Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 488: 116970, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38777098

RESUMO

Soman produces excitotoxic effects by inhibiting acetylcholinesterase in the cholinergic synapses and neuromuscular junctions, resulting in soman-induced sustained status epilepticus (SSE). Our previous work showed delayed intramuscular (i.m.) treatment with A1 adenosine receptor agonist N-bicyclo-[2.2.1]-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA) alone suppressed soman-induced SSE and prevented neuropathology. Using this same rat soman seizure model, we tested if delayed therapy with ENBA (60 mg/kg, i.m.) would terminate seizure, protect neuropathology, and aid in survival when given in conjunction with current standard medical countermeasures (MCMs): atropine sulfate, 2-PAM, and midazolam (MDZ). Either 15- or 30-min following soman-induced SSE onset, male rats received atropine and 2-PAM plus either MDZ or MDZ + ENBA. Electroencephalographic (EEG) activity, physiologic parameters, and motor function were recorded. Either 2- or 14-days following exposure surviving rats were euthanized and perfused for histology. All animals treated with MDZ + ENBA at both time points had 100% EEG seizure termination and reduced total neuropathology compared to animals treated with MDZ (2-day, p = 0.015 for 15-min, p = 0.002 for 30-min; 14-day, p < 0.001 for 15-min, p = 0.006 for 30-min), showing ENBA enhanced MDZ's anticonvulsant and neuroprotectant efficacy. However, combined MDZ + ENBA treatment, when compared to MDZ treatment groups, had a reduction in the 14-day survival rate regardless of treatment time, indicating possible enhancement of MDZ's neuronal inhibitory effects by ENBA. Based on our findings, ENBA shows promise as an anticonvulsant and neuroprotectant in a combined treatment regimen following soman exposure; when given as an adjunct to standard MCMs, the dose of ENBA needs to be adjusted.


Assuntos
Agonistas do Receptor A1 de Adenosina , Ratos Sprague-Dawley , Convulsões , Soman , Animais , Soman/toxicidade , Masculino , Agonistas do Receptor A1 de Adenosina/farmacologia , Ratos , Injeções Intramusculares , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/uso terapêutico , Anticonvulsivantes/administração & dosagem , Eletroencefalografia/efeitos dos fármacos , Adenosina/análogos & derivados , Adenosina/administração & dosagem , Adenosina/farmacologia , Atropina/farmacologia , Atropina/administração & dosagem , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/tratamento farmacológico , Midazolam/farmacologia , Midazolam/uso terapêutico
2.
Neuropharmacology ; 253: 109983, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38704023

RESUMO

Exposure to organophosphorus compounds, such as soman (GD), cause widespread toxic effects, sustained status epilepticus, neuropathology, and death. The A1 adenosine receptor agonist N-bicyclo-(2.2.1)-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), when given 1 min after GD exposure, provides neuroprotection and prevents behavioral impairments. Here, we tested the ability of ENBA at delayed treatment times to improve behavioral outcomes via a two-way active avoidance task in two male animal models, each consisting of saline and GD exposure groups. In a rat model, animals received medical treatments (atropine sulfate [A], 2-PAM [P], and midazolam [MDZ]) or AP + MDZ + ENBA at 15 or 30 min after seizure onset and were subjected to behavioral testing for up to 14 days. In a human acetylcholinesterase knock-in serum carboxylesterase knock-out mouse model, animals received AP, AP + MDZ, AP + ENBA, or AP + MDZ + ENBA at 15 min post seizure onset and were subjected to the behavioral task on days 7 and 14. In rats, the GD/AP + MDZ + ENBA group recovered to saline-exposed avoidance levels while the GD/AP + MDZ group did not. In mice, in comparison with GD/AP + MDZ group, the GD/AP + MDZ + ENBA showed decreases in escape latency, response latency, and pre-session crossings, as well as increases in avoidances. In both models, only ENBA-treated groups showed control level inter-trial interval crossings by day 14. Our findings suggest that ENBA, alone and as an adjunct to medical treatments, can improve behavioral and cognitive outcomes when given at delayed time points after GD intoxication.


Assuntos
Acetilcolinesterase , Agonistas do Receptor A1 de Adenosina , Soman , Animais , Soman/toxicidade , Masculino , Agonistas do Receptor A1 de Adenosina/farmacologia , Ratos , Acetilcolinesterase/metabolismo , Humanos , Camundongos , Camundongos Knockout , Modelos Animais de Doenças , Ratos Sprague-Dawley , Memória/efeitos dos fármacos , Aprendizagem da Esquiva/efeitos dos fármacos , Adenosina/análogos & derivados , Adenosina/farmacologia
3.
Neuropharmacology ; 253: 109966, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38677446

RESUMO

Organophosphorus nerve agents, such as soman (GD), produce excitotoxic effects resulting in sustained status epilepticus (SSE) and brain damage. Previous work shows that neuronal inhibitory effects of A1 adenosine receptor (A1AR) agonists, such as N6- Bicyclo (2.2.1)-hept-2-yl-5'-chloro-5'-deoxyadenosine (Cl-ENBA), suppresses GD-induced SSE and improves neuropathology. Some other physiologic effects of these agonists are hypothermia, hypotension, and sedation. Hypothermia may also shield the brain from injury by slowing down chemical insults, lessening inflammation, and contributing to improved neurological outcomes. Therefore, we attempted to isolate the hypothermic effect from ENBA by assessing the neuroprotective efficacy of direct surface body cooling in a rat GD-induced SSE model, and comparing the effects on seizure termination, neuropathology, and survival. Male rats implanted with a body temperature (Tb) transponder and electroencephalographic (EEG) electrodes were primed with asoxime (HI-6), exposed to GD 30 min later, and then treated with Cl-ENBA or had Tb lowered directly via body cooling at 30 min after the onset of seizure activity. Afterwards, they were either allowed to develop hypothermia as expected, or received thermal support to maintain normothermic Tb for a period of 6-h. Neuropathology was assessed at 24 h. Regardless of Cl-ENBA or surface cooling, all hypothermic GD-exposed groups had significantly improved 24-h survival compared to rats with normothermic Tb (81% vs. 39%, p < 0.001). Cl-ENBA offered neuroprotection independently of hypothermic Tb. While hypothermia enhanced the overall efficacy of Cl-ENBA by improving survival outcomes, body cooling didn't reduce seizure activity or neuropathology following GD-induced SSE.


Assuntos
Agonistas do Receptor A1 de Adenosina , Hipotermia Induzida , Ratos Sprague-Dawley , Convulsões , Soman , Animais , Masculino , Agonistas do Receptor A1 de Adenosina/farmacologia , Soman/toxicidade , Hipotermia Induzida/métodos , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/prevenção & controle , Ratos , Adenosina/análogos & derivados , Adenosina/farmacologia , Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Eletroencefalografia , Modelos Animais de Doenças
4.
Toxicol Appl Pharmacol ; 464: 116437, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36849019

RESUMO

Recently a novel humanized mouse strain has been successfully generated, in which serum carboxylesterase (CES) knock out (KO) mice (Es1-/-) were further genetically modified by knocking in (KI), or adding, the gene that encodes the human form of acetylcholinesterase (AChE). The resulting human AChE KI and serum CES KO (or KIKO) mouse strain should not only exhibit organophosphorus nerve agent (NA) intoxication in a manner more similar to humans, but also display AChE-specific treatment responses more closely mimicking those of humans to facilitate data translation to pre-clinic trials. In this study, we utilized the KIKO mouse to develop a seizure model for NA medical countermeasure investigation, and then applied it to evaluate the anticonvulsant and neuroprotectant (A/N) efficacy of a specific A1 adenosine receptor (A1AR) agonist, N-bicyclo-(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), which has been shown in a rat seizure model to be a potent A/N compound. Male mice surgically implanted with cortical electroencephalographic (EEG) electrodes a week earlier were pretreated with HI-6 and challenged with various doses (26 to 47 µg/kg, SC) of soman (GD) to determine a minimum effective dose (MED) that induced sustained status epilepticus (SSE) activity in 100% of animals while causing minimum lethality at 24 h. The GD dose selected was then used to investigate the MED doses of ENBA when given either immediately following SSE initiation (similar to wartime military first aid application) or at 15 min after ongoing SSE seizure activity (applicable to civilian chemical attack emergency triage). The selected GD dose of 33 µg/kg (1.4 x LD50) generated SSE in 100% of KIKO mice and produced only 30% mortality. ENBA at a dose as little as 10 mg/kg, IP, caused isoelectric EEG activity within minutes after administration in naïve un-exposed KIKO mice. The MED doses of ENBA to terminate GD-induced SSE activity were determined to be 10 and 15 mg/kg when treatment was given at the time of SSE onset and when seizure activity was ongoing for 15 min, respectively. These doses were much lower than in the non-genetically modified rat model, which required an ENBA dose of 60 mg/kg to terminate SSE in 100% GD-exposed rats. At MED doses, all mice survived for 24 h, and no neuropathology was observed when the SSE was stopped. The findings confirmed that ENBA is a potent A/N for both immediate and delayed (i.e., dual purposed) therapy to victims of NA exposure and serves as a promising neuroprotective antidotal and adjunctive medical countermeasure candidate for pre-clinical research and development for human application.


Assuntos
Agentes Neurotóxicos , Fármacos Neuroprotetores , Soman , Estado Epiléptico , Animais , Masculino , Camundongos , Ratos , Acetilcolinesterase , Anticonvulsivantes/efeitos adversos , Agentes Neurotóxicos/toxicidade , Fármacos Neuroprotetores/efeitos adversos , Compostos Organofosforados/uso terapêutico , Agonistas do Receptor Purinérgico P1/efeitos adversos , Receptores Purinérgicos P1 , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/prevenção & controle , Soman/toxicidade , Soman/uso terapêutico , Estado Epiléptico/induzido quimicamente
5.
Toxicol Rep ; 8: 896-907, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996503

RESUMO

Animal models are essential for evaluating the toxicity of chemical warfare nerve agents (CWNAs) to extrapolate to human risk and are necessary to evaluate the efficacy of medical countermeasures. The Göttingen minipig is increasingly used for toxicological studies because it has anatomical and physiological characteristics that are similar to those of humans. Our objective was to determine whether the minipig would be a useful large animal model to evaluate the toxic effects of soman (GD). We determined the intramuscular (IM) median lethal dose (LD50) of GD in adult male Göttingen minipigs using an up-and-down dosing method. In addition to lethality estimates, we characterized the observable signs of toxicity, blood and tissue cholinesterase (ChE) activity and brain pathology following GD exposure. The 24 h LD50 of GD was estimated to be 4.7 µg/kg, with 95 % confidence limits of 3.6 and 6.3 µg/kg. As anticipated, GD inhibited ChE activity in blood and several tissues. Neurohistopathological analysis showed neurodegeneration and neuroinflammation in survivors exposed to 4.7 µg/kg of GD, including in the primary visual cortex and various thalamic nuclei. These findings suggest that the minipig will be a useful large animal model for assessing drugs to mitigate neuropathological effects of exposure to CWNAs.

6.
Toxicol Appl Pharmacol ; 419: 115515, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33798593

RESUMO

Exposure to organophosphorus nerve agents (NAs) like sarin (GB) and soman (GD) can lead to sustained seizure activity, or status epilepticus (SE). Previous research has shown that activation of A1 adenosine receptors (A1ARs) can inhibit neuronal excitability, which could aid in SE termination. Two A1AR agonists, 2-Chloro-N6-cyclopentyladenosine (CCPA) and N-Bicyclo(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), were effective in terminating GD-induced SE in rats when administered via intraperitoneal (IP) injection. However, IP injection is not a clinically relevant route of administration. This study evaluated the efficacy of these agonists in terminating NA-induced SE when administered via intramuscular (IM) route. Adult male rats were exposed subcutaneously (SC) to either GB (150 µg/kg) or GD (90 µg/kg) and were treated with ENBA or CCPA at 15, 30, or 60 min after seizure onset or left untreated. Up to 7 days after exposure, deeply anesthetized rats were euthanized and perfused brains were removed for histologic assessment of neuropathology (i.e., neuronal damage) in six brain regions (amygdala, cerebral cortex, piriform cortex, thalamus, dorsal hippocampus, and ventral hippocampus). A total neuropathy score (0-24) was determined for each rat by adding the scores from each of the six regions. The higher the total score the more severe the neuropathology. With the GB model and 60 min treatment delay, ENBA-treated rats experienced 78.6% seizure termination (N = 14) and reduced neuropathology (11.6 ± 2.6, N = 5), CCPA-treated rats experienced 85.7% seizure termination (N = 14) and slightly reduced neuropathology (20.7 ± 1.8, N = 6), and untreated rats experienced no seizure termination (N = 13) and severe neuropathology (22.3 ± 1.0, N = 4). With the GD model and 60 min treatment delay, ENBA-treated rats experienced 92.9% seizure termination (N = 14) and reduced neuropathology (13.96 ± 1.8, N = 9), CCPA-treated rats experienced 78.6% seizure termination (N = 14) and slightly reduced neuropathology (22.0 ± 0.9, N = 10); and untreated rats experienced 16.7% seizure termination (N = 12) and severe neuropathology (22.0 ± 1.8, N = 5). While ENBA and CCPA both demonstrate a clear ability to terminate SE when administered up to 60 min after seizure onset, ENBA offers more neuroprotection, making it a promising candidate for NA-induced SE.


Assuntos
Agonistas do Receptor A1 de Adenosina/administração & dosagem , Adenosina/análogos & derivados , Anticonvulsivantes/administração & dosagem , Encéfalo/efeitos dos fármacos , Desoxiadenosinas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Norbornanos/administração & dosagem , Sarina , Soman , Estado Epiléptico/prevenção & controle , Adenosina/administração & dosagem , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Esquema de Medicação , Injeções Intramusculares , Masculino , Ratos Sprague-Dawley , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/metabolismo , Estado Epiléptico/patologia , Fatores de Tempo
7.
Toxicol Appl Pharmacol ; 416: 115466, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33631229

RESUMO

Soman, an organophosphorus (OP) compound, disrupts nervous system function through inactivation of acetylcholinesterase (AChE), the enzyme that breaks down acetylcholine at synapses. Left untreated, a state of prolonged seizure activity (status epilepticus, SE) is induced, causing widespread neuronal damage and associated cognitive and behavioral impairments. Previous research demonstrated that therapeutic stimulation of A1 adenosine receptors (A1ARs) can prevent or terminate soman-induced seizure. This study examined the ability of three potent A1AR agonists to provide neuroprotection and, ultimately, prevent observable cognitive and behavioral deficits following exposure to soman. Sprague Dawley rats were challenged with a seizure-inducing dose of soman (1.2 x LD50) and treated 1 min later with one of the following A1AR agonists: (6)-Cyclopentyladenosine (CPA), 2-Chloro-N6-cyclopentyladenosine (CCPA) or N-bicyclo(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (cdENBA). An active avoidance shuttle box task was used to evaluate locomotor responses to aversive stimuli at 3, 7 and 14 days post-exposure. Animals treated with CPA, CCPA or cdENBA demonstrated a higher number of avoidance responses and a faster reaction to the aversive stimulus than the soman/saline control group across all three sessions. Findings suggest that A1AR agonism is a promising neuroprotective countermeasure, capable of preventing the long-term deficits in learning and memory that are characteristic of soman intoxication.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Anticonvulsivantes/farmacologia , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Intoxicação por Organofosfatos/prevenção & controle , Receptor A1 de Adenosina/efeitos dos fármacos , Convulsões/prevenção & controle , Soman , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Desoxiadenosinas/farmacologia , Modelos Animais de Doenças , Masculino , Intoxicação por Organofosfatos/etiologia , Intoxicação por Organofosfatos/metabolismo , Intoxicação por Organofosfatos/fisiopatologia , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo , Convulsões/patologia
8.
Toxicol Appl Pharmacol ; 415: 115443, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33548273

RESUMO

The brain is a critical target for the toxic action of organophosphorus (OP) inhibitors of acetylcholinesterase (AChE) such as the nerve agent sarin. However, the available oxime antidote 2-PAM only reactivates OP-inhibited AChE in peripheral tissues. Monoisonitrosoacetone (MINA), a tertiary oxime, reportedly reactivates AChE in the central nervous system (CNS). The current study investigated whether MINA would be beneficial as a supplemental oxime treatment in preventing lethality and reducing morbidity following lethal sarin exposure, MINA supplement would improve AChE recovery in the body, and MINA would be detectable in the CNS. Guinea pigs were exposed to sarin and treated with atropine sulfate and 2-PAM at one minute. Additional 2-PAM or MINA was administered at 3, 5, 15, or 30 min after sarin exposure. Survival and morbidity were assessed at 2 and 24 h. AChE activity in brain and peripheral tissues was evaluated one hour after MINA and 2-PAM treatment. An in vivo microdialysis technique was used to determine partitioning of MINA into the brain. A liquid chromatography-tandem mass spectrometry method was developed for the analysis of MINA in microdialysates. MINA-treated animals exhibited significantly higher survival and lower morbidity compared to 2-PAM-treated animals. 2-PAM was significantly more effective in reactivating AChE in peripheral tissues, but only MINA reactivated AChE in the CNS. MINA was found in guinea pig brain microdialysate samples beginning at ~10 min after administration in a dose-related manner. The data strongly suggest that a centrally penetrating oxime could provide significant benefit as an adjunct to atropine and 2-PAM therapy for OP intoxication.


Assuntos
Acetilcolinesterase/metabolismo , Antídotos/farmacologia , Encéfalo/efeitos dos fármacos , Reativadores da Colinesterase/farmacologia , Intoxicação por Organofosfatos/prevenção & controle , Oximas/farmacologia , Sarina , Animais , Antídotos/metabolismo , Encéfalo/enzimologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ativação Enzimática , Cobaias , Masculino , Microdiálise , Intoxicação por Organofosfatos/enzimologia , Oximas/metabolismo , Permeabilidade , Compostos de Pralidoxima/metabolismo , Compostos de Pralidoxima/farmacologia , Distribuição Tecidual
9.
Toxics ; 8(4)2020 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-33213094

RESUMO

Organophosphorus compound exposure remains a present threat through agricultural accidents, warfare, or terrorist activity. The primary mechanism of organophosphorus toxicity is through inhibition of the enzyme acetylcholinesterase, with current emergency treatment including anticholinergics, benzodiazepines, and oxime reactivators. However, a need for more effective and broadly acting countermeasures remains. This study aimed to develop larval zebrafish as a high-throughput model for evaluating novel therapeutics against acute organophosphorus exposure. Larval zebrafish at six days post-fertilization were exposed to acute concentrations of seven organophosphorus compounds and treated with one of three oximes. Lethality studies indicated similar relative toxicity to that seen in the established rodent model, with chemical warfare agents proving more lethal than organophosphorus pesticides. Additionally, the organophosphorus-specific response for oxime reactivation of acetylcholinesterase was comparable to what has been previously reported. Behavioral studies measuring the visual motor response demonstrated greater efficacy for centrally acting oxime compounds than for those that are contained to the peripheral tissue. Overall, these results support the use of this larval zebrafish model as a high-throughput screening platform for evaluating novel treatments following acute organophosphorus exposure.

10.
Neurotox Res ; 36(2): 323-333, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31069755

RESUMO

Organophosphorus nerve agents (NAs) irreversibly inhibit acetylcholinesterase, which results in the accumulation of acetylcholine and widespread excitotoxic seizure activity. Because current medical countermeasures (anticholinergics, AChE reactivators, and benzodiazepines) lack sufficient anti-seizure efficacy when treatment is delayed, those intoxicated are at risk for severe brain damage or death if treatment is not immediately available. Toward developing a more effective anti-seizure treatment for NA intoxication, this study evaluated the efficacy of A1 adenosine (ADO) receptor (A1AR) agonists in a rat soman seizure model. One minute after exposure to soman (1.6 × LD50, subcutaneous), rats were treated intraperitoneally with one of the following agonists at increasing dose levels until anti-seizure efficacy was achieved: N6-cyclopentaladenosine (CPA), 2-chloro-N6-cyclopentyladenosine (CCPA), and (±)-5'-chloro-5'-deoxy-ENBA (ENBA). All A1AR agonists were efficacious in preventing seizure and promoting survival. The effective doses for the A1AR agonists were 60 mg/kg CPA, 36 mg/kg CCPA, and 62 mg/kg ENBA. Whereas vehicle-treated rats experienced 100% seizure and 21% survival (N = 28), ADO treatments reduced seizure occurrence and improved survival rates: 8% seizure and 83% survival with CPA (60 mg/kg, N = 12), 17% seizure and 75% survival with CCPA (36 mg/kg, N = 12), and 8% seizure, 83% survival with ENBA (62 mg/kg, N = 12). The brains of ADO-treated rats were also protected from damage as indicated by neurohistopathological analysis. While all ADO agonists provided neuroprotection, rats receiving CCPA and ENBA experienced less severe ADO-induced side effects (e.g., sedation, hypothermia, bradycardia) than with CPA. The data from this study suggest that the ADO signaling pathway is a promising mechanism for countering seizure activity induced by NAs.


Assuntos
Agonistas do Receptor A1 de Adenosina/uso terapêutico , Anticonvulsivantes/uso terapêutico , Contramedidas Médicas , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Soman/toxicidade , Animais , Relação Dose-Resposta a Droga , Masculino , Agentes Neurotóxicos/toxicidade , Ratos , Ratos Sprague-Dawley , Convulsões/fisiopatologia
11.
Toxicol Mech Methods ; 29(5): 322-333, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30558458

RESUMO

The efficacy of anticonvulsant therapies to stop seizure activities following organophosphorus nerve agents (NAs) has been documented as being time-dependent. We utilized the guinea pig NA-seizure model to compare the effectiveness of phencynonate (PCH) and scopolamine (SCP) when given at the early (at time of seizure onset) or late (40 min after seizure onset) phase of seizure progression. PCH possesses both anticholinergic and anti-NMDA activities, while SCP is a purely anti-muscarinic compound. Animals with cortical electrodes were pretreated with pyridostigmine bromide 30 min prior to exposure to a 2.0 x LD50 subcutaneous dose of a NA (GA, GB, GD, GF, VR, or VX), followed one min later with atropine sulfate and 2-PAM. At either early or late phase, animals were treated with either PCH or SCP and the 24-h anticonvulsant ED50 doses were determined. When administered at seizure onset, PCH, and SCP were both effective at terminating seizure activity against all NAs, with ED50 values for SCP generally being lower. At the 40 min time, ED50 values were obtained following GA, GD, GF, and VR challenges for SCP, but ED50 value was obtained only following GD for PCH, indicating a superior efficacy of SCP. When seizure activity was controlled, a significant improvement in weight loss, neuropathology, and survival was observed, regardless of treatment or NA. Overall, these results demonstrate the differing efficacies of these two similarly structured anticholinergic compounds with delayed administration and warrant further investigation into the timing and mechanisms of the seizure maintenance phase in different animal models.


Assuntos
Anticonvulsivantes/farmacologia , Compostos Aza/farmacologia , Encéfalo/efeitos dos fármacos , Glicolatos/farmacologia , Agentes Neurotóxicos/toxicidade , Compostos Organofosforados/toxicidade , Escopolamina/farmacologia , Convulsões/prevenção & controle , Idade de Início , Animais , Anticonvulsivantes/administração & dosagem , Compostos Aza/administração & dosagem , Encéfalo/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glicolatos/administração & dosagem , Cobaias , Dose Letal Mediana , Masculino , Escopolamina/administração & dosagem , Convulsões/induzido quimicamente , Convulsões/patologia
12.
Toxicology ; 410: 10-15, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30172647

RESUMO

Genetics likely play a role in various responses to nerve agent (NA) exposure, as genetic background plays an important role in behavioral, neurological, and physiological responses. This study uses different mouse strains to identify if mouse strain differences in sarin exposure exist. In Experiment 1, basal levels of acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and carboxylesterase (CE) were measured in different strains of naïve mice to account for potential pharmacokinetic determinants of individual differences. In Experiment 2, median lethal dose (MLD) levels were estimated in 8 inbred mouse strains following subcutaneous (s.c.) administration of sarin. Few strain or sex differences in esterase activity levels were observed, with the exception of erythrocyte AChE activity in the C57BL/6J strain. Both sex and strain differences in toxicity were observed, with the most resistant strains being the BALB/cByJ and FVB/NJ strains and the most sensitive strain being the DBA/2J strain. These findings can be expanded to explore pathways involved in NA response, which may provide an avenue to develop therapeutics for preventing and treating the damaging effects of NA exposure.


Assuntos
Substâncias para a Guerra Química/toxicidade , Esterases/efeitos dos fármacos , Esterases/metabolismo , Camundongos Endogâmicos , Agentes Neurotóxicos/toxicidade , Sarina/toxicidade , Acetilcolinesterase/efeitos dos fármacos , Acetilcolinesterase/metabolismo , Animais , Butirilcolinesterase/efeitos dos fármacos , Butirilcolinesterase/metabolismo , Hidrolases de Éster Carboxílico/efeitos dos fármacos , Hidrolases de Éster Carboxílico/metabolismo , Inibidores da Colinesterase/toxicidade , Eritrócitos/efeitos dos fármacos , Eritrócitos/enzimologia , Feminino , Injeções Subcutâneas , Dose Letal Mediana , Masculino , Camundongos , Especificidade da Espécie
13.
Comput Biol Chem ; 75: 74-81, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29747078

RESUMO

Organophosphorus nerve agents (NAs) irreversibly inhibit acetylcholinesterase (AChE), the enzyme responsible for breaking down the neurotransmitter acetylcholine (ACh). The over accumulation of ACh after NA exposure leads to cholinergic toxicity, seizure, and death. Current medical countermeasures effectively mitigate peripheral symptoms, however; the brain is often unprotected. Alternative acute treatment with the adenosine A1 receptor agonist N6-cyclopentyladensosine (CPA) has previously been demonstrated to prevent AChE inhibition as well as to suppress neuronal activity. The mechanism of AChE protection is unknown. To elucidate the feasibility of potential CPA-AChE interaction mechanisms, we applied a truncated molecular model approach and density functional theory. The candidate mechanisms studied are reversible enzyme inhibition, enzyme reactivation, and NA blocking prior to enzyme conjugation. Our thermodynamic data suggest that CPA can compete with the NAs sarin and soman for the active site of AChE, but may, in contrast to NAs, undergo back-reaction. We found a strong interaction between CPA and NA conjugated AChE, making enzyme reactivation unlikely but possibly allowing for CPA protection through the prevention of NA aging. The data also indicates that there is an affinity between CPA and unbound NAs. The results from this study support the hypothesis that CPA counters NA toxicity via multiple mechanisms and is a promising therapeutic strategy that warrants further development.


Assuntos
Acetilcolinesterase/metabolismo , Agonistas do Receptor A1 de Adenosina/metabolismo , Adenosina/análogos & derivados , Agentes Neurotóxicos/metabolismo , Compostos Organofosforados/metabolismo , Sarina/metabolismo , Soman/metabolismo , Adenosina/química , Adenosina/metabolismo , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina/química , Agonistas do Receptor A1 de Adenosina/farmacologia , Animais , Estrutura Molecular , Agentes Neurotóxicos/química , Agentes Neurotóxicos/farmacologia , Compostos Organofosforados/química , Compostos Organofosforados/farmacologia , Teoria Quântica , Ratos , Sarina/química , Sarina/farmacologia , Soman/química , Soman/farmacologia , Termodinâmica
14.
Toxicol Mech Methods ; 26(5): 378-88, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27329284

RESUMO

Organophosphorus nerve agents such as soman (GD) inhibit acetylcholinesterase, producing an excess of acetylcholine (ACh), which results in respiratory distress, convulsions and status epilepticus that leads to neuropathology. Several drugs (topiramate, clobazam, pregnanolone, allopregnanolone, UBP 302, cyclopentyladenosine [CPA], ketamine, midazolam and scopolamine) have been identified as potential neuroprotectants that may terminate seizures and reduce brain damage. To systematically evaluate their efficacy, this study employed in vivo striatal microdialysis and liquid chromatography to respectively collect and analyze extracellular ACh in freely moving rats treated with these drugs 20 min after seizure onset induced by a high dose of GD. Along with microdialysis, EEG activity was recorded and neuropathology assessed at 24 h. GD induced a marked increase of ACh, which peaked at 30 min post-exposure to 800% of control levels and then steadily decreased toward baseline levels. Approximately 40 min after treatment, only midazolam (10 mg/kg) and CPA (60 mg/kg) caused a significant reduction of ACh levels, with CPA reducing ACh levels more rapidly than midazolam. Both drugs facilitated a return to baseline levels at least 55 min after treatment. At 24 h, only animals treated with CPA (67%), midazolam (18%) and scopolamine (27%) exhibited seizure termination. While all treatments except for topiramate reduced neuropathology, CPA, midazolam and scopolamine showed the greatest reduction in pathology. Our results suggest that delayed treatment with CPA, midazolam, or scopolamine is effective at reducing GD-induced seizure activity and neuropathology, with CPA and midazolam capable of facilitating a reduction in GD-induced ACh elevation.


Assuntos
Acetilcolina/metabolismo , Encéfalo , Agentes Neurotóxicos/toxicidade , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/prevenção & controle , Convulsões/prevenção & controle , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Relação Dose-Resposta a Droga , Eletroencefalografia , Masculino , Microdiálise , Fármacos Neuroprotetores/administração & dosagem , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Ratos Sprague-Dawley , Convulsões/metabolismo , Convulsões/patologia , Análise de Sobrevida , Fatores de Tempo
15.
Chem Biol Interact ; 259(Pt B): 133-141, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27062893

RESUMO

Currently fielded treatments for nerve agent intoxication include atropine, an acetylcholine receptor antagonist, and pralidoxime (2PAM), a small molecule reactivator of acetylcholinesterase (AChE). 2PAM reactivates nerve agent-inhibited AChE via direct nucleophilic attack by the oxime moiety on the phosphorus center of the bound nerve agent. Due to a permanently charged pyridinium motif, 2PAM is not thought to cross the blood brain barrier and therefore cannot act directly in the neuronal junctions of the brain. In this study, ADOC, a non-permanently charged, non-oxime molecule initially identified using pesticide-inhibited AChE, was characterized in vitro against nerve agent-inhibited recombinant human AChE. The inhibitory and reactivation potentials of ADOC were determined with native AChE and AChE inhibited with tabun, sarin, soman, cyclosarin, VX, or VR and then compared to those of 2PAM. Several structural analogs of ADOC were used to probe the reactivation mechanism of the molecule. Finally, guinea pigs were used to examine the protective efficacy of the compound after exposure to sarin. The results of both in vitro and in vivo testing will be useful in the design of future small molecule reactivators.


Assuntos
Acetilcolinesterase/metabolismo , Reativadores da Colinesterase/metabolismo , Agentes Neurotóxicos/metabolismo , Oximas/química , Fenóis/metabolismo , Compostos de Pralidoxima/metabolismo , Acetilcolinesterase/química , Acetilcolinesterase/genética , Animais , Reativadores da Colinesterase/química , Reativadores da Colinesterase/uso terapêutico , Eritrócitos/enzimologia , Cobaias , Meia-Vida , Humanos , Cinética , Masculino , Agentes Neurotóxicos/química , Agentes Neurotóxicos/intoxicação , Intoxicação por Organofosfatos/tratamento farmacológico , Organofosfatos/química , Organofosfatos/metabolismo , Fenóis/química , Fenóis/uso terapêutico , Compostos de Pralidoxima/química , Compostos de Pralidoxima/uso terapêutico , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Sarina/química , Sarina/metabolismo , Soman/química , Soman/metabolismo , Relação Estrutura-Atividade
16.
Ann N Y Acad Sci ; 1374(1): 68-77, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27123828

RESUMO

The current research progression efforts for investigating novel treatments for exposure to organophosphorus (OP) compounds that inhibit acetylcholinesterase (AChE), including pesticides and chemical warfare nerve agents (CWNAs), rely solely on in vitro cell assays and in vivo rodent models. The zebrafish (Danio rerio) is a popular, well-established vertebrate model in biomedical research that offers high-throughput capabilities and genetic manipulation not readily available with rodents. A number of research studies have investigated the effects of subacute developmental exposure to OP pesticides in zebrafish, observing detrimental effects on gross morphology, neuronal development, and behavior. Few studies, however, have utilized this model to evaluate treatments, such as oxime reactivators, anticholinergics, or anticonvulsants, following acute exposure. Preliminary work has investigated the effects of CWNA exposure. The results clearly demonstrated relative toxicity and oxime efficacy similar to that reported for the rodent model. This review surveys the current literature utilizing zebrafish as a model for OP exposure and highlights its potential use as a high-throughput system for evaluating AChE reactivator antidotal treatments to acute pesticide and CWNA exposure.


Assuntos
Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/toxicidade , Compostos Organofosforados/toxicidade , Oximas/farmacologia , Peixe-Zebra/fisiologia , Animais , Ativação Enzimática/efeitos dos fármacos , Modelos Animais
17.
Toxicol Mech Methods ; 24(6): 385-95, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24785252

RESUMO

The current regimen for treating nerve agent poisoning does not sufficiently suppress the excitotoxic activity that causes severe brain damage, especially in cases where treatment is delayed and nerve agent-induced status epilepticus develops. New therapeutic targets are required to improve survivability and minimize neuropathology after irreversible acetylcholinesterase inactivation. Earlier studies have shown that systemic delivery of adenosine agonists decreases nerve agent lethality; however, the mechanism of protection remains to be understood. The primary aim of this study was to investigate the role of central adenosine receptor (AR) stimulation in neuroprotection by directly injecting (6)-cyclopentyladenosine (CPA), an adenosine agonist specific to the A1 receptor subtype (A1R), into the brain intracerebroventricularly (ICV) in a soman seizure rat model. In addition to general A1R stimulation, we hypothesized that bilateral micro-injection of CPA into the cholinergic basal forebrain (BF) could also suppress excitotoxic activity. The results from these studies demonstrated that centrally administered adenosine agonists are anti-seizure and neuroprotective. CPA-delivered ICV prevented seizure and convulsion in 100% of the animals. Moreover, neuropathological evaluation indicated that adenosine treatments reduced brain damage from severe to minimal. Inhibition of the BF via CPA had varied results. Some animals were protected by treatment; however, others displayed similar pathology to the control. Overall, these data suggest that stimulating central ARs could be an effective target for the next generation countermeasures for nerve agent intoxication.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Adenosina/análogos & derivados , Anticonvulsivantes/farmacologia , Receptor A1 de Adenosina/metabolismo , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Soman/toxicidade , Adenosina/farmacologia , Animais , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Substâncias para a Guerra Química/toxicidade , Ratos
18.
Neurotoxicology ; 33(3): 463-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22774227

RESUMO

Pharmacological control of seizure activity following nerve agent exposure is critical in reducing neuropathology and improving survival in casualties. Three classes of drugs, anticholinergics, benzodiazepines and excitatory amino acid (EAA) antagonists, have been shown to be effective at moderating nerve agent-induced seizures. However, little is known about which brain structures are involved in producing the anticonvulsant response. This study evaluated drugs from each class, injected directly into one of three specific brain structures, the perirhinal cortex, the entorhinal cortex, or the mediodorsal thalamus, for their ability to modulate seizures induced by the nerve agent soman. The drugs evaluated were the anticholinergic scopolamine, the benzodiazepine midazolam, and the EAA antagonist MK-801. For each drug treatment in each brain area, anticonvulsant ED50 values were calculated using an up-down dosing procedure over successive animals. There was no statistical difference in the anticonvulsant ED50 values for scopolamine and MK-801 in the perirhinal and entorhinal cortices. MK-801 pretreatment in the mediodorsal thalamus had a significantly lower anticonvulsant ED50 value than any other treatment/injection site combination. Midazolam required significantly higher doses than scopolamine and MK-801 in the perirhinal and entorhinal cortices to produce an anticonvulsant response and was ineffective in the mediodorsal thalamus. These findings support the contention that specific neuroanatomical pathways are activated during nerve agentinduced seizures and that the discrete brain structures involved have unique pharmacological thresholds for producing an anticonvulsant response. This study is also the first to show the involvement of the mediodorsal thalamus in the control of nerve agent-induced seizures.


Assuntos
Anticonvulsivantes/farmacologia , Encéfalo/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Convulsões/induzido quimicamente , Convulsões/prevenção & controle , Soman/toxicidade , Animais , Encéfalo/fisiopatologia , Mapeamento Encefálico/métodos , Antagonistas Colinérgicos/farmacologia , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Eletroencefalografia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Masculino , Midazolam/farmacologia , Ratos , Ratos Sprague-Dawley , Escopolamina/farmacologia , Convulsões/fisiopatologia , Soman/administração & dosagem
19.
Arch Toxicol ; 85(12): 1607-16, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21695469

RESUMO

Organophosphorus nerve agents such as sarin (GB) and VX irreversibly inhibit acetylcholinesterase, causing a buildup of acetylcholine (ACh) in synapses and neuromuscular junctions, which leads to excess bronchial secretions, convulsions, seizures, coma, and death. Understanding the unique toxic characteristics of different nerve agents is vital in the effort to develop broad spectrum medical countermeasures. To this end, we employed a repeated measure multivariate design with striatal microdialysis collection and high-performance liquid chromatography analysis to measure changes in concentrations of several neurotransmitters (ACh, glutamate, aspartate, GABA) in the same samples during acute exposure to GB or VX in freely moving guinea pigs. Concurrent with microdialysis collection, we used cortical electrodes to monitor brain seizure activity. This robust double multivariate design provides greater fidelity when comparing data while also reducing the required number of subjects. No correlation between nerve agents' propensity for causing seizure and seizure-related lethality was observed. The GB seizure group experienced more rapid and severe cholinergic toxicity and lethality than that of the VX seizure group. Seizures generated from GB and VX exposure resulted in further elevation of ACh level and then a gradual return to baseline. Glutamate levels increased in the GB, but not in the VX, seizure group. There were no consistent changes in either aspartate or GABA as a result of either nerve agent. These observations reinforce findings with other nerve agents that seizure activity per se contributes to the elevated levels of brain ACh observed after nerve agent exposure.


Assuntos
Substâncias para a Guerra Química/toxicidade , Compostos Organotiofosforados/toxicidade , Sarina/toxicidade , Convulsões/induzido quimicamente , Acetilcolina/metabolismo , Animais , Encéfalo/metabolismo , Inibidores da Colinesterase/toxicidade , Cromatografia Líquida de Alta Pressão , Eletroencefalografia , Ácido Glutâmico/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Cobaias , Masculino , Microdiálise , Análise Multivariada , Convulsões/fisiopatologia , Índice de Gravidade de Doença
20.
Toxicol Mech Methods ; 21(1): 53-62, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21117832

RESUMO

This study examined whether pro-2-PAM, a pro-drug dihydropyridine derivative of the oxime 2-pralidoxime (2-PAM) that can penetrate the brain, could prevent or reverse the central toxic effects of three nerve agents; sarin, cyclosarin, and VX. The first experiment tested whether pro-2-PAM could reactivate guinea pig cholinesterase (ChE) in vivo in central and peripheral tissues inhibited by these nerve agents. Pro-2-PAM produced a dose-dependent reactivation of sarin- or VX-inhibited ChE in both peripheral and brain tissues, but with substantially greater reactivation in peripheral tissues compared to brain. Pro-2-PAM produced 9-25% reactivation of cyclosarin-inhibited ChE in blood, heart, and spinal cord, but no reactivation in brain or muscle tissues. In a second experiment, the ability of pro-2-PAM to block or terminate nerve agent-induced electroencephalographic seizure activity was evaluated. Pro-2-PAM was able to block sarin- or VX-induced seizures (16-33%) over a dose range of 24-32 mg/kg, but was ineffective against cyclosarin-induced seizures. Animals that were protected from seizures showed significantly less weight loss and greater behavioral function 24 h after exposure than those animals that were not protected. Additionally, brains were free from neuropathology when pro-2-PAM prevented seizures. In summary, pro-2-PAM provided modest reactivation of sarin- and VX-inhibited ChE in the brain and periphery, which was reflected by a limited ability to block or terminate seizures elicited by these agents. Pro-2-PAM was able to reactivate blood, heart, and spinal cord ChE inhibited by cyclosarin, but was not effective against cyclosarin-induced seizures.


Assuntos
Reativadores da Colinesterase/farmacologia , Compostos Organofosforados/toxicidade , Compostos Organotiofosforados/toxicidade , Compostos de Pralidoxima/farmacologia , Sarina/toxicidade , Animais , Cobaias , Masculino , Pró-Fármacos , Convulsões/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA