Neurosci. 2012;36:3194–203.PubMedCrossRef Beamer E, Otahal J, Sills GJ, Thippeswamy T. N w -Propyl- l -arginine (L-NPA) reduces status epilepticus and early epileptogenic events in a mouse model of epilepsy: behavioural, EEG and immunohistochemical analyses: L-NPA modulates SE and epileptogenesis. Eur J Neurosci. 2012;36:3194–203.PubMedCrossRef
17.
go back to reference Cosgrave AS, McKay JS, Bubb V, Morris R, Quinn JP, Thippeswamy T. Regulation of activity-dependent neuroprotective protein (ADNP) by the NO-cGMP pathway in the hippocampus during kainic acid-induced seizure. Neurobiol Dis. 2008;30:281–92.PubMedCrossRef Cosgrave AS, McKay JS, Bubb V, Morris R, Quinn JP, Thippeswamy T. Regulation of activity-dependent neuroprotective protein (ADNP) by the NO-cGMP pathway in the hippocampus during kainic acid-induced seizure. Neurobiol Dis. 2008;30:281–92.PubMedCrossRef
18.
go back to reference Guzik TJ, Korbut R, Adamek-Guzik T. Nitric oxide and superoxide in inflammation and immune regulation. J Physiol Pharmacol Off J Pol Physiol Soc. 2003;54:469–87. Guzik TJ, Korbut R, Adamek-Guzik T. Nitric oxide and superoxide in inflammation and immune regulation. J Physiol Pharmacol Off J Pol Physiol Soc. 2003;54:469–87.
19.
go back to reference Liy PM, Puzi NNA, Jose S, Vidyadaran S. Nitric oxide modulation in neuroinflammation and the role of mesenchymal stem cells. Exp Biol Med Maywood NJ. 2021;246:2399–406.CrossRef Liy PM, Puzi NNA, Jose S, Vidyadaran S. Nitric oxide modulation in neuroinflammation and the role of mesenchymal stem cells. Exp Biol Med Maywood NJ. 2021;246:2399–406.CrossRef
20.
go back to reference Puttachary S, Sharma S, Verma S, Yang Y, Putra M, Thippeswamy A, Luo D, Thippeswamy T. 1400W, a highly selective inducible nitric oxide synthase inhibitor is a potential disease modifier in the rat kainate model of temporal lobe epilepsy. Neurobiol Dis. 2016;93:184–200.PubMedCrossRef Puttachary S, Sharma S, Verma S, Yang Y, Putra M, Thippeswamy A, Luo D, Thippeswamy T. 1400W, a highly selective inducible nitric oxide synthase inhibitor is a potential disease modifier in the rat kainate model of temporal lobe epilepsy. Neurobiol Dis. 2016;93:184–200.PubMedCrossRef
21.
go back to reference Gage M, Putra M, Estrada CG, Golden M, Wachter L, Gard M, Thippeswamy T. Differential impact of severity and duration of status epilepticus, medical countermeasures, and a disease-modifier, saracatinib (AZD0530), on brain regions in the rat diisopropylfluorophosphate (DFP) model. Front Cell Neurosci. 2021;0:426. Gage M, Putra M, Estrada CG, Golden M, Wachter L, Gard M, Thippeswamy T. Differential impact of severity and duration of status epilepticus, medical countermeasures, and a disease-modifier, saracatinib (AZD0530), on brain regions in the rat diisopropylfluorophosphate (DFP) model. Front Cell Neurosci. 2021;0:426.
22.
go back to reference Tse K, Puttachary S, Beamer E, Sills GJ, Thippeswamy T. Advantages of repeated low dose against single high dose of kainate in C57BL/6J mouse model of status epilepticus: behavioral and electroencephalographic studies. PLoS ONE. 2014;9: e96622.PubMedPubMedCentralCrossRef Tse K, Puttachary S, Beamer E, Sills GJ, Thippeswamy T. Advantages of repeated low dose against single high dose of kainate in C57BL/6J mouse model of status epilepticus: behavioral and electroencephalographic studies. PLoS ONE. 2014;9: e96622.PubMedPubMedCentralCrossRef
23.
go back to reference Wyatt-Johnson SK, Sommer AL, Shim KY, Brewster AL. Suppression of microgliosis with the colony-stimulating factor 1 receptor inhibitor PLX3397 does not attenuate memory defects during epileptogenesis in the rat. Front Neurol. 2021;12: Wyatt-Johnson SK, Sommer AL, Shim KY, Brewster AL. Suppression of microgliosis with the colony-stimulating factor 1 receptor inhibitor PLX3397 does not attenuate memory defects during epileptogenesis in the rat. Front Neurol. 2021;12:
24.
go back to reference Leger M, Quiedeville A, Bouet V, Haelewyn B, Boulouard M, Schumann-Bard P, Freret T. Object recognition test in mice. Nat Protoc. 2013;8:2531–7.PubMedCrossRef Leger M, Quiedeville A, Bouet V, Haelewyn B, Boulouard M, Schumann-Bard P, Freret T. Object recognition test in mice. Nat Protoc. 2013;8:2531–7.PubMedCrossRef
25.
go back to reference Gage M, Golden M, Putra M, Sharma S, Thippeswamy T. Sex as a biological variable in the rat model of diisopropylfluorophosphate-induced long-term neurotoxicity. Ann N Y Acad Sci. 2020;1479:44–64.PubMedCrossRef Gage M, Golden M, Putra M, Sharma S, Thippeswamy T. Sex as a biological variable in the rat model of diisopropylfluorophosphate-induced long-term neurotoxicity. Ann N Y Acad Sci. 2020;1479:44–64.PubMedCrossRef
26.
go back to reference Garza-Lombó C, Thang M, Greve HJ, Mumaw CL, Messenger EJ, Ahmed C, Quinn E, Sullivan K, Block ML. Circulating HMGB1 is elevated in veterans with Gulf War Illness and triggers the persistent pro-inflammatory microglia phenotype in male C57Bl/6J mice. Transl Psychiatry. 2021;11:390.PubMedPubMedCentralCrossRef Garza-Lombó C, Thang M, Greve HJ, Mumaw CL, Messenger EJ, Ahmed C, Quinn E, Sullivan K, Block ML. Circulating HMGB1 is elevated in veterans with Gulf War Illness and triggers the persistent pro-inflammatory microglia phenotype in male C57Bl/6J mice. Transl Psychiatry. 2021;11:390.PubMedPubMedCentralCrossRef
27.
28.
go back to reference Wyatt-Johnson SK, Herr SA, Brewster AL. Status epilepticus triggers time-dependent alterations in microglia abundance and morphological phenotypes in the hippocampus. Front Neurol. 2017;8:700.PubMedPubMedCentralCrossRef Wyatt-Johnson SK, Herr SA, Brewster AL. Status epilepticus triggers time-dependent alterations in microglia abundance and morphological phenotypes in the hippocampus. Front Neurol. 2017;8:700.PubMedPubMedCentralCrossRef
29.
go back to reference Zhang Y, Huo M, Zhou J, Xie S. PKSolver: an add-in program for pharmacokinetic and pharmacodynamic data analysis in Microsoft Excel. Comput Methods Programs Biomed. 2010;99:306–14.PubMedCrossRef Zhang Y, Huo M, Zhou J, Xie S. PKSolver: an add-in program for pharmacokinetic and pharmacodynamic data analysis in Microsoft Excel. Comput Methods Programs Biomed. 2010;99:306–14.PubMedCrossRef
31.
go back to reference Fracassi A, Marcatti M, Tumurbaatar B, Woltjer R, Moreno S, Taglialatela G. TREM2-induced activation of microglia contributes to synaptic integrity in cognitively intact aged individuals with Alzheimer’s neuropathology. Brain Pathol Zurich Switz. 2023;33: e13108. Fracassi A, Marcatti M, Tumurbaatar B, Woltjer R, Moreno S, Taglialatela G. TREM2-induced activation of microglia contributes to synaptic integrity in cognitively intact aged individuals with Alzheimer’s neuropathology. Brain Pathol Zurich Switz. 2023;33: e13108.
33.
go back to reference Gharagozloo M, Smith MD, Jin J, et al. Complement component 3 from astrocytes mediates retinal ganglion cell loss during neuroinflammation. Acta Neuropathol (Berl). 2021;142:899–915.PubMedCrossRef Gharagozloo M, Smith MD, Jin J, et al. Complement component 3 from astrocytes mediates retinal ganglion cell loss during neuroinflammation. Acta Neuropathol (Berl). 2021;142:899–915.PubMedCrossRef
34.
go back to reference Wei Y, Chen T, Bosco DB, Xie M, Zheng J, Dheer A, Ying Y, Wu Q, Lennon VA, Wu L-J. The complement C3–C3aR pathway mediates microglia-astrocyte interaction following status epilepticus. Glia. 2021;69:1155–69.PubMedCrossRef Wei Y, Chen T, Bosco DB, Xie M, Zheng J, Dheer A, Ying Y, Wu Q, Lennon VA, Wu L-J. The complement C3–C3aR pathway mediates microglia-astrocyte interaction following status epilepticus. Glia. 2021;69:1155–69.PubMedCrossRef
35.
go back to reference Coubard S, Béracochéa D, Collombet J-M, Philippin J-N, Krazem A, Liscia P, Lallement G, Piérard C. Long-term consequences of soman poisoning in mice: part 2. Emotional behavior. Behav Brain Res. 2008;191:95–103.PubMedCrossRef Coubard S, Béracochéa D, Collombet J-M, Philippin J-N, Krazem A, Liscia P, Lallement G, Piérard C. Long-term consequences of soman poisoning in mice: part 2. Emotional behavior. Behav Brain Res. 2008;191:95–103.PubMedCrossRef
36.
go back to reference Reddy SD, Wu X, Kuruba R, Sridhar V, Reddy DS. Magnetic resonance imaging analysis of long-term neuropathology after exposure to the nerve agent soman: correlation with histopathology and neurological dysfunction. Ann N Y Acad Sci. 2020;1480:116–35.PubMedPubMedCentralCrossRef Reddy SD, Wu X, Kuruba R, Sridhar V, Reddy DS. Magnetic resonance imaging analysis of long-term neuropathology after exposure to the nerve agent soman: correlation with histopathology and neurological dysfunction. Ann N Y Acad Sci. 2020;1480:116–35.PubMedPubMedCentralCrossRef
38.
39.
go back to reference Zhang Y, Zhang M, Zhu W, Yu J, Wang Q, Zhang J, Cui Y, Pan X, Gao X, Sun H. Succinate accumulation induces mitochondrial reactive oxygen species generation and promotes status epilepticus in the kainic acid rat model. Redox Biol. 2020;28: 101365.PubMedCrossRef Zhang Y, Zhang M, Zhu W, Yu J, Wang Q, Zhang J, Cui Y, Pan X, Gao X, Sun H. Succinate accumulation induces mitochondrial reactive oxygen species generation and promotes status epilepticus in the kainic acid rat model. Redox Biol. 2020;28: 101365.PubMedCrossRef
40.
41.
go back to reference Pauletti A, Terrone G, Shekh-Ahmad T, et al. Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy. Brain. 2019;142: e39.PubMedPubMedCentralCrossRef Pauletti A, Terrone G, Shekh-Ahmad T, et al. Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy. Brain. 2019;142: e39.PubMedPubMedCentralCrossRef
43.
go back to reference Sharma S, Puttachary S, Thippeswamy A, Kanthasamy AG, Thippeswamy T. Status epilepticus: behavioral and electroencephalography seizure correlates in kainate experimental models. Front Neurol. 2018;9:1–8.CrossRef Sharma S, Puttachary S, Thippeswamy A, Kanthasamy AG, Thippeswamy T. Status epilepticus: behavioral and electroencephalography seizure correlates in kainate experimental models. Front Neurol. 2018;9:1–8.CrossRef
44.
go back to reference Gage M, Vasanthi SS, Meyer CM, Rao NS, Thedens DR, Kannurpatti SS, Thippeswamy T. Sex-based structural and functional MRI outcomes in the rat brain after soman (GD) exposure-induced status epilepticus. Epilepsia Open. 2023;8(2):399–410.PubMedPubMedCentralCrossRef Gage M, Vasanthi SS, Meyer CM, Rao NS, Thedens DR, Kannurpatti SS, Thippeswamy T. Sex-based structural and functional MRI outcomes in the rat brain after soman (GD) exposure-induced status epilepticus. Epilepsia Open. 2023;8(2):399–410.PubMedPubMedCentralCrossRef
45.
go back to reference De Araujo FM, Lumley LA, Robison C, Tong LC, Lichtenstein S, Yourick DL. Spontaneous recurrent seizures after status epilepticus induced by soman in Sprague-Dawley rats. Epilepsia. 2010;51:1503–10.CrossRef De Araujo FM, Lumley LA, Robison C, Tong LC, Lichtenstein S, Yourick DL. Spontaneous recurrent seizures after status epilepticus induced by soman in Sprague-Dawley rats. Epilepsia. 2010;51:1503–10.CrossRef
46.
go back to reference Prager EM, Aroniadou-Anderjaska V, Almeida-Suhett CP, Figueiredo TH, Apland JP, Rossetti F, Olsen CH, Braga MFM. The recovery of acetylcholinesterase activity and the progression of neuropathological and pathophysiological alterations in the rat basolateral amygdala after soman-induced status epilepticus: relation to anxiety-like behavior. Neuropharmacology. 2014;81:64–74.PubMedPubMedCentralCrossRef Prager EM, Aroniadou-Anderjaska V, Almeida-Suhett CP, Figueiredo TH, Apland JP, Rossetti F, Olsen CH, Braga MFM. The recovery of acetylcholinesterase activity and the progression of neuropathological and pathophysiological alterations in the rat basolateral amygdala after soman-induced status epilepticus: relation to anxiety-like behavior. Neuropharmacology. 2014;81:64–74.PubMedPubMedCentralCrossRef
47.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
48.
go back to reference Rojas A, McCarren HS, Wang J, Wang W, Abreu-Melon J, Wang S, McDonough JH, Dingledine R. Comparison of neuropathology in rats following status epilepticus induced by diisopropylfluorophosphate and soman. Neurotoxicology. 2021;83:14–27.PubMedCrossRef Rojas A, McCarren HS, Wang J, Wang W, Abreu-Melon J, Wang S, McDonough JH, Dingledine R. Comparison of neuropathology in rats following status epilepticus induced by diisopropylfluorophosphate and soman. Neurotoxicology. 2021;83:14–27.PubMedCrossRef
49.
go back to reference Prager EM, Figueiredo TH, Long RP, Aroniadou-Anderjaska V, Apland JP, Braga MFM. Ly293558 prevents soman-induced pathophysiological alterations in the basolateral amygdala and the development of anxiety. Neuropharmacology. 2015;89:11–8.PubMedCrossRef Prager EM, Figueiredo TH, Long RP, Aroniadou-Anderjaska V, Apland JP, Braga MFM. Ly293558 prevents soman-induced pathophysiological alterations in the basolateral amygdala and the development of anxiety. Neuropharmacology. 2015;89:11–8.PubMedCrossRef
50.
go back to reference Figueiredo TH, Aroniadou-Anderjaska V, Qashu F, Apland JP, Pidoplichko V, Stevens D, Ferrara TM, Braga MFM. Neuroprotective efficacy of caramiphen against soman and mechanisms of its action. Br J Pharmacol. 2011;164:1495–505.PubMedPubMedCentralCrossRef Figueiredo TH, Aroniadou-Anderjaska V, Qashu F, Apland JP, Pidoplichko V, Stevens D, Ferrara TM, Braga MFM. Neuroprotective efficacy of caramiphen against soman and mechanisms of its action. Br J Pharmacol. 2011;164:1495–505.PubMedPubMedCentralCrossRef
51.
go back to reference Kundrick ER, Marrero-Rosado BM, de Araujo FM, Stone M, Schultz CR, Lumley LA. Cannabidiol reduces soman-induced lethality and seizure severity in female plasma carboxylesterase knockout mice treated with midazolam. Neurotoxicology. 2021;82:130–6.PubMedCrossRef Kundrick ER, Marrero-Rosado BM, de Araujo FM, Stone M, Schultz CR, Lumley LA. Cannabidiol reduces soman-induced lethality and seizure severity in female plasma carboxylesterase knockout mice treated with midazolam. Neurotoxicology. 2021;82:130–6.PubMedCrossRef
52.
go back to reference Lumley LA, Marrero-Rosado B, Rossetti F, Schultz CR, Stone MF, Niquet J, Wasterlain CG. Combination of antiseizure medications phenobarbital, ketamine, and midazolam reduces soman-induced epileptogenesis and brain pathology in rats. Epilepsia Open. 2021;6:757–69.PubMedPubMedCentralCrossRef Lumley LA, Marrero-Rosado B, Rossetti F, Schultz CR, Stone MF, Niquet J, Wasterlain CG. Combination of antiseizure medications phenobarbital, ketamine, and midazolam reduces soman-induced epileptogenesis and brain pathology in rats. Epilepsia Open. 2021;6:757–69.PubMedPubMedCentralCrossRef
53.
go back to reference Alexandrova EA, Aracava Y, Pereira EFR, Albuquerque EX. Pretreatment of Guinea pigs with galantamine prevents immediate and delayed effects of soman on inhibitory synaptic transmission in the hippocampus. J Pharmacol Exp Ther. 2010;334:1051–8.PubMedPubMedCentralCrossRef Alexandrova EA, Aracava Y, Pereira EFR, Albuquerque EX. Pretreatment of Guinea pigs with galantamine prevents immediate and delayed effects of soman on inhibitory synaptic transmission in the hippocampus. J Pharmacol Exp Ther. 2010;334:1051–8.PubMedPubMedCentralCrossRef
54.
go back to reference Bohnert S, van den Berg RM, Mikler J, Klaassen SD, Joosen MJA. Pharmacokinetics of three oximes in a guinea pig model and efficacy of combined oxime therapy. Toxicol Lett. 2020;324:86–94.PubMedCrossRef Bohnert S, van den Berg RM, Mikler J, Klaassen SD, Joosen MJA. Pharmacokinetics of three oximes in a guinea pig model and efficacy of combined oxime therapy. Toxicol Lett. 2020;324:86–94.PubMedCrossRef
55.
go back to reference Putra M, Gage M, Sharma S, Gardner C, Gasser G, Anantharam V, Thippeswamy T. NADPH oxidase inhibitor, diapocynin, counteracts diisopropylfluorophosphate (DFP)-induced long-term neurotoxicity in the rat model. Ann N Y Acad Sci. 2020;1479:75–93.PubMedPubMedCentralCrossRef Putra M, Gage M, Sharma S, Gardner C, Gasser G, Anantharam V, Thippeswamy T. NADPH oxidase inhibitor, diapocynin, counteracts diisopropylfluorophosphate (DFP)-induced long-term neurotoxicity in the rat model. Ann N Y Acad Sci. 2020;1479:75–93.PubMedPubMedCentralCrossRef
56.
go back to reference Pearson-Smith JN, Patel M. Antioxidant drug therapy as a neuroprotective countermeasure of nerve agent toxicity. Neurobiol Dis. 2020;133: 104457.PubMedCrossRef Pearson-Smith JN, Patel M. Antioxidant drug therapy as a neuroprotective countermeasure of nerve agent toxicity. Neurobiol Dis. 2020;133: 104457.PubMedCrossRef
57.
go back to reference Aroniadou-Anderjaska V, Figueiredo TH, Apland JP, Prager EM, Pidoplichko VI, Miller SL, Braga MFM. Long-term neuropathological and behavioral impairments after exposure to nerve agents: long-term health effects of nerve agent exposure. Ann N Y Acad Sci. 2016;1374:17–28.PubMedPubMedCentralCrossRef Aroniadou-Anderjaska V, Figueiredo TH, Apland JP, Prager EM, Pidoplichko VI, Miller SL, Braga MFM. Long-term neuropathological and behavioral impairments after exposure to nerve agents: long-term health effects of nerve agent exposure. Ann N Y Acad Sci. 2016;1374:17–28.PubMedPubMedCentralCrossRef
58.
go back to reference Koplovitz I, Romano JA, Stewart JR. Assessment of motor performance decrement following soman poisoning in mice. Drug Chem Toxicol. 1989;12:221–35.PubMedCrossRef Koplovitz I, Romano JA, Stewart JR. Assessment of motor performance decrement following soman poisoning in mice. Drug Chem Toxicol. 1989;12:221–35.PubMedCrossRef
59.
go back to reference Moffett MC, Schultz MK, Schwartz JE, Stone MF, Lumley LA. Impaired auditory and contextual fear conditioning in soman-exposed rats. Pharmacol Biochem Behav. 2011;98:120–9.PubMedCrossRef Moffett MC, Schultz MK, Schwartz JE, Stone MF, Lumley LA. Impaired auditory and contextual fear conditioning in soman-exposed rats. Pharmacol Biochem Behav. 2011;98:120–9.PubMedCrossRef
62.
go back to reference Walker DL, Miles LA, Davis M. Selective participation of the bed nucleus of the stria terminalis and CRF in sustained anxiety-like versus phasic fear-like responses. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:1291–308.PubMedPubMedCentralCrossRef Walker DL, Miles LA, Davis M. Selective participation of the bed nucleus of the stria terminalis and CRF in sustained anxiety-like versus phasic fear-like responses. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:1291–308.PubMedPubMedCentralCrossRef
63.
go back to reference Walker DL, Davis M. Double dissociation between the involvement of the bed nucleus of the stria terminalis and the central nucleus of the amygdala in startle increases produced by conditioned versus unconditioned fear. J Neurosci Off J Soc Neurosci. 1997;17:9375–83.CrossRef Walker DL, Davis M. Double dissociation between the involvement of the bed nucleus of the stria terminalis and the central nucleus of the amygdala in startle increases produced by conditioned versus unconditioned fear. J Neurosci Off J Soc Neurosci. 1997;17:9375–83.CrossRef
64.
go back to reference Aroniadou-Anderjaska V, Qashu F, Braga MFM. Mechanisms regulating GABAergic inhibitory transmission in the basolateral amygdala: implications for epilepsy and anxiety disorders. Amino Acids. 2007;32:305–15.PubMedCrossRef Aroniadou-Anderjaska V, Qashu F, Braga MFM. Mechanisms regulating GABAergic inhibitory transmission in the basolateral amygdala: implications for epilepsy and anxiety disorders. Amino Acids. 2007;32:305–15.PubMedCrossRef
65.
go back to reference Figueiredo TH, Aroniadou-Anderjaska V, Pidoplichko VI, Apland JP, Braga MFM. Antiseizure and neuroprotective efficacy of midazolam in comparison with tezampanel (LY293558) against soman-induced status epilepticus. Toxics. 2022;10:409.PubMedPubMedCentralCrossRef Figueiredo TH, Aroniadou-Anderjaska V, Pidoplichko VI, Apland JP, Braga MFM. Antiseizure and neuroprotective efficacy of midazolam in comparison with tezampanel (LY293558) against soman-induced status epilepticus. Toxics. 2022;10:409.PubMedPubMedCentralCrossRef
66.
go back to reference Figueiredo TH, Qashu F, Apland JP, Aroniadou-Anderjaska V, Souza AP, Braga MFM. The GluK1 (GluR5) Kainate/{alpha}-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor antagonist LY293558 reduces soman-induced seizures and neuropathology. J Pharmacol Exp Ther. 2011;336:303–12.PubMedPubMedCentralCrossRef Figueiredo TH, Qashu F, Apland JP, Aroniadou-Anderjaska V, Souza AP, Braga MFM. The GluK1 (GluR5) Kainate/{alpha}-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor antagonist LY293558 reduces soman-induced seizures and neuropathology. J Pharmacol Exp Ther. 2011;336:303–12.PubMedPubMedCentralCrossRef
67.
go back to reference Prager EM, Pidoplichko VI, Aroniadou-Anderjaska V, Apland JP, Braga MFM (2014) Pathophysiological mechanisms underlying increased anxiety after soman exposure: reduced GABAergic inhibition in the basolateral amygdala. Neurotoxicology 0:335–343 Prager EM, Pidoplichko VI, Aroniadou-Anderjaska V, Apland JP, Braga MFM (2014) Pathophysiological mechanisms underlying increased anxiety after soman exposure: reduced GABAergic inhibition in the basolateral amygdala. Neurotoxicology 0:335–343
68.
go back to reference Davis M, Rainnie D, Cassell M. Neurotransmission in the rat amygdala related to fear and anxiety. Trends Neurosci. 1994;17:208–14.PubMedCrossRef Davis M, Rainnie D, Cassell M. Neurotransmission in the rat amygdala related to fear and anxiety. Trends Neurosci. 1994;17:208–14.PubMedCrossRef
69.
go back to reference Tuunanen J, Halonen T, Pitkänen A. Status epilepticus causes selective regional damage and loss of GABAergic neurons in the rat amygdaloid complex. Eur J Neurosci. 1996;8:2711–25.PubMedCrossRef Tuunanen J, Halonen T, Pitkänen A. Status epilepticus causes selective regional damage and loss of GABAergic neurons in the rat amygdaloid complex. Eur J Neurosci. 1996;8:2711–25.PubMedCrossRef
70.
go back to reference Zhou R, Wang S, Zhu X. Prenatal ethanol exposure attenuates GABAergic inhibition in basolateral amygdala leading to neuronal hyperexcitability and anxiety-like behavior of adult rat offspring. Neuroscience. 2010;170:749–57.PubMedCrossRef Zhou R, Wang S, Zhu X. Prenatal ethanol exposure attenuates GABAergic inhibition in basolateral amygdala leading to neuronal hyperexcitability and anxiety-like behavior of adult rat offspring. Neuroscience. 2010;170:749–57.PubMedCrossRef
71.
go back to reference Iwata J, LeDoux JE, Meeley MP, Arneric S, Reis DJ. Intrinsic neurons in the amygdaloid field projected to by the medial geniculate body mediate emotional responses conditioned to acoustic stimuli. Brain Res. 1986;383:195–214.PubMedCrossRef Iwata J, LeDoux JE, Meeley MP, Arneric S, Reis DJ. Intrinsic neurons in the amygdaloid field projected to by the medial geniculate body mediate emotional responses conditioned to acoustic stimuli. Brain Res. 1986;383:195–214.PubMedCrossRef
72.
go back to reference LeDoux JE, Iwata J, Cicchetti P, Reis DJ. Different projections of the central amygdaloid nucleus mediate autonomic and behavioral correlates of conditioned fear. J Neurosci Off J Soc Neurosci. 1988;8:2517–29.CrossRef LeDoux JE, Iwata J, Cicchetti P, Reis DJ. Different projections of the central amygdaloid nucleus mediate autonomic and behavioral correlates of conditioned fear. J Neurosci Off J Soc Neurosci. 1988;8:2517–29.CrossRef
73.
go back to reference Hitchcock J, Davis M. Lesions of the amygdala, but not of the cerebellum or red nucleus, block conditioned fear as measured with the potentiated startle paradigm. Behav Neurosci. 1986;100:11–22.PubMedCrossRef Hitchcock J, Davis M. Lesions of the amygdala, but not of the cerebellum or red nucleus, block conditioned fear as measured with the potentiated startle paradigm. Behav Neurosci. 1986;100:11–22.PubMedCrossRef
74.
go back to reference Hitchcock JM, Davis M. Efferent pathway of the amygdala involved in conditioned fear as measured with the fear-potentiated startle paradigm. Behav Neurosci. 1991;105:826–42.PubMedCrossRef Hitchcock JM, Davis M. Efferent pathway of the amygdala involved in conditioned fear as measured with the fear-potentiated startle paradigm. Behav Neurosci. 1991;105:826–42.PubMedCrossRef
75.
go back to reference Hitchcock JM, Davis M. Fear-potentiated startle using an auditory conditioned stimulus: effect of lesions of the amygdala. Physiol Behav. 1987;39:403–8.PubMedCrossRef Hitchcock JM, Davis M. Fear-potentiated startle using an auditory conditioned stimulus: effect of lesions of the amygdala. Physiol Behav. 1987;39:403–8.PubMedCrossRef
76.
go back to reference Staunton CA, Barrett-Jolley R, Djouhri L, Thippeswamy T. Inducible nitric oxide synthase inhibition by 1400W limits pain hypersensitivity in a neuropathic pain rat model. Exp Physiol. 2018;103:535–44.PubMedCrossRef Staunton CA, Barrett-Jolley R, Djouhri L, Thippeswamy T. Inducible nitric oxide synthase inhibition by 1400W limits pain hypersensitivity in a neuropathic pain rat model. Exp Physiol. 2018;103:535–44.PubMedCrossRef
77.
go back to reference Puttachary S, Sharma S, Tse K, Beamer E, Sexton A, Crutison J, Thippeswamy T. Immediate epileptogenesis after kainate-induced status epilepticus in C57BL/6J mice: evidence from long term continuous video-EEG telemetry. PLoS ONE. 2015;10: e0131705.PubMedPubMedCentralCrossRef Puttachary S, Sharma S, Tse K, Beamer E, Sexton A, Crutison J, Thippeswamy T. Immediate epileptogenesis after kainate-induced status epilepticus in C57BL/6J mice: evidence from long term continuous video-EEG telemetry. PLoS ONE. 2015;10: e0131705.PubMedPubMedCentralCrossRef
78.
go back to reference Long Ii RP, Aroniadou-Anderjaska V, Prager EM, Pidoplichko VI, Figueiredo TH, Braga MFM. Repeated isoflurane exposures impair long-term potentiation and increase basal GABAergic activity in the basolateral amygdala. Neural Plast. 2016;2016:8524560.PubMedPubMedCentralCrossRef Long Ii RP, Aroniadou-Anderjaska V, Prager EM, Pidoplichko VI, Figueiredo TH, Braga MFM. Repeated isoflurane exposures impair long-term potentiation and increase basal GABAergic activity in the basolateral amygdala. Neural Plast. 2016;2016:8524560.PubMedPubMedCentralCrossRef
79.
go back to reference Bar-Klein G, Klee R, Brandt C, Bankstahl M, Bascuñana P, Töllner K, Dalipaj H, Bankstahl JP, Friedman A, Löscher W. Isoflurane prevents acquired epilepsy in rat models of temporal lobe epilepsy. Ann Neurol. 2016;80:896–908.PubMedCrossRef Bar-Klein G, Klee R, Brandt C, Bankstahl M, Bascuñana P, Töllner K, Dalipaj H, Bankstahl JP, Friedman A, Löscher W. Isoflurane prevents acquired epilepsy in rat models of temporal lobe epilepsy. Ann Neurol. 2016;80:896–908.PubMedCrossRef
80.
go back to reference Gage MC, Thippeswamy T. Inhibitors of Src family kinases, inducible nitric oxide synthase, and NADPH oxidase as potential CNS drug targets for neurological diseases. CNS Drugs. 2021;35:1–20.PubMedPubMedCentralCrossRef Gage MC, Thippeswamy T. Inhibitors of Src family kinases, inducible nitric oxide synthase, and NADPH oxidase as potential CNS drug targets for neurological diseases. CNS Drugs. 2021;35:1–20.PubMedPubMedCentralCrossRef
81.
go back to reference Sharma S, Carlson S, Gregory-Flores A, Hinojo-Perez A, Olson A, Thippeswamy T. Mechanisms of disease-modifying effect of saracatinib (AZD0530), a Src/Fyn tyrosine kinase inhibitor, in the rat kainate model of temporal lobe epilepsy. Neurobiol Dis. 2021;156: 105410.PubMedPubMedCentralCrossRef Sharma S, Carlson S, Gregory-Flores A, Hinojo-Perez A, Olson A, Thippeswamy T. Mechanisms of disease-modifying effect of saracatinib (AZD0530), a Src/Fyn tyrosine kinase inhibitor, in the rat kainate model of temporal lobe epilepsy. Neurobiol Dis. 2021;156: 105410.PubMedPubMedCentralCrossRef
82.
go back to reference Sharma S, Puttachary S, Thippeswamy T. Glial source of nitric oxide in epileptogenesis: a target for disease modification in epilepsy. J Neurosci Res. 2019;97:1363–77.PubMedCrossRef Sharma S, Puttachary S, Thippeswamy T. Glial source of nitric oxide in epileptogenesis: a target for disease modification in epilepsy. J Neurosci Res. 2019;97:1363–77.PubMedCrossRef
83.
go back to reference Vezzani A, Ravizza T, Balosso S, Aronica E. Glia as a source of cytokines: implications for neuronal excitability and survival. Epilepsia. 2008;49(Suppl 2):24–32.PubMedCrossRef Vezzani A, Ravizza T, Balosso S, Aronica E. Glia as a source of cytokines: implications for neuronal excitability and survival. Epilepsia. 2008;49(Suppl 2):24–32.PubMedCrossRef
84.
go back to reference Benoit ME, Tenner AJ. Complement protein C1q-mediated neuroprotection is correlated with regulation of neuronal gene and microRNA expression. J Neurosci Off J Soc Neurosci. 2011;31:3459–69.CrossRef Benoit ME, Tenner AJ. Complement protein C1q-mediated neuroprotection is correlated with regulation of neuronal gene and microRNA expression. J Neurosci Off J Soc Neurosci. 2011;31:3459–69.CrossRef
85.
go back to reference Shinjyo N, Ståhlberg A, Dragunow M, Pekny M, Pekna M. Complement-derived anaphylatoxin C3a regulates in vitro differentiation and migration of neural progenitor cells. Stem Cells Dayt Ohio. 2009;27:2824–32.CrossRef Shinjyo N, Ståhlberg A, Dragunow M, Pekny M, Pekna M. Complement-derived anaphylatoxin C3a regulates in vitro differentiation and migration of neural progenitor cells. Stem Cells Dayt Ohio. 2009;27:2824–32.CrossRef
86.
go back to reference Stevens B, Allen NJ, Vazquez LE, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131:1164–78.PubMedCrossRef Stevens B, Allen NJ, Vazquez LE, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131:1164–78.PubMedCrossRef
Metadata
Title
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
Authors
Suraj S. Vasanthi
Nikhil S. Rao
Manikandan Samidurai
Nyzil Massey
Christina Meyer
Meghan Gage
Mihir Kharate
Aida Almanza
Logan Wachter
Candide Mafuta
Lily Trevino
Adriana M. Carlo
Elijah Bryant
Brooke E. Corson
Morgan Wohlgemuth
Morgan Ostrander
Lucas Showman
Chong Wang
Thimmasettappa Thippeswamy
Publication date
01-12-2023
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2023
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-023-02847-1

Other articles of this Issue 1/2023

Journal of Neuroinflammation 1/2023 Go to the issue