<<

1521-0103/360/1/23–32$25.00 http://dx.doi.org/10.1124/jpet.116.236125 THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS J Pharmacol Exp Ther 360:23–32, January 2017 Copyright ª 2016 by The American Society for Pharmacology and Experimental Therapeutics

The M1 Muscarinic VU0255035 Delays the Development of Status Epilepticus after Organophosphate Exposure and Prevents Hyperexcitability in the Basolateral Amygdala

Steven L. Miller, Vassiliki Aroniadou-Anderjaska, Volodymyr I. Pidoplichko, Taiza H. Figueiredo, James P. Apland, Jishnu K. S. Krishnan, and Maria F. M. Braga Departments of Anatomy, Physiology, and Genetics (S.L.M., V.A.-A., V.I.P., T.H.F., J.K.S.K., M.F.M.B.) and Psychiatry (V.A.-A.,

M.F.M.B.), and Program in Neuroscience (S.L.M., V.A.-A., M.F.M.B.), F. Edward Hébert School of Medicine, Uniformed Services Downloaded from University of the Health Sciences, Bethesda, Maryland; and Neurotoxicology Branch, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland (J.P.A.) Received June 22, 2016; accepted October 27, 2016

ABSTRACT jpet.aspetjournals.org Exposure to organophosphorus toxins induces seizures that key role in seizure initiation after exposure), progress to status epilepticus (SE), which can cause brain damage VU0255035 blocked the effects produced by bath application or death. Seizures are generated by hyperstimulation of musca- of —namely, a brief barrage of spontaneous in- rinic receptors, subsequent to inhibition of ; hibitory postsynaptic currents, followed by a significant in- this is followed by glutamatergic hyperactivity, which sustains and crease in the ratio of the total charge transferred by reinforces seizure activity. It has been unclear which muscarinic spontaneous excitatory postsynaptic currents over that of receptor subtypes are involved in seizure initiation and the the inhibitory postsynaptic currents. Furthermore, paraoxon at ASPET Journals on September 28, 2021 development of SE in the early phases after exposure. Here, we enhanced the hyperpolarization-activated cation current Ih show that pretreatment of rats with the selective M1 receptor in basolateral amygdala principal cells, which could be one antagonist, VU0255035 [N-(3-oxo-3-(4-(pyridine-4-yl)piperazin- of the mechanisms underlying the increased glutamatergic 1-yl)propyl)-benzo[c][1,2,5]thiadiazole-4 sulfonamide], significantly activity, an effect that was also blocked in the presence suppressed seizure severity and prevented the development of VU0255035. Thus, selective M1 antagonists may be an of SE for about 40 minutes after exposure to paraoxon or efficacious pretreatment in contexts in which there is risk for , suggesting an important role of the M1 receptor in the exposure to organophosphates, as these antagonists will early phases of seizure generation. In addition, in in vitro brain delay the development of SE long enough for medical assis- slices of the basolateral amygdala (a brain region that plays a tance to arrive.

Introduction at synapses. Although both nico- tinic acetylcholine receptors and mAChRs can be expected to There is ample evidence to suggest that the primary mecha- be activated by the elevated acetylcholine, it is primarily the nism by which organophosphates (OPs), used as or activation of mAChRs that elicits the generation of seizures. in (nerve agents), induce prolonged, severe Evidence for this has been provided by studies showing that seizures or status epilepticus (SE) involves hyperstimulation of mAChR antagonists prevent the induction of seizures if muscarinic acetylcholine receptors (mAChRs). Thus, phosphor- administered before OP exposure (Ashani and Catravas, ylation of acetylcholinesterase by these toxins inactivates 1981; McDonough et al., 1987; Capacio and Shih, 1991; the enzyme (Sirin et al., 2012), resulting in accumulation of Skovira et al., 2010) and can arrest seizures if administered shortly after exposure (McDonough et al., 1989; Anderson et al., This research was supported by the National Institutes of Health 1997), whereas nicotinic receptor antagonists do not suppress CounterACT Program, the National Institutes of Health Office of the Director, and the National Institutes of Health National Institute of Neurologic OP-induced seizures in vivo (Shih et al., 1991; Dekundy et al., Disorders and Stroke [Grant 5U01NS058162-07]. The views of the authors 2003) or epileptiform activity in vitro (Harrison et al., 2004). do not purport to reflect the position or policies of the Department of Defense or the U.S. Army. Antagonists of mAChRs are ineffective in stopping seizures if dx.doi.org/10.1124/jpet.116.236125. injected at delayed time points after OP exposure, because

ABBREVIATIONS: ACSF, artificial cerebrospinal fluid; D-AP5, D(2)-2-amino-5-phosphonopentanoic ; BLA, basolateral amygdala; DMSO, dimethylsulfoxide; LY341495, (2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl)propanoic acid; mAChR, muscarinic ; OP, organophosphate; SCH50911, (2S)-(1)-5,5-dimethyl-2-morpholineacetic acid; SE, status epilepticus; sEPSC, spontaneous excitatory postsynaptic current; sIPSC, spontaneous inhibitory postsynaptic current; VU0255035, N-(3-oxo-3-(4-(pyridine-4-yl)piperazin-1-yl)propyl)-benzo[c] [1,2,5]thiadiazole-4 sulfonamide.

23 24 Miller et al. after the initial phase of SE, glutamatergic mechanisms that and the U.S. Army Medical Research Institute of Chemical Defense have come into play are primarily responsible for reinforcing (Aberdeen Proving Ground, MD) are accredited by the Association for and sustaining seizures (McDonough and Shih, 1997; Assessment and Accreditation of Laboratory Animal Care Interna- Weissman and Raveh, 2008). It has not been clear, however, tional. All animal experiments were conducted following the Guide for which of the mAChR subtypes are involved in the initiation of the Care and Use of Laboratory Animals by the Institute of Laboratory Animal Resources and the U.S. National Research Council and were seizures, leading to and sustaining SE in the early phases approved by the Institutional Animal Care and Use Committees of postexposure. both of our institutions. – There are five subtypes of mAChRs: M1 M5. They all are G Paraoxon and Soman Exposure. Ten-week-old male rats were protein coupled; M2 and M4 are preferentially coupled to the administered 25 mg/kg (i.p.) of the selective M1 receptor antagonist Gi class of G proteins, whereas M1,M3, and M5 are coupled to VU0255035 (Tocris Bioscience, Ellisville, MO) diluted at 25 mg/ml in the Gq family (Wess, 2003; Kruse et al., 2014). Hamilton et al. dimethylsulfoxide (DMSO) (Sigma-Aldrich, St. Louis, MO), or 1 ml/kg (1997) and Bymaster et al. (2003) found that , a (i.p.) DMSO as the vehicle, 15 minutes before exposure to soman muscarinic , induces SE in wild-type mice but not in (pinacoyl methylphosphonofluoridate; obtained from the Edgewood Chemical Biologic Center, Aberdeen Proving Ground, Edgewood, mice lacking the M1 receptor, suggesting a key role for M1 receptors in the induction of SE by pilocarpine. In addition, a MD), or 30 minutes before exposure to paraoxon (paraoxon-ethyl; Sigma-Aldrich). The concentration of VU0255035 that we used is highly selective M receptor antagonist was developed re- 1 within the range that has no significant effect on cognitive functions cently, VU0255035 [N-(3-oxo-3-(4-(pyridine-4-yl)piperazin-1-

(Sheffler et al., 2009). For the soman experiments, animals were Downloaded from yl)propyl)-benzo[c][1,2,5]thiadiazole-4 sulfonamide] (Weaver subcutaneously injected with 1.8 Â LD50 soman (198 mg/kg, diluted in et al., 2009), and was tested against seizures induced by cold physiologic saline). For the paraoxon experiments, animals were pilocarpine. Mice pretreated with VU0255035 did not develop subcutaneously injected with 4 mg/kg paraoxon; paraoxon solutions SE, at least for 40 minutes after injection of pilocarpine were prepared fresh by adding 10-ml stock solution (1.274 g/ml) to 3 ml (Sheffler et al., 2009). Because pilocarpine and OPs share ice-cold -buffered saline in a glass vial and mixing thor- the primary mechanism of seizure induction (muscarinic oughly. Soman was used because of its high toxicity and the difficulty receptor hyperstimulation), it has been suggested that in counteracting soman-induced seizures, which implies that if a drug jpet.aspetjournals.org findings in the pilocarpine model of SE may also apply to is effective in suppressing seizures induced by soman, it is likely to also be effective against other nerve agents. Paraoxon was used OP-induced SE (Tetz et al., 2006; Tang et al., 2011). Whether because it is highly potent, has similar effects to those of soman, and M1 receptor activation, which is necessary for induction of there is extensive literature on its use as a model of OP poisoning, seizures by pilocarpine, is also necessary for induction of including its use as a surrogate for chemical warfare nerve agents. The seizures by OPs was recently tested in wild-type and M1- peripheral toxic cholinergic effects of soman and paraoxon were knockout mice (Kow et al., 2015) exposed to paraoxon (O,O- controlled by intramuscular injection of either 2 mg/kg diethyl O-p-nitrophenyl phosphate), which is the active methylnitrate, or 2 mg/kg atropine sulfate along with 25 mg/kg at ASPET Journals on September 28, 2021 metabolite of the (Garcia et al., 2003). (all three drugs from Sigma-Aldrich), administered No difference was found in paraoxon-induced seizure sever- within 1 minute after injection of the OP. Seizures were monitored behaviorally in a blind fashion by two expert observers. Seizure ity between the wild-type and the M1-knockout mice, leading to the conclusion that in contrast to the significant role that severity was scored for 1 hour, using a modified version of the Racine the M receptor plays in pilocarpine-induced seizures, these (1972) scale as we described previously (Figueiredo et al., 2011): stage 0, 1 no behavioral response; stage 1, behavioral arrest; stage 2, oral/facial receptors are not involved in the generation of seizures by movements, chewing, head nodding; stage 3, unilateral/bilateral fore- paraoxon exposure (Kow et al., 2015). limb clonus without rearing, Straub tail, extended body posture; stage Delineating the mechanisms that are involved in the 4, bilateral forelimb clonus plus rearing; stage 5, rearing and falling; generation of seizures after OP exposure can result in more and stage 6, full tonic-clonic seizures. Behavioral seizures above stage targeted pretreatments and early-phase, postexposure treat- 3 indicate SE (Abdullah and Rafiqul Islam, 2012; Rossetti et al., 2012; ments that have minor or no side effects. Therefore, to shed Apland et al., 2014; Deshpande et al., 2014). – more light on the role of the M1 receptor in seizure generation Electrophysiology. Male rats, aged 25 30 days, were anesthe- by OPs, we examined the effect of pretreatment with the tized with prior to decapitation for preparation of brain slices. Coronal slices (400-mm thick) containing the amygdala were cut selective M1 antagonist VU0255035 on seizure development and severity by exposure of rats to soman or paraoxon. We as described previously (Aroniadou-Anderjaska et al., 2012). Record- ing solution [artificial cerebrospinal fluid (ACSF)] consisted of the further examined the effects of paraoxon on neuronal excit- following: 125 mM NaCl, 2.5 mM KCl, 1.25 mM NaH2PO4,21mM ability parameters in in vitro brain slices and whether M1 NaHCO3, 2 mM CaCl2, 1 mM MgCl2, and 11 mM D-glucose. The ACSF receptors are involved in these effects. The in vitro studies was saturated with 95% O2/5% CO2 to achieve a pH near 7.4, and the were performed in the basolateral amygdala (BLA), a seizure- osmolarity was adjusted to 325 mOsm with D-glucose. The slice prone amygdala nucleus (Aroniadou-Anderjaska et al., 2008), chamber (0.7-ml capacity) had continuously flowing ACSF (approxi- because there is strong evidence to suggest that the BLA plays mately 8 ml/min) at a temperature of 31–32°C. Tight-seal (.1GV), a key role in seizure generation after nerve agent exposure whole-cell recordings from principal neurons in the BLA were (McDonough et al., 1987; Prager et al., 2013). performed as described previously (Pidoplichko et al., 2014). Neurons were visualized under infrared light using Nomarski optics of an upright microscope (Axioskop 2; Zeiss, Thornwood, NY) through a Â40 Materials and Methods water immersion objective, equipped with a CCD-100 camera (Dage- MTI, Michigan City, IN). BLA principal cells were identified by their Animals. Male Sprague-Dawley rats (Charles River Laboratories, pyramidal shape and the presence of the hyperpolarization-activated

Wilmington, MA) were housed in an environmentally controlled room cationic current (Ih; Aroniadou-Anderjaska et al., 2012). Borosilicate (20–23°C, 12-hour light/dark cycle, lights on 06:00 AM), with food and glass patch electrodes had a resistance of 3.5–4.5 MV when filled with water available ad libitum. The animal care and use programs at the an internal solution of the following composition: 60 mM CsCH3SO3, Uniformed Services University of the Health Sciences (Bethesda, MD) 60 mM KCH3SO3, 10 mM EGTA, 10 mM HEPES, 5 mM KCl, 5 mM M1 Receptor Role in Seizure Generation by Organophosphates 25

Mg-ATP, and 0.3 mM Na3GTP, pH 7.2, 290 mOsm. The 5 mM KCl (198 mg/kg). All rats developed seizures, and there was no allowed the simultaneous recordings of spontaneous inhibitory post- statistically significant difference in seizure scores between synaptic currents (sIPSCs) and excitatory postsynaptic currents the two groups at 5 minutes (0.75 6 0.75 for the vehicle group – V (sEPSCs). Access resistance (15 24 M ) was regularly monitored during and 2.5 6 0.5 for the VU0255035 group; P 5 0.1) or . recordings, and cells were rejected if the resistance changed by 15% 10 minutes (3 6 0 for the vehicle group and 2.75 6 0.25 for during the experiment. Ionic currents were amplified using an Axopatch the VU0255035 group; P 5 0.3) after injection of soman. 200B amplifier (Axon Instruments, Foster City, CA) and filtered (1 kHz) with a four-pole, low-pass Bessel filter. Currents were digitally sampled However, for the next 35 minutes, Racine scale scores for the (up to 2 kHz) using pClamp 10.2 software (Molecular Devices, Sunnyvale, VU2055035 group were significantly lower compared with CA) and further analyzed using the Mini Analysis Program (Synaptosoft, the vehicle group (Fig. 1A). At 15 minutes after soman injection, Fort Lee, NJ) and Origin (OriginLab, Northampton, MA). Drugs used for the seizure score was 5.25 6 0.75 for the vehicle group and 2.75 6 the in vitro experiments were paraoxon-ethyl, VU0255035, atropine 0.25 for the VU0255035 group (P 5 0.025). At 20 minutes sulfate, D-AP5 [D-(2)-2-amino-5-phosphonopentanoic acid, an N-methyl- postexposure, seizure scores were 5.25 6 0.75 for the vehicle D-aspartate receptor antagonist; Tocris Bioscience], SCH50911 [(2S)- group and 3 6 0 for the VU0255035 group (P 5 0.025). At 1 ( )-5,5-dimethyl-2-morpholineacetic acid, an GABAB receptor antagonist; 25 minutes, seizure scores were 5.25 6 0.75 for the vehicle group Tocris Bioscience], and LY341495 [(2S)-2-amino-2-[(1S,2S)-2- and 2.75 6 0.25 for the VU0255035 group (P 5 0.020). At carboxycycloprop-1-yl]-3-(xanth-9-yl)propanoic acid, a metabotropic 30 minutes, seizure scores were 5.25 6 0.75 for the vehicle group glutamate group II/III receptor antagonist; Tocris Bioscience]. and 2.5 6 0.3 for the VU0255035 group (P 5 0.015). At 35 minutes

Statistical Analysis. Seizure score differences between the Downloaded from OP-exposed and the vehicle groups were tested for statistical signif- after soman injection, seizure scores were 5.25 6 0.75 for the icance using independent-samples t tests. In vitro electrophysiological vehicle group and 2.25 6 0.25 for the VU0255035 group (P 5 results were analyzed using paired t tests. Differences were consid- 0.009). At 40 minutes, seizure scores were 5 6 1 for the vehicle ered statistically significant when P , 0.05. Data are presented as group and 2.25 6 0.25 for the VU0255035 group (P 5 0.026). At means 6 S.E.M. Sample sizes (n) refer to the number of animals for the 45 minutes postexposure, seizure scores were 5 6 1 for the vehicle in vivo experiments and the number of recorded neurons for the whole- group and 2.25 6 0.25 for the VU0255035 group (P 5 0.026). cell experiments. During the last 15 minutes of observation, there were no jpet.aspetjournals.org significant differences in the Racine scale scores between vehicle 6 Results and VU0255035 groups, respectively, (Fig. 1A; at 50 minutes, 5 1 and 2.75 6 0.25, P 5 0.052; 55 minutes, 5 6 1and36 0.41, P 5 Effects of VU0255035 Pretreatment on Seizure Severity 0.09; and 60 minutes, 5 6 1 and 3.25 6 0.25, P 5 0.10). These after Exposure to Soman. Eight rats were randomly divided results are summarized in Table 1. into two groups: a group that was injected with VU0255035 Effects of VU0255035 Pretreatment on Seizure Se-

(25 mg/kg, n 5 4) and a group injected with the vehicle (DMSO; verity after Exposure to Paraoxon. Next, we examined at ASPET Journals on September 28, 2021 1ml/kg,n 5 4), at 15 minutes before exposure to soman whether pretreatment with VU0255035 also decreases seizure

Fig. 1. Pretreatment with the selective M1 receptor antagonist VU0255035 reduces sei- zure severity after exposure to soman or paraoxon. (A) Administration of VU0255035 (25 mg/kg), 15 minutes before exposure to soman (1.8 Â LD50), significantly reduced seizure severity scores from 15 minutes to 45 minutes after soman injection (n =4in each of the two groups). (B) Administration of VU0255035 (25 mg/kg), 30 minutes before exposure to paraoxon (4 mg/kg), significantly reduced seizure severity scores from 15 min- utes to 40 minutes after paraoxon injection (n = 6 in the VU0255035 group and n =5in the vehicle group). *P , 0.05; **P , 0.01; ***P , 0.001. 26 Miller et al.

TABLE 1 D-AP5 (50 mM), SCH50911 (10 mM), and LY341495 (3 mM); Seizure severity after exposure to soman, in VU0255035-pretreated rats under these conditions, outward currents are mediated by compared with vehicle-pretreated rats GABAA receptors, whereas inward currents are mediated by Data are presented as means 6 S.E.M. a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/kainate Behavioral Seizure Score receptors (Pidoplichko et al., 2014). Bath application of para- Time after Soman Injection P Value , at three different concentrations (0.1, 1, and 10 mM), Vehicle Group VU0255035 Group induced a transient barrage of large-amplitude sIPSCs with a min duration of 1.1 6 0.1 minutes (n 5 21), which was followed by a 6 6 5 0.75 0.75 2.5 0.5 0.1 lasting enhancement of preferentially sEPSCs (Fig. 2A). Para- 10 3 6 0 2.75 6 0.25 0.3 15 5.25 6 0.75 2.75 6 0.25 0.025* oxon took effect (appearance of the barrage of sIPSCs) within 20 5.25 6 0.75 3 6 0 0.025* 2–8 minutes after application, depending on the concentration 25 5.25 6 0.75 2.75 6 0.25 0.02* (the lower concentration of paraoxon was slower to take effect). 30 5.25 6 0.75 2.5 6 0.3 0.015* 35 5.25 6 0.75 2.25 6 0.25 0.009** To quantify the relative increase of excitatory synaptic activ- 40 5 6 1 2.25 6 0.25 0.026* ity by paraoxon, we calculated the charge transferred by the 45 5 6 1 2.25 6 0.25 0.026* excitatory and the inhibitory currents, during a 20-second time 6 6 50 5 1 2.75 0.25 0.052 window, in control conditions and then again after paraoxon 55 5 6 136 0.41 0.09 60 5 6 1 3.25 6 0.25 0.1 had taken effect. The charge transferred, in picocoulombs, was calculated as the area delimited by the excitatory or inhibitory Downloaded from , , *P 0.05; **P 0.01. current and the baseline. The ratio of the charge transferred by sEPSCs to the charge transferred by sIPSCs was increased severity after exposure to paraoxon. Because behavioral seizure significantly in the presence of paraoxon. When 0.1 mMpara- scores did not differ between the VU0255035 group and the oxon was applied, the sEPSC/sIPSC ratio was increased from vehicle group during the first 10 minutes after soman exposure 0.6 6 0.1 in control conditions to 1.4 6 0.3 at 17 minutes after (Fig. 1A), with VU0255035 administered 15 minutes before paraoxon application (P , 0.05, n 5 7). When 1 mM paraoxon jpet.aspetjournals.org soman injection, this time we administered VU0255035 30 min- was applied, the sEPSC/sIPSC ratio was increased from 0.6 6 utes before paraoxon exposure, considering that perhaps 15 min- 0.1 in control conditions to 0.9 6 0.1 at 9 minutes after paraoxon utes were not sufficient for VU0255035 to take full effect. Eleven application (P , 0.05, n 5 4). Finally, when 10 mM paraoxon rats were randomly divided into two groups: a group that was was applied, the ratio of sEPSC/sIPSC charge transfer was injected with 25 mg/kg VU0255035 (n 5 6)andagroupinjected increased from 0.8 6 0.1 in control conditions to 1.1 6 0.1 at with the vehicle (DMSO; 1 ml/kg, n 5 5), at 30 minutes before 6 minutes after paraoxon application (P , 0.01, n 5 10; Fig. 2B). exposure to paraoxon (4 mg/kg). All rats developed seizures. To determine whether mAChRs were involved in mediating at ASPET Journals on September 28, 2021 Again,theRacinescoredidnotdiffersignificantlybetweenthe the effects of paraoxon, first, we pretreated the slices with vehicle group and the VU0255035 group, respectively, during atropine (1 mM) for about 4 minutes, and then applied 10 mM the first 10 minutes after paraoxon injection (at 5 minutes, paraoxon, while recording from principal BLA neurons. The 3.33 6 0.24 and 3.4 6 0.21, P 5 0.8; and at 10 minutes, 4.33 6 holding potential was again 258 mV, and D-AP5 (50 mM), 0.8 and 3.8 6 0.33, P 5 0.5). However, for the next 30 minutes, SCH50911 (10 mM), and LY341495 (3 mM) were present in the Racine scale scores for the VU2055035 group were significantly slice medium. Paraoxon, in the presence of atropine, had no lower compared with the vehicle group (Fig. 1B). At 15 minutes significant effect on sIPSCs or sEPSCs (Fig. 3A, top traces). after paraoxon injection, the seizure score was 4 6 0.37 for the The total charge transferred by sIPSCs or sEPSCs during a vehicle group and 2.2 6 0.26 for the VU0255035 group (P 5 20-second time window, 10 minutes after paraoxon applica- 0.003). At 20 minutes postexposure, seizure scores were 5 6 0.51 tion, was expressed as a percentage of the total charge for the vehicle group and 2.4 6 0.37 for the VU0255035 group transferred, during 20 seconds, in control conditions (in the (P 5 0.002). At 25 minutes, seizure scores were 5.33 6 0.37 for the vehicle group and 2.2 6 0.33 for the VU0255035 group (P 5 TABLE 2 0.0001). At 30 minutes, seizure scores were 4.67 6 0.51 for the 6 5 Seizure severity after exposure to paraoxon in VU0255035-pretreated vehicle group and 2.4 0.33 for the VU0255035 group (P rats compared with vehicle-pretreated rats 0.004). At 35 minutes after paraoxon injection, seizure scores Data are presented as means 6 S.E.M. were 4.83 6 0.58 for the vehicle group and 2.8 6 0.25 for the Behavioral Seizure Score VU0255035 group (P 5 0.010). At 40 minutes, seizure scores Time after Paraoxon Injection P Value were 4.33 6 0.51 for the vehicle group and 2.6 6 0.21 for the Vehicle Group VU0255035 Group VU0255035 group (P 5 0.008). During the last 20 minutes of min observation, there were no significant differences in the Racine 5 3.33 6 0.24 3.4 6 0.21 0.8 scale scores between vehicle and VU0255035 groups, respec- 10 4.33 6 0.8 3.8 6 0.33 0.5 6 6 5 15 4 6 0.37 2.2 6 0.26 0.003* tively (Fig. 1B; at 45 minutes, 4.67 0.51 and 3.6 0.33, P 6 6 6 6 5 20 5 0.51 2.4 0.37 0.002* 0.104; at 50 minutes, 4.83 0.2 and 4.2 0.31, P 0.134; at 25 5.33 6 0.37 2.2 6 0.33 0.0001** 55 minutes, 5 6 0.24 and 4.4 6 0.26, P 5 0.131; and at 30 4.67 6 0.51 2.4 6 0.33 0.004* 60 minutes, 4.83 6 0.32 and 5 6 0.17, P 5 0.635). These results 35 4.83 6 0.58 2.8 6 0.25 0.01*** 40 4.33 6 0.51 2.6 6 0.21 0.008* are summarized in Table 2. 45 4.67 6 0.51 3.6 6 0.33 0.104 Effects of Paraoxon on Spontaneous Synaptic Activ- 50 4.83 6 0.2 4.2 6 0.31 0.134 ity in the BLA and the Role of M Receptors. Whole-cell 55 5 6 0.24 4.4 6 0.26 0.131 1 6 6 simultaneous recordings of sIPSCs and sEPSCs were obtained 60 4.83 0.32 5 0.17 0.635 from principal BLA neurons, at Vh of 258 mV, in the presence of *P , 0.05; **P , 0.01; ***P , 0.001. M1 Receptor Role in Seizure Generation by Organophosphates 27 Downloaded from jpet.aspetjournals.org at ASPET Journals on September 28, 2021

Fig. 2. Paraoxon enhances both sIPSCs and sEPSCs, with a significantly greater, lasting effect on the sEPSCs, in the BLA. Whole-cell simultaneous recordings of sIPSCs and sEPSCs were obtained from BLA principal cells (Vh = 258). (A) Representative examples of the effects of paraoxon at 0.1 mM, 1 mM, and 10 mM (traces are from three different neurons). Outward currents (upward deflections) are GABAergic and inward currents (downward deflections) are glutamatergic. The number of minutes between successive traces in a row is the time lapse from the time point paraoxon was applied, and then the interval between the two successive recordings. (B) Group data of the effects of paraoxon on the ratio of the charge transferred by sEPSCs over the charge transferred by sIPSCs. The charge transferred was calculated during a 20-second time window, in control conditions and after bath application of paraoxon. Calculations were made at 17 minutes after application of 0.1 mM paraoxon (n = 7; left), at 9 minutes after application of 1 mM paraoxon (n = 4; middle), and at 6 minutes after application of 10 mM paraoxon (n = 10; right). *P , 0.05; **P , 0.01. 28 Miller et al. presence of atropine). When 1 mM atropine was present in the effects of 10 mM paraoxon in the absence of muscarinic slice medium, the charge transferred by sIPSCs after 10 min- receptor antagonists (Fig. 2A, third row of traces). Under utes in paraoxon was 72% 6 25% of the control (n 5 7, P 5 these conditions, the charge transferred by sIPSCs was 0.203), whereas the charge transferred by sEPSCs was 79% 6 increased by paraoxon to 275% 6 57% of the control (n 5 10, 23% of the control (n 5 7, P 5 0.055; Fig. 3B, left bar graph). P 5 0.0001), and the charge transferred by sEPSCs was Next, we performed similar experiments in the presence of increased by paraoxon to 435% 6 80% of the control (n 5 10, VU0255035, to determine whether the M1 receptor was P 5 0.00002; Fig. 3B, right bar graph). The difference between involved in mediating the effects of paraoxon. In 10 mM the increase in sIPSCs and the increase in sEPSCs was VU0255035, paraoxon had no significant effect on sIPSCs or statistically significant (P , 0.001). sEPSCs (Fig. 3A, lower traces); the charge transferred by Effects of Paraoxon on the Ih Current. Previous sIPSCs after 10 minutes in paraoxon was 75% 6 17% of the studies have shown that one of the mechanisms by which M1 VU0255035-containing control (n 5 7, P 5 0.156), whereas receptor activation depolarizes pyramidal neurons in the hippo- the charge transferred by sEPSCs was 81% 6 16% of the campus is the increase of the hyperpolarization-activated, mixed 1 1 VU0255035-containing control (n 5 7, P 5 0.389; Fig. 3B, Na /K current (Ih; Fisahn et al., 2002). The preferential middle bar graph). These results are in contrast with the increase of spontaneous glutamatergic activity by paraoxon, in Downloaded from jpet.aspetjournals.org at ASPET Journals on September 28, 2021

Fig. 3. M1 muscarinic receptors mediate the effects of paraoxon on sIPSCs and sEPSCs in the BLA. Whole-cell simultaneous recordings of sIPSCs and sEPSCs were obtained from BLA principal cells (Vh = 258). (A) Representative traces show that 10 mM paraoxon had no significant effect when applied in the presence of atropine (1 mM, top row of traces) or VU0255035 (10 mM, second row of traces). (B) Effects of 10 mM paraoxon on sIPSCs and sEPSCs under three conditions: pretreatment with atropine (left bar graph; n = 7), pretreatment with VU0255035 (middle bar graph; n = 7), and in the absence of muscarinic receptor antagonists (right bar graph; n = 10). The total charge transferred by sIPSCs or sEPSCs during a 20-second time window, 10 minutes after paraoxon application, was expressed as a percentage of the total charge transferred, during 20 seconds, in control conditions. The total charge transferred by sIPSCs and sEPSCs was increased by paraoxon only when there was no atropine or VU0255035 in the slice medium, and the difference between the increase in sIPSCs and the increase in sEPSCs was statistically significant. ***P , 0.001. M1 Receptor Role in Seizure Generation by Organophosphates 29 the BLA, suggests that paraoxon may depolarize principal such that the ratio of the charge transferred by sEPSCs over BLA neurons. To determine whether such an effect could be that of sIPSCs is increased; these effects of paraoxon are mediated via enhancement of the Ih current, we evoked the Ih in blocked in the presence of VU0255035. Finally, we found that 1 1 principal BLA neurons by voltage steps from a Vh of 270 mV paraoxon increases the Ih Na /K current in BLA principal to 2110 mV, in 10-mV increments. Bath application of 10 mM cells, an effect that is also blocked by VU0255035. The paraoxon had a consistent and clear enhancing effect on the Ih depolarization that the increase of Ih can induce may be one current (n 5 4; Fig. 4A). Addition of 10 mM VU0255035 to the of the mechanisms by which paraoxon increases spontane- slice medium reversed the effect of paraoxon (Fig. 4B). Bath ous excitatory activity in the BLA. application of 10 mM VU0255035 alone (in the absence of Knowledge of the specific subtypes of mAChRs that are paraoxon) had no effect on the Ih (Fig. 4C). When 10 mM involved in the initiation of SE after exposure to an OP can paraoxon was added to the recording solution in the presence of lead to the development of pretreatments or early postexpo- 10 mM VU0255035, there was no effect on the Ih (Fig. 4D). sure treatments that do not have the side effects of nonselective These results suggest that the enhancement of Ih by paraoxon mAChR antagonists. The role of M1 receptors, however, in was mediated by M1 receptor activation. seizure induction by OPs has been unclear. Previous studies have shown that pilocarpine, a , induces Discussion severe seizures in wild-type mice and in mice lacking the M2, M3,M4,orM5 receptor (Bymaster et al., 2003), but not in mice Downloaded from This study demonstrated that pretreatment with the selec- lacking the M1 receptor (Hamilton et al., 1997; Bymaster et al., tive M1 receptor antagonist VU0255035 significantly sup- 2003). In consistency with these studies, pretreatment of wild- presses behavioral seizures induced by either soman or type mice with VU0255035 decreased the severity of seizures paraoxon, for at least 40 minutes after exposure, preventing induced by pilocarpine; the effect was significant at 35 and the development of SE during this time period. The in vitro 40 minutes after pilocarpine injection, resulting in seizure experiments aiming to uncover some of the mechanisms severity below the level of SE at least for the first 40 minutes involved in seizure generation by paraoxon and the involve- postinjection (Sheffler et al., 2009). Since both pilocarpine and jpet.aspetjournals.org ment of M1 receptors showed that paraoxon enhances both OPs are known to induce seizures by mAChR hyperstimulation, inhibitory and excitatory spontaneous activity recorded from it is reasonable to suggest that M1 receptors may also be BLA principal cells, with a lasting greater increase in sEPSCs, important in the induction of seizures by OPs, including nerve at ASPET Journals on September 28, 2021

Fig. 4. Paraoxon enhances the hyperpo- larization-activated cation current (Ih)in principal BLA neurons, via M1 muscarinic receptor activation. (A) Bath application of 10 mM paraoxon increased the Ih current elicited by hyperpolarizing steps (at 10-mV increments) from a Vh of 270 mV. The recordings shown are 3 minutes after appli- cation of paraoxon. (B) Addition of 10 mM VU0255035 to the slice medium reversed the effects of paraoxon. (A) and (B) are from thesameneuron.(C)Applicationof10mM VU0255035 alone had no effect on the Ih.(D) Bath application of 10 mMparaoxoninthe presence of 10 mM VU0255035 had no effect on the Ih. (C) and (D) are from the same neuron [different from (A) and (B)]. 30 Miller et al. agents (Bhattacharjee et al., 2013). However, Kow et al. (2015) develop SE when exposed to high doses of soman, the activity of found no differences between wild-type and M1 knockout mice acetylcholinesterase is reduced in the hippocampus and the in seizure severity scores or proportion of animals displaying at piriform cortex to a similar extent as in the rats who develop least one tonic-clonic seizure after exposure to paraoxon; it was SE, but to a significantly lesser extent in the BLA; this suggests concluded that pilocarpine and paraoxon induce seizures by a critical role of acetylcholinesterase reduction in the BLA for different mechanisms. This study, however, in rats shows that SE to be induced. The importance of the amygdala in nerve the M1 receptor plays a central role in the severity of seizures agent–induced seizure generation is also highlighted by the induced by paraoxon, at least during the first 40 minutes after findings that the amygdala displays the earliest increase in the exposure. This discrepancy may imply species differences in extracellular glutamate after exposure to soman (Lallement the role of the M1 receptor in OP-induced seizures. et al., 1991) and generates ictal-like discharges after in vitro The data in our study make it evident that during the first application of soman (Apland et al., 2009). It is to be expected, 10 minutes after OP injection, low-intensity seizures were therefore, that when the BLA is exposed to an OP, neuronal initiated despite the VU0255035 pretreatment. Behavioral excitability will increase, which will be reflected in an increase seizures stage 2 to 3 and slightly higher, which correspond to of spontaneous glutamatergic activity. Indeed, in this study, epileptic discharges on EEG (Abdullah and Rafiqul Islam, paraoxon increased sEPSCs in the BLA significantly more than 2012), were observed during the first 10 minutes after soman the increase of sIPSCs. or paraoxon injection in the VU0255035-treated rats as in the The in vitro effects of paraoxon were blocked in the presence vehicle-treated rats, whereas the effects of VU0255035 pre- of VU0255035, suggesting that both the increase in sIPSCs Downloaded from treatment became significant after 10 minutes postexposure. and that in sEPSCs were mediated via M1 receptor activation. Pharmacokinetic studies have shown that VU0255035 has The cellular location of these receptors where paraoxon acted already reached high concentrations in the brain at 30 min- remains to be determined. Cholinergic synapses in the BLA utes after the injection (Sheffler et al., 2009). Therefore, the are formed by the dense cholinergic afferents arising from the absence of a significant effect of the VU0255035 pretreat- basal forebrain (Ben-Ari et al., 1977; Nagai et al., 1982; ment during the first 10 minutes is probably not attributable Carlsen and Heimer, 1986; Muller et al., 2011) but also by jpet.aspetjournals.org to a delay of the drug reaching the brain. Other mAChR an intrinsic population of small cholinergic neurons (Carlsen subtypes may be involved in the generation of seizure activity and Heimer, 1986). Cholinergic nerve terminals synapse on at this early phase, or an increase of glutamate may be the both pyramidal cells and interneurons (Carlsen and Heimer, culprit, since extracellular glutamate has been found to 1986; Nitecka and Frotscher, 1989; Muller et al., 2011), but the increase in the amygdala within the first 10 minutes after majority of the cholinergic input is directed onto pyramidal exposure to soman (Lallement et al., 1991). During the next neurons (Muller et al., 2011). From the different types of

30 minutes (after the initial 10-minute period), the M1 mAChRs, both the M1 and M2 subtypes, are present in the rat at ASPET Journals on September 28, 2021 antagonist in the VU0255035-treated group prevented SE BLA (Cortés and Palacios, 1986; Mash and Potter, 1986; (stage 3 and higher), but seizures below stage 3 were ongoing; Spencer et al., 1986; Mash et al., 1988), as well as the M3 again, other mAChR subtypes and/or glutamatergic mecha- and/or M4 receptors (Smith et al., 1991), but the M1 receptors nisms may be involved in sustaining low-intensity seizures predominate (Buckley et al., 1988). Immunocytochemistry during this postexposure period. After the 40-minute postexpo- along with electron microscopy have revealed that M1 recep- sure, the seizure-suppressing effects of VU0255035 pretreatment tors in the rat BLA are located on somata, dendritic shafts, were no longer significant. Since VU0255035 concentrations in and, to a lesser extent, spines of pyramidal neurons, as well as the brain remain relatively high for 2 hours after injection on aspiny dendrites with morphologic features typical of (Sheffler et al., 2009), the reduction in the seizure-suppressing interneurons (Muller et al., 2013). In addition, M1 immunore- effect of VU0255035 after the 40-minute period postexposure activity has been found on presynaptic terminals, most of may not be attributable to the clearance of the compound from which formed asymmetric synapses with dendritic spines, but the brain but mainly reflects reduced significance of the M1 some of which formed symmetrical synapses with neuronal receptors, because glutamatergic mechanisms take over the somata and dendritic shafts (Muller et al., 2013). It appears, primary role in sustaining and reinforcing seizures (McDonough therefore, that M1 receptors in the rat BLA are in a position to and Shih, 1997; Weissman and Raveh, 2008). Thus, M1 receptors affect the activity of both glutamatergic and GABAergic appear to be important during a certain time period after OP neurons, by postsynaptic or presynaptic mechanisms. The exposure (after the first 10 minutes and before 1 hour). This increase in GABAergic inhibitory activity by paraoxon could implies that if the effects of an M1 antagonist or the consequences be due to M1 receptor–mediated direct depolarization of of knocking out the M1 receptor (in mice) on seizure induction by GABAergic interneurons, facilitation of GABA release from pilocarpine or OPs are averaged over a period of time, instead of GABAergic presynaptic terminals, or depolarization of gluta- being evaluated every 5 minutes or during the 10–40 minutes matergic neurons or terminals synapsing onto inhibitory postexposure, a significant role of the receptor may be missed. interneurons; presynaptic facilitation of GABA release may The in vitro experiments were performed in the BLA because, be the least likely of the three possibilities, because M1 in addition to the importance of other brain regions in seizure receptor activation inhibits GABA release in the rat lateral generation by OPs (Myhrer, 2007; Myhrer et al., 2007, 2010), amygdala (Sugita et al., 1991). The increase in sEPSCs by there is strong evidence to suggest that the BLA plays a key role paraoxon could be the result of M1 receptor–mediated de- (Aroniadou-Anderjaska et al., 2009). Thus, when McDonough polarization of BLA principal neurons via enhancement of the et al. (1987) microinjected nerve agents into different brain Ih current, as shown previously in CA3 pyramidal neurons in regions, they observed that convulsions were elicited only when the hippocampus (Fisahn et al., 2002) and in BLA principal the nerve agent was injected into the BLA. Prager et al. (2013) neurons in this study. In addition, M1 receptors on gluta- recently found that in the small proportion of rats who do not matergic terminals may facilitate glutamate release; this M1 Receptor Role in Seizure Generation by Organophosphates 31 remains to be demonstrated in the BLA, but in granule cells Cortés R and Palacios JM (1986) Muscarinic cholinergic receptor subtypes in the rat brain. I. Quantitative autoradiographic studies. Brain Res 362:227–238. of the hippocampus, the frequency and amplitude of sEPSCs Dekundy A, Kaminski RM, and Turski WA (2003) improves beneficial are increased by application of paraoxon via a presynaptic effects of cholinergic antagonists in anticholinesterase-treated mice. Toxicol Sci 72: 289–295. mechanism, and this effect is blocked by preapplication of Deshpande LS, Carter DS, Phillips KF, Blair RE, and DeLorenzo RJ (2014) Devel- atropine (Kozhemyakin et al., 2010). opment of status epilepticus, sustained calcium elevations and neuronal injury in a Up to 30 mg/kg VU0255035 has been used in mice to block rat survival model of lethal paraoxon intoxication. Neurotoxicology 44:17–26. Figueiredo TH, Qashu F, Apland JP, Aroniadou-Anderjaska V, Souza AP, and Braga M1 receptors, and no effect was observed on learning as de- MF (2011) The GluK1 (GluR5) kainate/alpha-amino-3-hydroxy-5-methyl-4- termined by contextual fear conditioning (Sheffler et al., 2009). In isoxazolepropionic acid receptor antagonist LY293558 reduces soman-induced seizures and neuropathology. J Pharmacol Exp Ther 336:303–312. M1 knockout mice, Miyakawa et al. (2001) found evidence of Fisahn A, Yamada M, Duttaroy A, Gan JW, Deng CX, McBain CJ, and Wess J (2002) hyperactivity under stressful conditions and no significant cogni- Muscarinic induction of hippocampal gamma oscillations requires coupling of the M1 receptor to two mixed cation currents. Neuron 33:615–624. tive impairments in the Morris water maze and in contextual fear Garcia SJ, Abu-Qare AW, Meeker-O’Connell WA, Borton AJ, and Abou-Donia MB conditioning, whereas Anagnostaras et al. (2003) found enhance- (2003) Methyl parathion: a review of health effects. J Toxicol Environ Health B Crit Rev 6:185–210. ments, or deficits, or no effect depending on the type of memory. Hamilton SE, Loose MD, Qi M, Levey AI, Hille B, McKnight GS, Idzerda RL, Taken together with the results presented here, it is suggested and Nathanson NM (1997) Disruption of the m1 receptor gene ablates muscarinic receptor-dependent M current regulation and seizure activity in mice. Proc Natl that selective M1 antagonists may be an efficacious pretreatment Acad Sci USA 94:13311–13316. in situations in which there is risk for exposure to OPs because the Harrison PK, Sheridan RD, Green AC, Scott IR, and Tattersall JE (2004) A guinea pig hippocampal slice model of organophosphate-induced seizure activity. JPharmacol antagonist may produce only minimal or no significant side effects – Exp Ther 310:678 686. Downloaded from on cognitive function, whereas, upon exposure, it will delay the Kow RL, Cheng EM, Jiang K, Le JH, Stella N, and Nathanson NM (2015) Muscarinic development of SE long enough for medical assistance to arrive. M1 receptor and cannabinoid CB1 receptor do not modulate paraoxon-induced seizures. Pharmacol Res Perspect 3:e00100. Kozhemyakin M, Rajasekaran K, and Kapur J (2010) Central in- Authorship Contributions hibition enhances glutamatergic synaptic transmission. J Neurophysiol 103: 1748–1757. Participated in research design: Braga, Miller, Aroniadou-Anderjaska, Kruse AC, Kobilka BK, Gautam D, Sexton PM, Christopoulos A, and Wess J (2014) Muscarinic acetylcholine receptors: novel opportunities for drug development. Nat Apland. – Rev Drug Discov 13:549 560. jpet.aspetjournals.org Conducted experiments: Miller, Pidoplichko, Figueiredo, Apland, Lallement G, Carpentier P, Collet A, Pernot-Marino I, Baubichon D, and Blanchet G Krishnan. (1991) Effects of soman-induced seizures on different extracellular amino acid Performed data analysis: Miller, Pidoplichko, Figueiredo. levels and on glutamate uptake in rat hippocampus. Brain Res 563:234–240. Mash DC and Potter LT (1986) Autoradiographic localization of M1 and M2 mus- Wrote or contributed to the writing of the manuscript: Aroniadou- carine receptors in the rat brain. Neuroscience 19:551–564. Anderjaska, Apland, Braga. Mash DC, White WF, and Mesulam MM (1988) Distribution of muscarinic receptor subtypes within architectonic subregions of the primate cerebral cortex. J Comp Neurol 278:265–274. References McDonough JH, Jr, Jaax NK, Crowley RA, Mays MZ, and Modrow HE (1989) Atro- pine and/or therapy protects against soman-induced neural and cardiac

Abdullah JM and Rafiqul Islam M (2012) Telemetric EEG and the rat: a guide for at ASPET Journals on September 28, 2021 neuroscientists. Malays J Med Sci 19:1–5. pathology. Fundam Appl Toxicol 13:256–276. Anagnostaras SG, Murphy GG, Hamilton SE, Mitchell SL, Rahnama NP, Nathanson McDonough JH, Jr, McLeod CG, Jr, and Nipwoda MT (1987) Direct microinjection of NM, and Silva AJ (2003) Selective cognitive dysfunction in acetylcholine M1 soman or VX into the amygdala produces repetitive limbic convulsions and neu- muscarinic receptor mutant mice. Nat Neurosci 6:51–58. ropathology. Brain Res 435:123–137. Anderson DR, Harris LW, Chang FC, Baze WB, Capacio BR, Byers SL, and Lennox McDonough JH, Jr and Shih TM (1997) Neuropharmacological mechanisms of nerve WJ (1997) Antagonism of soman-induced convulsions by midazolam, diazepam and agent-induced seizure and neuropathology. Neurosci Biobehav Rev 21:559–579. . Drug Chem Toxicol 20:115–131. Miyakawa T, Yamada M, Duttaroy A, and Wess J (2001) Hyperactivity and intact Apland JP, Aroniadou-Anderjaska V, and Braga MF (2009) Soman induces hippocampus-dependent learning in mice lacking the M1 muscarinic acetylcholine ictogenesis in the amygdala and interictal activity in the hippocampus that are receptor. J Neurosci 21:5239–5250. blocked by a GluR5 kainate receptor antagonist in vitro. Neuroscience 159: Muller JF, Mascagni F, and McDonald AJ (2011) Cholinergic innervation of pyra- 380–389. midal cells and parvalbumin-immunoreactive interneurons in the rat basolateral Apland JP, Aroniadou-Anderjaska V, Figueiredo TH, Rossetti F, Miller SL, amygdala. J Comp Neurol 519:790–805. and Braga MF (2014) The limitations of diazepam as a treatment for nerve agent- Muller JF, Mascagni F, Zaric V, and McDonald AJ (2013) Muscarinic cholinergic induced seizures and neuropathology in rats: comparison with UBP302. JPharmacol receptor M1 in the rat basolateral amygdala: ultrastructural localization and Exp Ther 351:359–372. synaptic relationships to cholinergic axons. J Comp Neurol 521:1743–1759. Aroniadou-Anderjaska V, Figueiredo TH, Apland JP, Qashu F, and Braga MF (2009) Myhrer T (2007) Neuronal structures involved in the induction and propagation of Primary brain targets of nerve agents: the role of the amygdala in comparison to seizures caused by nerve agents: implications for medical treatment. Toxicology the hippocampus. Neurotoxicology 30:772–776. 239:1–14. Aroniadou-Anderjaska V, Fritsch B, Qashu F, and Braga MF (2008) Pathology and Myhrer T, Enger S, and Aas P (2007) Anticonvulsant effects of damage to structures pathophysiology of the amygdala in epileptogenesis and epilepsy. Epilepsy Res 78: involved in seizure induction in rats exposed to soman. Neurotoxicology 28:819–828. 102–116. Myhrer T, Enger S, and Aas P (2010) Roles of perirhinal and posterior piriform Aroniadou-Anderjaska V, Pidoplichko VI, Figueiredo TH, Almeida-Suhett CP, Prager cortices in control and generation of seizures: a microinfusion study in rats exposed EM, and Braga MF (2012) Presynaptic facilitation of glutamate release in the to soman. Neurotoxicology 31:147–153. basolateral amygdala: a mechanism for the anxiogenic and seizurogenic function of Nagai T, Kimura H, Maeda T, McGeer PL, Peng F, and McGeer EG (1982) Cholin- GluK1 receptors. Neuroscience 221:157–169. ergic projections from the basal forebrain of rat to the amygdala. J Neurosci 2: Ashani Y and Catravas GN (1981) Seizure-induced changes in the permeability of the 513–520. blood-brain barrier following administration of anticholinesterase drugs to rats. Nitecka L and Frotscher M (1989) Organization and synaptic interconnections of Biochem Pharmacol 30:2593–2601. GABAergic and cholinergic elements in the rat amygdaloid nuclei: single- and Ben-Ari Y, Zigmond RE, Shute CC, and Lewis PR (1977) Regional distribution of double-immunolabeling studies. J Comp Neurol 279:470–488. acetyltransferase and acetylcholinesterase within the amygdaloid complex Pidoplichko VI, Aroniadou-Anderjaska V, Prager EM, Figueiredo TH, Almeida- and stria terminalis system. Brain Res 120:435–444. Suhett CP, Miller SL, and Braga MF (2014) ASIC1a activation enhances inhibition Bhattacharjee AK, Pomponio JW, Evans SA, Pervitsky D, and Gordon RK (2013) in the basolateral amygdala and reduces anxiety. J Neurosci 34:3130–3141. Discovery of subtype selective muscarinic receptor antagonists as alternatives to Prager EM, Aroniadou-Anderjaska V, Almeida-Suhett CP, Figueiredo TH, Apland atropine using in silico pharmacophore modeling and virtual screening methods. JP, and Braga MF (2013) Acetylcholinesterase inhibition in the basolateral amyg- Bioorg Med Chem 21:2651–2662. dala plays a key role in the induction of status epilepticus after soman exposure. Buckley NJ, Bonner TI, and Brann MR (1988) Localization of a family of muscarinic Neurotoxicology 38:84–90. receptor mRNAs in rat brain. J Neurosci 8:4646–4652. Racine RJ (1972) Modification of seizure activity by electrical stimulation. II. Motor Bymaster FP, McKinzie DL, Felder CC, and Wess J (2003) Use of M1-M5 muscarinic seizure. Electroencephalogr Clin Neurophysiol 32:281–294. receptor knockout mice as novel tools to delineate the physiological roles of the Rossetti F, de Araujo Furtado M, Pak T, Bailey K, Shields M, Chanda S, Addis M, muscarinic cholinergic system. Neurochem Res 28:437–442. Robertson BD, Moffett M, Lumley LA, et al. (2012) Combined diazepam and HDAC Capacio BR and Shih TM (1991) Anticonvulsant actions of drugs in inhibitor treatment protects against seizures and neuronal damage caused by soman poisoning. Epilepsia 32:604–615. soman exposure. Neurotoxicology 33:500–511. Carlsen J and Heimer L (1986) A correlated light and electron microscopic immu- Sheffler DJ, Williams R, Bridges TM, Xiang Z, Kane AS, Byun NE, Jadhav S, Mock nocytochemical study of cholinergic terminals and neurons in the rat amygdaloid MM, Zheng F, Lewis LM, et al. (2009) A novel selective muscarinic acetylcholine body with special emphasis on the basolateral amygdaloid nucleus. J Comp Neurol receptor subtype 1 antagonist reduces seizures without impairing hippocampus- 244:121–136. dependent learning. Mol Pharmacol 76:356–368. 32 Miller et al.

Shih TM, Koviak TA, and Capacio BR (1991) Anticonvulsants for poisoning by the Tetz LM, Rezk PE, Ratcliffe RH, Gordon RK, Steele KE, and Nambiar MP (2006) organophosphorus compound soman: pharmacological mechanisms. Neurosci Bio- Development of a rat pilocarpine model of seizure/status epilepticus that mimics behav Rev 15:349–362. chemical warfare nerve agent exposure. Toxicol Ind Health 22:255–266. Sirin GS, Zhou Y, Lior-Hoffmann L, Wang S, and Zhang Y (2012) Aging mechanism of Weaver CD, Sheffler DJ, Lewis LM, Bridges TM, Williams R, Nalywajko NT, soman inhibited acetylcholinesterase. J Phys Chem B 116:12199–12207. Kennedy JP, Mulder MM, Jadhav S, Aldrich LA, et al. (2009) Discovery and Skovira JW, McDonough JH, and Shih TM (2010) Protection against -induced development of a potent and highly selective small molecule muscarinic acetyl- seizures in rats by direct brain microinjection of scopolamine, midazolam or choline receptor subtype I (mAChR 1 or M1) antagonist in vitro and in vivo probe. MK-801. J Mol Neurosci 40:56–62. Curr Top Med Chem 9:1217–1226. Smith TD, Annis SJ, Ehlert FJ, and Leslie FM (1991) N-[3H]methylscopolamine Weissman BA and Raveh L (2008) Therapy against organophosphate poisoning: the labeling of non-M1, non-M2 muscarinic receptor binding sites in rat brain. J importance of anticholinergic drugs with antiglutamatergic properties. Toxicol Pharmacol Exp Ther 256:1173–1181. Appl Pharmacol 232:351–358. Spencer DG, Jr, Horváth E, and Traber J (1986) Direct autoradiographic de- Wess J (2003) Novel insights into muscarinic acetylcholine receptor function using termination of M1 and M2 muscarinic acetylcholine receptor distribution in the rat gene targeting technology. Trends Pharmacol Sci 24:414–420. brain: relation to cholinergic nuclei and projections. Brain Res 380:59–68. Sugita S, Uchimura N, Jiang ZG, and North RA (1991) Distinct muscarinic receptors inhibit release of gamma-aminobutyric acid and excitatory amino in mam- Address correspondence to: Dr. Maria F. M. Braga, Department of malian brain. Proc Natl Acad Sci USA 88:2608–2611. Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Tang FR, Loke WK, and Ling EA (2011) Comparison of status epilepticus models Uniformed Services University of the Health Sciences, 4301 Jones Bridge induced by pilocarpine and nerve agents - a systematic review of the underlying Road, Bethesda, MD 20814. E-mail: [email protected] aetiology and adopted therapeutic approaches. Curr Med Chem 18:886–899. Downloaded from jpet.aspetjournals.org at ASPET Journals on September 28, 2021