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










Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 388(2): 432-450, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-37739807

RESUMEN

Acute exposure to nerve agents induces a peripheral cholinergic crisis and prolonged status epilepticus (SE), causing death or long-term brain damage. To provide preclinical data pertinent to the protection of infants and newborns, we compared the antiseizure and neuroprotective effects of treating soman-induced SE with midazolam (MDZ) versus tezampanel (LY293558) in combination with caramiphen (CRM) in 12- and 7-day-old rats. The anticonvulsants were administered 1 hour after soman exposure; neuropathology data were collected up to 6 months postexposure. In both ages, the total duration of SE within 24 hours after soman exposure was significantly shorter in the LY293558 plus CRM groups compared with the MDZ groups. Neuronal degeneration was substantial in the MDZ-treated groups but absent or minimal in the groups treated with LY293558 plus CRM. Loss of neurons and interneurons in the basolateral amygdala and CA1 hippocampal area was significant in the MDZ-treated groups but virtually absent in the LY293558 plus CRM groups. Atrophy of the amygdala and hippocampus occurred only in MDZ-treated groups. Neuronal/interneuronal loss and atrophy of the amygdala and hippocampus deteriorated over time. Reduction of inhibitory activity in the basolateral amygdala and increased anxiety were found only in MDZ groups. Spontaneous recurrent seizures developed in the MDZ groups, deteriorating over time; a small percentage of rats from the LY293558 plus CRM groups also developed seizures. These results suggest that brain damage can be long lasting or permanent if nerve agent-induced SE in infant victims is treated with midazolam at a delayed timepoint after SE onset, whereas antiglutamatergic treatment with tezampanel and caramiphen provides significant neuroprotection. SIGNIFICANCE STATEMENT: To protect the brain and the lives of infants in a mass exposure to nerve agents, an anticonvulsant treatment must be administered that will effectively stop seizures and prevent neuropathology, even if offered with a relative delay after seizure onset. The present study shows that midazolam, which was recently approved by the Food and Drug Administration for the treatment of nerve agent-induced status epilepticus, is not an effective neuroprotectant, whereas brain damage can be prevented by targeting glutamate receptors.


Asunto(s)
Lesiones Encefálicas , Ciclopentanos , Isoquinolinas , Agentes Nerviosos , Fármacos Neuroprotectores , Soman , Estado Epiléptico , Tetrazoles , Humanos , Recién Nacido , Ratas , Animales , Agentes Nerviosos/toxicidad , Midazolam/farmacología , Midazolam/uso terapéutico , Soman/toxicidad , Neuroprotección , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Anticonvulsivantes/efectos adversos , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/tratamiento farmacológico , Encéfalo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Atrofia/tratamiento farmacológico
2.
Toxics ; 11(10)2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37888716

RESUMEN

Organophosphorus compounds (OPs) have applications in agriculture (e.g., pesticides), industry (e.g., flame retardants), and chemical warfare (nerve agents). In high doses or chronic exposure, they can be toxic or lethal. The primary mechanism, common among all OPs, that initiates their toxic effects is the inhibition of acetylcholinesterase. In acute OP exposure, the subsequent surge of acetylcholine in cholinergic synapses causes a peripheral cholinergic crisis and status epilepticus (SE), either of which can lead to death. If death is averted without effective seizure control, long-term brain damage ensues. This review describes the mechanisms by which elevated acetylcholine can cause respiratory failure and trigger SE; the role of the amygdala in seizure initiation; the role of M1 muscarinic receptors in the early stages of SE; the neurotoxic pathways activated by SE (excitotoxicity/Ca++ overload/oxidative stress, neuroinflammation); and neurotoxic mechanisms linked to low-dose, chronic exposure (Ca++ dyshomeostasis/oxidative stress, inflammation), which do not depend on SE and do not necessarily involve acetylcholinesterase inhibition. The evidence so far indicates that brain damage from acute OP exposure is a direct result of SE, while the neurotoxic mechanisms activated by low-dose chronic exposure are independent of SE and may not be associated with acetylcholinesterase inhibition.

3.
Exp Biol Med (Maywood) ; 248(7): 612-623, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37300407

RESUMEN

Prolonged status epilepticus (SE) can cause brain damage; therefore, treatment must be administered promptly after seizure onset to limit SE duration and prevent neuropathology. Timely treatment of SE is not always feasible; this would be particularly true in a mass exposure to an SE-inducing agent such as a nerve agent. Therefore, the availability of anticonvulsant treatments that have neuroprotective efficacy even if administered with a delay after SE onset is an imperative. Here, we compared the long-term neuropathology resulting from acutely exposing 21-day-old male and female rats to the nerve agent soman, and treating them with midazolam (3 mg/kg) or co-administration of tezampanel (10 mg/kg) and caramiphen (50 mg/kg), at 1 h postexposure (~50 min after SE onset). Midazolam-treated rats had significant neuronal degeneration in limbic structures, mainly at one month postexposure, followed by neuronal loss in the basolateral amygdala and the CA1 hippocampal area. Neuronal loss resulted in significant amygdala and hippocampal atrophy, deteriorating from one to six months postexposure. Rats treated with tezampanel-caramiphen had no evidence of neuropathology, except for neuronal loss in the basolateral amygdala at the six-month timepoint. Anxiety was increased only in the midazolam-treated rats, at one, three, and six months postexposure. Spontaneous recurrent seizures appeared only in midazolam-treated rats, at three and six months postexposure in males and only at six months in females. These findings suggest that delayed treatment of nerve agent-induced SE with midazolam may result in long-lasting or permanent brain damage, while antiglutamatergic anticonvulsant treatment consisting of tezampanel and caramiphen may provide full neuroprotection.


Asunto(s)
Lesiones Encefálicas , Agentes Nerviosos , Soman , Estado Epiléptico , Femenino , Ratas , Masculino , Animales , Soman/toxicidad , Soman/uso terapéutico , Midazolam/farmacología , Midazolam/uso terapéutico , Anticonvulsivantes/efectos adversos , Agentes Nerviosos/efectos adversos , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/patología , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/patología , Lesiones Encefálicas/tratamiento farmacológico , Encéfalo/patología
4.
Exp Biol Med (Maywood) ; 248(7): 596-604, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37208920

RESUMEN

Hyperexcitability is a major mechanism implicated in several neuropsychiatric disorders, such as organophosphate-induced status epilepticus (SE), primary epilepsy, stroke, spinal cord injury, traumatic brain injury, schizophrenia, and autism spectrum disorders. Underlying mechanisms are diverse, but a functional impairment and loss of GABAergic inhibitory neurons are common features in many of these disorders. While novel therapies abound to correct for the loss of GABAergic inhibitory neurons, it has been difficult at best to improve the activities of daily living for the majority of patients. Alpha-linolenic acid (ALA) is an essential omega-3 polyunsaturated fatty acid found in plants. ALA exerts pleiotropic effects in the brain that attenuate injury in chronic and acute brain disease models. However, the effect of ALA on GABAergic neurotransmission in hyperexcitable brain regions involved in neuropsychiatric disorders, such as the basolateral amygdala (BLA) and CA1 subfield of the hippocampus, is unknown. Administration of a single dose of ALA (1500 nmol/kg) subcutaneously increased the charge transfer of inhibitory postsynaptic potential currents mediated by GABAA receptors in pyramidal neurons by 52% in the BLA and by 92% in the CA1 compared to vehicle animals a day later. Similar results were obtained in pyramidal neurons from the BLA and CA1 when ALA was bath-applied in slices from naïve animals. Importantly, pretreatment with the high-affinity, selective TrkB inhibitor, k252, completely abolished the ALA-induced increase in GABAergic neurotransmission in the BLA and CA1, suggesting a brain-derived neurotrophic factor (BDNF)-mediated mechanism. Addition of mature BDNF (20 ng/mL) significantly increased GABAA receptor inhibitory activity in the BLA and CA1 pyramidal neurons similar to the results obtained with ALA. ALA may be an effective treatment for neuropsychiatric disorders where hyperexcitability is a major feature.


Asunto(s)
Complejo Nuclear Basolateral , Ratas , Humanos , Animales , Complejo Nuclear Basolateral/metabolismo , Ácido alfa-Linolénico/farmacología , Factor Neurotrófico Derivado del Encéfalo , Ratas Sprague-Dawley , Actividades Cotidianas , Transmisión Sináptica/fisiología , Receptores de GABA-A/metabolismo
5.
Toxics ; 10(8)2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35893842

RESUMEN

Acute exposure to nerve agents induces status epilepticus (SE), which can cause death or long-term brain damage. Diazepam is approved by the FDA for the treatment of nerve agent-induced SE, and midazolam (MDZ) is currently under consideration to replace diazepam. However, animal studies have raised questions about the neuroprotective efficacy of benzodiazepines. Here, we compared the antiseizure and neuroprotective efficacy of MDZ (5 mg/kg) with that of tezampanel (LY293558; 10 mg/kg), an AMPA/GluK1 receptor antagonist, administered 1 h after injection of the nerve agent, soman (1.2 × LD50), in adult male rats. Both of the anticonvulsants promptly stopped SE, with MDZ having a more rapid effect. However, SE reoccurred to a greater extent in the MDZ-treated group, resulting in a significantly longer total duration of SE within 24 h post-exposure compared with the LY293558-treated group. The neuroprotective efficacy of the two drugs was studied in the basolateral amygdala, 30 days post-exposure. Significant neuronal and inter-neuronal loss, reduced ratio of interneurons to the total number of neurons, and reduction in spontaneous inhibitory postsynaptic currents accompanied by increased anxiety were found in the MDZ-treated group. The rats treated with LY293558 did not differ from the control rats (not exposed to soman) in any of these measurements. Thus, LY293558 has significantly greater efficacy than midazolam in protecting against prolonged seizures and brain damage caused by acute nerve agent exposure.

6.
Amino Acids ; 54(9): 1229-1249, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35798984

RESUMEN

Traumatic brain injury (TBI) has reached epidemic proportions around the world and is a major public health concern in the United States. Approximately 2.8 million individuals sustain a traumatic brain injury and are treated in an Emergency Department yearly in the U.S., and about 50,000 of them die. Persistent symptoms develop in 10-15% of the cases including neuropsychiatric disorders. Anxiety is the second most common neuropsychiatric disorder that develops in those with persistent neuropsychiatric symptoms after TBI. Abnormalities or atrophy in the temporal lobe has been shown in the overwhelming number of TBI cases. The basolateral amygdala (BLA), a temporal lobe structure that consolidates, stores and generates fear and anxiety-based behavioral outputs, is a critical brain region in the anxiety circuitry. In this review, we sought to capture studies that characterized the relationship between human post-traumatic anxiety and structural/functional alterations in the amygdala. We compared the human findings with results obtained with a reproducible mild TBI animal model that demonstrated a direct relationship between the alterations in the BLA and an anxiety-like phenotype. From this analysis, both preliminary insights, and gaps in knowledge, have emerged which may open new directions for the development of rational and more efficacious treatments.


Asunto(s)
Complejo Nuclear Basolateral , Lesiones Traumáticas del Encéfalo , Animales , Ansiedad , Encéfalo , Humanos
7.
Brain Res ; 1770: 147628, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34454948

RESUMEN

The amygdala is central to emotional behavior, and the excitability level of the basolateral nucleus of the amygdala (BLA) is associated with the level of anxiety. The excitability of neuronal networks is significantly controlled by GABAergic inhibition. Here, we investigated whether GABAergic inhibition in the BLA is altered during the rat estrous cycle. In rat amygdala slices, most principal BLA neurons display spontaneous IPSCs (sIPSCs) in the form of "bursts" of inhibitory currents, occurring rhythmically at a frequency of about 0.5 Hz. The percentage of BLA neurons displaying sIPSC bursts, along with the inhibitory charge transferred by sIPSCs and the frequency of sIPSC bursts, were significantly increased during the estrus phase; increased inhibition was accompanied by reduced anxiety in the open field, the light-dark box, and the acoustic startle response tests. sIPSC bursts were blocked by ibuprofen, an antagonist of acid-sensing-1a channels (ASIC1a), whose activity is known to increase by decreasing temperature. A transient reduction in the temperature of the slice medium, strengthened the sIPSCs bursts; this effect was blocked in the presence of ibuprofen. Further analysis of the sIPSC bursts during estrus showed significantly stronger rhythmic inhibitory activity in early estrus, when body temperature drops, compared with late estrus. To the extent that these results may relate to humans, it is suggested that "a calmer amygdala" due to increased inhibitory activity may underlie the positive affect in women around ovulation time. ASIC1a may contribute to increased inhibition, with their activity facilitated by the body-temperature drop preceding ovulation.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Ansiedad/metabolismo , Complejo Nuclear Basolateral/metabolismo , Estro/fisiología , Inhibición Neural/fisiología , Canales Iónicos Sensibles al Ácido/genética , Animales , Ansiedad/fisiopatología , Complejo Nuclear Basolateral/fisiopatología , Conducta Exploratoria/fisiología , Femenino , Potenciales Postsinápticos Inhibidores/fisiología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
8.
Neuropharmacology ; 181: 108298, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32898558

RESUMEN

Nerve agents are organophosphorus acetylcholinesterase inhibitors. Acute exposure to nerve agents can cause rapid death. In this review, we summarize the history of nerve agent development and use in warfare, the mechanisms by which these agents cause death or long-term brain damage, and the treatments for preventing death or long-term morbidity. The G-series nerve agents, tabun, sarin, soman, ethyl sarin, and cyclosarin, were developed by the Nazis. VX, the best-known of the V-series agents, was synthesized in the 1950's by a British scientist. Little is known about the development of the novichoks (the "A-series") by the former Soviet Union. Nerve agents were used for the first time in the battlefield by the Iraqi government in the Iran-Iraq War, in the 1980s. The Chemical Weapons Convention, in 1993, banned all chemical weapons production and use, yet, sarin was subsequently used in terrorist attacks in Japan and, recently, in the war in Syria. Pyridostigmine has been used as a prophylactic treatment, and bioscavengers are presently investigated as a better alternative. Atropine, along with an oxime, can prevent rapid death from the nerve agent-induced peripheral cholinergic crisis. Treatment with diazepam or midazolam for the cessation of nerve agent-induced status epilepticus cannot protect against brain damage, and, therefore, these benzodiazepines should be replaced by novel anticonvulsants and neuroprotectants. The AMPA/GluK1 receptor antagonist LY293558 (tezampanel) has shown superior antiseizure and neuroprotective efficacy against soman, particularly when administered in combination with caramiphen, an antagonist of muscarinic and NMDA receptors. This article is part of the special issue entitled 'Acetylcholinesterase Inhibitors: From Bench to Bedside to Battlefield'.


Asunto(s)
Sustancias para la Guerra Química/toxicidad , Inhibidores de la Colinesterasa/toxicidad , Reactivadores de la Colinesterasa/uso terapéutico , Agentes Nerviosos/toxicidad , Armas de Destrucción Masiva , Animales , Inhibidores de la Colinesterasa/historia , Historia del Siglo XX , Humanos , Contramedidas Médicas , Agentes Nerviosos/historia
9.
Ann N Y Acad Sci ; 1479(1): 122-133, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32237259

RESUMEN

Acute nerve agent exposure induces status epilepticus (SE), which can cause brain damage or death. Research aiming at developing effective therapies for controlling nerve agent-induced SE is commonly performed in adult rats. The characteristics of nerve agent-induced SE in young rats are less clear; relevant knowledge is necessary for developing effective pediatric therapies. Here, we have used electroencephalographic (EEG) recordings and analysis to study seizures in postnatal day 21 rats exposed to 1.2 × LD50 of soman, and compared the antiseizure efficacy of midazolam (MDZ)-currently considered by the Food and Drug Administration to replace diazepam for treating SE in victims of nerve agent exposure-with that of LY293558, an AMPA/GluK1 receptor antagonist, administered in combination with caramiphen, an antimuscarinic with N-methyl-d-aspartate receptor antagonistic properties. Prolonged SE developed in 80% of the rats and was reflected in behavioral seizures/convulsions. Both MDZ and LY293558 + caramiphen stopped the SE induced by soman, but there was a significant recurrence of seizures within 24 h postexposure only in the MDZ-treated group, as revealed in the raw EEG data and their representation in the frequency domain using a fast Fourier transform and in spectral analysis over 24 hours. In contrast to the high efficacy of LY293558 + caramiphen, MDZ is not an effective treatment for SE induced by soman in young animals.


Asunto(s)
Antídotos/farmacología , Ciclopentanos/farmacología , Electrocardiografía , Isoquinolinas/farmacología , Midazolam/farmacología , Agentes Nerviosos/toxicidad , Soman/toxicidad , Estado Epiléptico , Tetrazoles/farmacología , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/fisiopatología , Estado Epiléptico/prevención & control
10.
Mol Neurobiol ; 57(1): 23-31, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31520315

RESUMEN

Traumatic brain injury (TBI) is a major public health concern in the USA. There are approximately 2.5 million brain injuries annually, 90% of which may be classified as mild since these individuals do not display clear morphological abnormalities following injury on imaging. The majority of individuals develop neurocognitive deficits such as learning and memory impairment and recovery occurs over 3 to 6 months after mild TBI (mTBI). The hippocampus is highly susceptible to injury from mTBI due to the anatomic localization and has been implicated in the neurocognitive impairments after mTBI. Here, we investigated whether the mTBI-induced morphological and pathophysiological alterations of GABAergic interneurons in the CA1 subfield of the hippocampus recovers after 30 days in the controlled cortical impact (CCI) model of TBI. Design-based stereology shows a significant reduction in the number of GABAergic interneurons 7 days after CCI. However, the number of GABAergic interneurons is not significantly reduced at 30 days after CCI. The total number of neurons is not altered over the course of 30 days. GABAergic inhibitory currents in the CA1 subfield also show that, although there is a significant reduction in the CCI group at 7 days, the currents are not significantly different from sham controls at 30 days. We suggest that the recovery of GABAergic function in the CA1 subfield of the hippocampus observed 30 days after CCI is one of the mechanisms associated with the recovery of memory after mTBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/fisiopatología , Región CA1 Hipocampal/fisiopatología , Hipocampo/fisiopatología , Potenciales Postsinápticos Inhibidores/fisiología , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/fisiopatología , Lesiones Traumáticas del Encéfalo/complicaciones , Modelos Animales de Enfermedad , Neuronas GABAérgicas , Interneuronas/metabolismo , Masculino , Memoria/fisiología , Trastornos de la Memoria/complicaciones , Ratas Sprague-Dawley
11.
Neurobiol Dis ; 133: 104406, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30798006

RESUMEN

One of the devastating effects of acute exposure to organophosphates, like nerve agents, is the induction of severe and prolonged status epilepticus (SE), which can cause death, or brain damage if death is prevented. Seizures after exposure are initiated by muscarinic receptor hyperstimulation-after inhibition of acetylcholinesterase by the organophosphorus agent and subsequent elevation of acetylcholine-but they are reinforced and sustained by glutamatergic hyperexcitation, which is the primary cause of brain damage. Diazepam is the FDA-approved anticonvulsant for the treatment of nerve agent-induced SE, and its replacement by midazolam is currently under consideration. However, clinical data derived from the treatment of SE of any etiology, as well as studies on the control of nerve agent-induced SE in animal models, have indicated that diazepam and midazolam control seizures only temporarily, their antiseizure efficacy is reduced as the latency of treatment from the onset of SE increases, and their neuroprotective efficacy is limited or absent. Here, we review data on the discovery of a novel anticonvulsant and neuroprotectant, LY293558, an AMPA/GluK1 receptor antagonist. Treatment of soman-exposed immature, young-adult, and aged rats with LY293558, terminates SE with limited recurrence of seizures, significantly protects from brain damage, and prevents long-term behavioral deficits, even when LY293558 is administered 1 h post-exposure. More beneficial effects and complete neuroprotection is obtained when LY293558 administration is combined with caramiphen, which antagonizes NMDA receptors. Further efficacy studies may bring the LY293558 + caramiphen combination therapy on the pathway to approval for human use.


Asunto(s)
Anticonvulsivantes/farmacología , Isoquinolinas/farmacología , Fármacos Neuroprotectores/farmacología , Intoxicación por Organofosfatos , Receptores AMPA/antagonistas & inhibidores , Receptores de Ácido Kaínico/antagonistas & inhibidores , Tetrazoles/farmacología , Animales , Inhibidores de la Colinesterasa/toxicidad , Humanos , Agentes Nerviosos/toxicidad , Intoxicación por Organofosfatos/tratamiento farmacológico , Ratas , Soman/toxicidad , Estado Epiléptico/inducido químicamente
12.
Epilepsia ; 59 Suppl 2: 92-99, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30159887

RESUMEN

Nerve agents are organophosphate (OP) compounds and among the most powerful poisons known to man. A terrorist attack on civilian or military populations causing mass casualties is a real threat. The OP nerve agents include soman, sarin, cyclosarin, tabun, and VX. The major mechanism of acute toxicity is the irreversible inhibition of acetylcholinesterase. Acetylcholinesterase inhibition results in the accumulation of excessive acetylcholine levels in synapses, leading to progression of toxic signs including hypersecretions, tremors, status epilepticus, respiratory distress, and death. Miosis and rhinorrhea are the most common clinical findings in those individuals acutely exposed to OP nerve agents. Prolonged seizures are responsible for the neuropathology. The brain region that shows the most severe damage is the amygdala, followed by the piriform cortex, hippocampus, cortex, thalamus, and caudate/putamen. Current medical countermeasures are only modestly effective in attenuating the seizures and neuropathology. Anticonvulsants such as benzodiazepines decrease seizure activity and improve outcome, but their efficacy depends upon the administration time after exposure to the nerve agent. Administration of benzodiazepines may increase the risk for seizure recurrence. Recent studies document long-term neurologic and behavior deficits, and technological advances demonstrate structural brain changes on magnetic resonance imaging.


Asunto(s)
Encéfalo/efectos de los fármacos , Agentes Nerviosos/toxicidad , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Encéfalo/metabolismo , Humanos , Miosis/tratamiento farmacológico , Miosis/etiología , Trastornos Respiratorios/inducido químicamente , Trastornos Respiratorios/tratamiento farmacológico , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Factores de Tiempo
13.
Neurotox Res ; 34(3): 511-524, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29713995

RESUMEN

Acute exposure to nerve agents induces status epilepticus (SE), which causes brain damage or death. LY293558, an antagonist of AMPA and GluK1 kainate receptors is a very effective anticonvulsant and neuroprotectant against soman; however, some neuronal damage is still present after treatment of soman-exposed rats with LY293558. Here, we have tested whether combining LY293558 with an NMDA receptor antagonist can eliminate the residual damage. For this purpose, we chose caramiphen (CRM), an antimuscarinic compound with NMDA receptor antagonistic properties. Adult male rats were exposed to 1.2 × LD50 soman, and at 20 min after soman exposure, were injected with atropine + HI-6, or atropine + HI-6 + LY293558 (15 mg/kg), or atropine + HI-6 + LY293558 + CRM (50 mg/kg). We found that (1) the LY293558 + CRM treatment terminated SE significantly faster than LY293558 alone; (2) after cessation of the initial SE, seizures did not return in the LY293558 + CRM-treated group, during 72 h of monitoring; (3) power spectrum analysis of continuous EEG recordings for 7 days post-exposure showed increased delta and decreased gamma power that lasted beyond 24 h post-exposure only in the rats who did not receive anticonvulsant treatment; (4) spontaneous recurrent seizures appeared on day 7 only in the group that did not receive anticonvulsant treatment; (5) significant neuroprotection was achieved by LY293558 administration, while the rats who received LY293558 + CRM displayed no neurodegeneration; (6) body weight loss and recovery in the LY293558 + CRM-treated rats did not differ from those in control rats who were not exposed to soman. The data show that treatment with LY293558 + CRM provides full antiseizure and neuroprotective efficacy against soman.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Ciclopentanos/uso terapéutico , Isoquinolinas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Convulsiones/tratamiento farmacológico , Tetrazoles/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Lesiones Encefálicas/inducido químicamente , Ondas Encefálicas/efectos de los fármacos , Inhibidores de la Colinesterasa/toxicidad , Modelos Animales de Enfermedad , Quimioterapia Combinada/métodos , Electroencefalografía , Fluoresceínas/metabolismo , Análisis de Fourier , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Soman/toxicidad , Factores de Tiempo
14.
J Pharmacol Exp Ther ; 365(2): 314-326, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29467308

RESUMEN

The currently Food and Drug Administration-approved anticonvulsant for the treatment of status epilepticus (SE) induced by nerve agents is the benzodiazepine diazepam; however, diazepam does not appear to offer neuroprotective benefits. This is of particular concern with respect to the protection of children because, in the developing brain, synaptic transmission mediated via GABAA receptors, the target of diazepam, is weak. In the present study, we exposed 21-day-old male rats to 1.2 × LD50 soman and compared the antiseizure, antilethality, and neuroprotective efficacy of diazepam (10 mg/kg), LY293558 (an AMPA/GluK1 receptor antagonist; 15 mg/kg), caramiphen (CRM, an antimuscarinic with NMDA receptor-antagonistic properties; 50 mg/kg), and LY293558 (15 mg/kg) + CRM (50 mg/kg), administered 1 hour after exposure. Diazepam, LY293558, and LY293558 + CRM, but not CRM alone, terminated SE; LY293558 + CRM treatment acted significantly faster and produced a survival rate greater than 85%. Thirty days after soman exposure, neurodegeneration in limbic regions was most severe in the CRM-treated group, minimal to severe-depending on the region-in the diazepam group, absent to moderate in the LY293558-treated group, and totally absent in the LY293558 + CRM group. Amygdala and hippocampal atrophy, a severe reduction in spontaneous inhibitory activity in the basolateral amygdala, and increased anxiety-like behavior in the open-field and acoustic startle response tests were present in the diazepam and CRM groups, whereas the LY293558 and LY293558 + CRM groups did not differ from controls. The combined administration of LY293558 and CRM, by blocking mainly AMPA, GluK1, and NMDA receptors, is a very effective anticonvulsant and neuroprotective therapy against soman in young rats.


Asunto(s)
Anticonvulsivantes/farmacología , Ciclopentanos/farmacología , Diazepam/farmacología , Isoquinolinas/farmacología , Fármacos Neuroprotectores/farmacología , Soman/farmacología , Estado Epiléptico/tratamiento farmacológico , Tetrazoles/farmacología , Animales , Anticonvulsivantes/uso terapéutico , Ansiedad/complicaciones , Ansiedad/prevención & control , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Complejo Nuclear Basolateral/patología , Conducta Animal/efectos de los fármacos , Niño , Ciclopentanos/uso terapéutico , Diazepam/uso terapéutico , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Humanos , Isoquinolinas/uso terapéutico , Masculino , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/complicaciones , Tetrazoles/uso terapéutico
15.
Neuroscience ; 373: 145-158, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29339324

RESUMEN

Synchronous, rhythmic firing of GABAergic interneurons is a fundamental mechanism underlying the generation of brain oscillations, and evidence suggests that NMDA receptors (NMDARs) play a key role in oscillatory activity by regulating the activity of interneurons. Consistent with this, derangement of brain rhythms in certain neuropsychiatric disorders, notably schizophrenia and autism, is associated with NMDAR hypofunction and loss of inhibitory interneurons. In the basolateral amygdala (BLA)-dysfunction of which is involved in a host of neuropsychiatric diseases-, principal neurons display spontaneous, rhythmic "bursts" of inhibitory activity, which could potentially be involved in the orchestration of oscillations in the BLA network; here, we investigated the role of NMDARs in these inhibitory oscillations. Rhythmic bursts of spontaneous IPSCs (0.5 Hz average burst frequency) recorded from rat BLA principal cells were blocked or significantly suppressed by D-AP5, and could be driven by NMDAR activation alone. BLA interneurons generated spontaneous bursts of suprathreshold EPSCs at a similar frequency, which were also blocked or reduced by D-AP5. PEAQX (GluN2A-NMDAR antagonist; 0.4 µM) or Ro-25-6981 (GluN2B-NMDAR antagonist; 5 µM) suppressed the IPSC and EPSC bursts; suppression by PEAQX was significantly greater than that by Ro-25-6981. Immunohistochemical labeling revealed the presence of both GluN2A- and GluN2B-NMDARs on GABAergic BLA interneurons, while, functionally, GluN2A-NMDARs have the dominant role, as suggested by a greater reduction of NMDA-evoked currents by PEAQX versus Ro-25-6981. Entrainment of BLA principal neurons in an oscillatory generation of inhibitory activity depends primarily on activation of GluN2A-NMDARs, and interneuronal GluN2A-NMDARs may play a significant role.


Asunto(s)
Complejo Nuclear Basolateral/metabolismo , Potenciales Postsinápticos Inhibidores/fisiología , Neuronas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Complejo Nuclear Basolateral/citología , Complejo Nuclear Basolateral/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Glutamato Descarboxilasa/metabolismo , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Periodicidad , Ratas Sprague-Dawley , Técnicas de Cultivo de Tejidos
16.
Mol Neurobiol ; 55(1): 187-200, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28844093

RESUMEN

Approximately, 1.7 million Americans suffer a TBI annually and TBI is a major cause of death and disability. The majority of the TBI cases are of the mild type and while most patients recover completely from mild TBI (mTBI) about 10% result in persistent symptoms and some result in lifelong disability. Anxiety disorders are the second most common diagnosis post-TBI. Of note, TBI-induced anxiety disorders are difficult to treat and remain a chronic condition suggesting that new therapies are needed. Previous work from our laboratory demonstrated that a mild TBI induced an anxiety-like phenotype, a key feature of the human condition, associated with loss of GABAergic interneurons and hyperexcitability in the basolateral amygdala (BLA) in rodents 7 and 30 days after a controlled cortical impact (CCI) injury. We now confirm that animals display significantly increased anxiety-like behavior 30 days after CCI. The anxiety-like behavior was associated with a significant loss of GABAergic interneurons and significant reductions in the frequency and amplitude of spontaneous and miniature GABAA-receptor-mediated inhibitory postsynaptic currents (IPSCs) in the BLA. Significantly, subchronic treatment with alpha-linolenic acid (ALA) after CCI prevents the development of anxiety-like behavior, the loss of GABAergic interneurons, hyperexcitability in the BLA and reduces the impact injury. Taken together, administration of ALA after CCI is a potent therapy against the neuropathology and pathophysiological effects of mTBI in the BLA.


Asunto(s)
Ansiedad/prevención & control , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Contusiones/tratamiento farmacológico , Ácido alfa-Linolénico/uso terapéutico , Animales , Ansiedad/etiología , Ansiedad/fisiopatología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/fisiopatología , Contusiones/etiología , Contusiones/fisiopatología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Ácido alfa-Linolénico/farmacología
17.
Neurotoxicology ; 63: 84-89, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28939237

RESUMEN

Organophosphate chemical threat agents (OP-CTA) exert toxic effects through cholinergic over-activation. However, after the initial cholinergic phase, the pathophysiology shifts to a non-cholinergic phase which leads to prolonged status epilepticus (SE), irreversible neuronal degeneration and long-term damage to the central nervous system. The efficacy of delayed treatments against OP-CTA is generally low due to the fact that most drugs fail to inhibit the later phase of non-cholinergic activation. Recently, we reported that intranasal brain delivery of obidoxime (OBD) provides complete neuroprotection against a lethal dose of paraoxon when administered 5min after intoxication. In follow-up studies, we examined the window of effectiveness and found that OBD lost effectiveness around 15min post-exposure, which corresponds to the onset of the non-cholinergic phase of intoxication. However, we observed that a brief isoflurane administration, the inhalation anesthetic used to facilitate intranasal drug administration, was effective against paraoxon-induced neurotoxicity. Thus, the present study aimed to investigate the time-course and dose-response efficacy of a brief 4min isoflurane administration as a treatment for neurotoxicity induced by OP-CTA. We found that isoflurane is a potent anti-seizure agent and neuroprotectant when administered between 20 and 30min after paraoxon exposure, stopping SE within 10min of administration and preventing acute neurodegeneration seen 24h later. We also found that the seizure blocking and neuroprotectant properties of isoflurane, when administered 30min after paraoxon, are dose-dependent. The effectiveness and current clinical use of isoflurane support its use as an innovative approach for post exposure treatment of organophosphate poisoning.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Isoflurano/administración & dosificación , Intoxicación por Organofosfatos/tratamiento farmacológico , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/patología , Animales , Inhibidores de la Colinesterasa/toxicidad , Reactivadores de la Colinesterasa/administración & dosificación , Modelos Animales de Enfermedad , Estudios de Seguimiento , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Cloruro de Obidoxima/administración & dosificación , Intoxicación por Organofosfatos/etiología , Intoxicación por Organofosfatos/patología , Paraoxon/toxicidad , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
18.
Neurotox Res ; 32(4): 694-706, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28776308

RESUMEN

Acute nerve agent exposure causes prolonged status epilepticus (SE), leading to death or long-term brain damage. We have previously demonstrated that LY293558, an AMPA/GluK1 kainate receptor antagonist, terminates SE induced by the nerve agent soman and protects from long-term brain damage, in immature rats and young-adult rats, even if administered with a relatively long latency from the time of exposure. However, susceptibility to the lethal consequences of SE increases with age, and mortality by SE induced by soman is substantially greater in older animals. Therefore, in the present study, we compared the susceptibility to soman toxicity of 10-month-old male rats with that of young-adult male rats (42 to 50 days old) and examined the protective efficacy of LY293558 in the older group. A lower percentage of the 10-month-old rats developed SE after injection of 1.2 × LD50 soman, compared to the young adults, the latency to seizure onset was longer in the older rats, and seizure intensity did not differ between the two age groups. However, mortality rate in the older rats who developed SE was higher than in the young adults. Acetylcholinesterase activity in the amygdala, hippocampus, and piriform cortex did not differ between the two age groups. Administration of LY293558 at 20 or 60 min post-exposure suppressed SE, increased 24-h survival rate, decreased the long-term risk of death, reduced neuronal degeneration in the amygdala, hippocampus, piriform, and entorhinal cortices, and facilitated recovery from body weight loss. Thus, LY293558 is an effective countermeasure against soman toxicity also in older animals.


Asunto(s)
Hipocampo/efectos de los fármacos , Isoquinolinas/farmacología , Degeneración Nerviosa/tratamiento farmacológico , Estado Epiléptico/tratamiento farmacológico , Tetrazoles/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Inhibidores de la Colinesterasa/farmacología , Masculino , Degeneración Nerviosa/patología , Neuropatología/métodos , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
19.
J Pharmacol Exp Ther ; 360(1): 23-32, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27799295

RESUMEN

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


Asunto(s)
Complejo Nuclear Basolateral/efectos de los fármacos , Paraoxon/toxicidad , Receptor Muscarínico M1/antagonistas & inhibidores , Soman/toxicidad , Estado Epiléptico/inducido químicamente , Estado Epiléptico/prevención & control , Sulfonamidas/farmacología , Tiadiazoles/farmacología , Animales , Complejo Nuclear Basolateral/patología , Complejo Nuclear Basolateral/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/patología , Estado Epiléptico/fisiopatología , Sinapsis/efectos de los fármacos , Sinapsis/patología
20.
Neural Plast ; 2016: 8524560, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27313904

RESUMEN

After surgery requiring general anesthesia, patients often experience emotional disturbances, but it is unclear if this is due to anesthetic exposure. In the present study, we examined whether isoflurane anesthesia produces long-term pathophysiological alterations in the basolateral amygdala (BLA), a brain region that plays a central role in emotional behavior. Ten-week-old, male rats were administered either a single, 1 h long isoflurane (1.5%) anesthesia or three, 1 h long isoflurane exposures, separated by 48 h. Long-term potentiation (LTP) and spontaneous GABAergic activity in the BLA were studied 1 day, 1 week, and 1 month later. Single isoflurane anesthesia had no significant effect on the magnitude of LTP. In contrast, after repeated isoflurane exposures, LTP was dramatically impaired at both 1 day and 1 week after the last exposure but was restored by 1 month after the exposures. Spontaneous GABAA receptor-mediated IPSCs were increased at 1 day and 1 week after repeated exposures but had returned to control levels by 1 month after exposure. Thus, repeated exposures to isoflurane cause a long-lasting-but not permanent-impairment of synaptic plasticity in the BLA, which could be due to increased basal GABAergic activity. These pathophysiological alterations may produce emotional disturbances and impaired fear-related learning.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Complejo Nuclear Basolateral/efectos de los fármacos , Neuronas GABAérgicas/efectos de los fármacos , Isoflurano/administración & dosificación , Potenciación a Largo Plazo/efectos de los fármacos , Animales , Complejo Nuclear Basolateral/fisiología , Neuronas GABAérgicas/fisiología , Potenciación a Largo Plazo/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...