Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 388(2): 432-450, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37739807

ABSTRACT

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.


Subject(s)
Brain Injuries , Cyclopentanes , Isoquinolines , Nerve Agents , Neuroprotective Agents , Soman , Status Epilepticus , Tetrazoles , Humans , Infant, Newborn , Rats , Animals , Nerve Agents/toxicity , Midazolam/pharmacology , Midazolam/therapeutic use , Soman/toxicity , Neuroprotection , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Seizures/drug therapy , Anticonvulsants/adverse effects , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Atrophy/drug therapy
2.
Toxics ; 11(10)2023 Oct 18.
Article in English | MEDLINE | ID: mdl-37888716

ABSTRACT

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.
Toxics ; 10(8)2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35893842

ABSTRACT

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.

4.
Amino Acids ; 54(9): 1229-1249, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35798984

ABSTRACT

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.


Subject(s)
Basolateral Nuclear Complex , Brain Injuries, Traumatic , Animals , Anxiety , Brain , Humans
5.
Brain Res ; 1770: 147628, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34454948

ABSTRACT

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.


Subject(s)
Acid Sensing Ion Channels/metabolism , Anxiety/metabolism , Basolateral Nuclear Complex/metabolism , Estrus/physiology , Neural Inhibition/physiology , Acid Sensing Ion Channels/genetics , Animals , Anxiety/physiopathology , Basolateral Nuclear Complex/physiopathology , Exploratory Behavior/physiology , Female , Inhibitory Postsynaptic Potentials/physiology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
6.
Ann N Y Acad Sci ; 1479(1): 122-133, 2020 11.
Article in English | MEDLINE | ID: mdl-32237259

ABSTRACT

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.


Subject(s)
Antidotes/pharmacology , Cyclopentanes/pharmacology , Electrocardiography , Isoquinolines/pharmacology , Midazolam/pharmacology , Nerve Agents/toxicity , Soman/toxicity , Status Epilepticus , Tetrazoles/pharmacology , Animals , Male , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Status Epilepticus/prevention & control
7.
Mol Neurobiol ; 57(1): 23-31, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31520315

ABSTRACT

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.


Subject(s)
Brain Injuries, Traumatic/physiopathology , CA1 Region, Hippocampal/physiopathology , Hippocampus/physiopathology , Inhibitory Postsynaptic Potentials/physiology , Animals , Brain Injuries/complications , Brain Injuries/physiopathology , Brain Injuries, Traumatic/complications , Disease Models, Animal , GABAergic Neurons , Interneurons/metabolism , Male , Memory/physiology , Memory Disorders/complications , Rats, Sprague-Dawley
8.
Epilepsia ; 59 Suppl 2: 92-99, 2018 10.
Article in English | MEDLINE | ID: mdl-30159887

ABSTRACT

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.


Subject(s)
Brain/drug effects , Nerve Agents/toxicity , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Brain/metabolism , Humans , Miosis/drug therapy , Miosis/etiology , Respiration Disorders/chemically induced , Respiration Disorders/drug therapy , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Time Factors
9.
Neurotox Res ; 34(3): 511-524, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29713995

ABSTRACT

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.


Subject(s)
Anticonvulsants/therapeutic use , Brain Injuries/drug therapy , Cyclopentanes/therapeutic use , Isoquinolines/therapeutic use , Neuroprotective Agents/therapeutic use , Seizures/drug therapy , Tetrazoles/therapeutic use , Animals , Body Weight/drug effects , Brain Injuries/chemically induced , Brain Waves/drug effects , Cholinesterase Inhibitors/toxicity , Disease Models, Animal , Drug Therapy, Combination/methods , Electroencephalography , Fluoresceins/metabolism , Fourier Analysis , Male , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Soman/toxicity , Time Factors
10.
J Pharmacol Exp Ther ; 365(2): 314-326, 2018 05.
Article in English | MEDLINE | ID: mdl-29467308

ABSTRACT

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.


Subject(s)
Anticonvulsants/pharmacology , Cyclopentanes/pharmacology , Diazepam/pharmacology , Isoquinolines/pharmacology , Neuroprotective Agents/pharmacology , Soman/pharmacology , Status Epilepticus/drug therapy , Tetrazoles/pharmacology , Animals , Anticonvulsants/therapeutic use , Anxiety/complications , Anxiety/prevention & control , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/pathology , Behavior, Animal/drug effects , Child , Cyclopentanes/therapeutic use , Diazepam/therapeutic use , Disease Models, Animal , Drug Interactions , Humans , Isoquinolines/therapeutic use , Male , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/complications , Tetrazoles/therapeutic use
11.
Neuroscience ; 373: 145-158, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29339324

ABSTRACT

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.


Subject(s)
Basolateral Nuclear Complex/metabolism , Inhibitory Postsynaptic Potentials/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamate Decarboxylase/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Male , Neurons/cytology , Neurons/drug effects , Neurotransmitter Agents/pharmacology , Periodicity , Rats, Sprague-Dawley , Tissue Culture Techniques
12.
Mol Neurobiol ; 55(1): 187-200, 2018 01.
Article in English | MEDLINE | ID: mdl-28844093

ABSTRACT

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.


Subject(s)
Anxiety/prevention & control , Brain Injuries, Traumatic/drug therapy , Contusions/drug therapy , alpha-Linolenic Acid/therapeutic use , Animals , Anxiety/etiology , Anxiety/physiopathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/physiopathology , Contusions/etiology , Contusions/physiopathology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Rats , Rats, Sprague-Dawley , Treatment Outcome , alpha-Linolenic Acid/pharmacology
13.
Neurotox Res ; 32(4): 694-706, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28776308

ABSTRACT

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.


Subject(s)
Hippocampus/drug effects , Isoquinolines/pharmacology , Nerve Degeneration/drug therapy , Status Epilepticus/drug therapy , Tetrazoles/pharmacology , Amygdala/drug effects , Animals , Cholinesterase Inhibitors/pharmacology , Male , Nerve Degeneration/pathology , Neuropathology/methods , Rats, Sprague-Dawley , Seizures/chemically induced
14.
J Pharmacol Exp Ther ; 360(1): 23-32, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27799295

ABSTRACT

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.


Subject(s)
Basolateral Nuclear Complex/drug effects , Paraoxon/toxicity , Receptor, Muscarinic M1/antagonists & inhibitors , Soman/toxicity , Status Epilepticus/chemically induced , Status Epilepticus/prevention & control , Sulfonamides/pharmacology , Thiadiazoles/pharmacology , Animals , Basolateral Nuclear Complex/pathology , Basolateral Nuclear Complex/physiopathology , Male , Rats , Rats, Sprague-Dawley , Status Epilepticus/pathology , Status Epilepticus/physiopathology , Synapses/drug effects , Synapses/pathology
15.
Neural Plast ; 2016: 8524560, 2016.
Article in English | MEDLINE | ID: mdl-27313904

ABSTRACT

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.


Subject(s)
Anesthetics, Inhalation/administration & dosage , Basolateral Nuclear Complex/drug effects , GABAergic Neurons/drug effects , Isoflurane/administration & dosage , Long-Term Potentiation/drug effects , Animals , Basolateral Nuclear Complex/physiology , GABAergic Neurons/physiology , Long-Term Potentiation/physiology , Male , Rats , Rats, Sprague-Dawley
16.
Ann N Y Acad Sci ; 1374(1): 17-28, 2016 06.
Article in English | MEDLINE | ID: mdl-27002925

ABSTRACT

One of the deleterious effects of acute nerve agent exposure is the induction of status epilepticus (SE). If SE is not controlled effectively, it causes extensive brain damage. Here, we review the neuropathology observed after nerve agent-induced SE, as well as the ensuing pathophysiological, neurological, and behavioral alterations, with an emphasis on their time course and longevity. Limbic structures are particularly vulnerable to damage by nerve agent exposure. The basolateral amygdala (BLA), which appears to be a key site for seizure initiation upon exposure, suffers severe neuronal loss; however, GABAergic BLA interneurons display a delayed death, perhaps providing a window of opportunity for rescuing intervention. The end result is a long-term reduction of GABAergic activity in the BLA, with a concomitant increase in spontaneous excitatory activity; such pathophysiological alterations are not observed in the CA1 hippocampal area, despite the extensive neuronal loss. Hyperexcitability in the BLA may be at least in part responsible for the development of recurrent seizures and increased anxiety, while hippocampal damage may underlie the long-term memory impairments. Effective control of SE after nerve agent exposure, such that brain damage is also minimized, is paramount for preventing lasting neurological and behavioral deficits.


Subject(s)
Behavior, Animal/drug effects , Nerve Agents/adverse effects , Nervous System/pathology , Animals , Cognition/drug effects , Interneurons/drug effects , Interneurons/pathology , Nervous System/drug effects , Nervous System/physiopathology , Time Factors
17.
Neurotoxicology ; 53: 64-73, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26751814

ABSTRACT

Intranasal delivery is an emerging method for bypassing the blood brain barrier (BBB) and targeting therapeutics to the CNS. Oximes are used to counteract the effects of organophosphate poisoning, but they do not readily cross the BBB. Therefore, they cannot effectively counteract the central neuropathologies caused by cholinergic over-activation when administered peripherally. For these reasons we examined intranasal administration of oximes in an animal model of severe organophosphate poisoning to determine their effectiveness in reducing mortality and seizure-induced neuronal degeneration. Using the paraoxon model of organophosphate poisoning, we administered the standard treatment (intramuscular pralidoxime plus atropine sulphate) to all animals and then compared the effectiveness of intranasal application of obidoxime (OBD) to saline in the control groups. Intranasally administered OBD was effective in partially reducing paraoxon-induced acetylcholinesterase inhibition in the brain and substantially reduced seizure severity and duration. Further, intranasal OBD completely prevented mortality, which was 41% in the animals given standard treatment plus intranasal saline. Fluoro-Jade-B staining revealed extensive neuronal degeneration in the surviving saline-treated animals 24h after paraoxon administration, whereas no detectable degenerating neurons were observed in any of the animals given intranasal OBD 30min before or 5min after paraoxon administration. These findings demonstrate that intranasally administered oximes bypass the BBB more effectively than those administered peripherally and provide an effective method for protecting the brain from organophosphates. The addition of intranasally administered oximes to the current treatment regimen for organophosphate poisoning would improve efficacy, reducing both brain damage and mortality.


Subject(s)
Brain/enzymology , Central Nervous System Diseases/prevention & control , Cholinesterase Reactivators/therapeutic use , Obidoxime Chloride/therapeutic use , Organophosphate Poisoning , Acetylcholinesterase/metabolism , Administration, Intranasal , Animals , Biological Availability , Brain/drug effects , Central Nervous System Diseases/etiology , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Male , Organophosphate Poisoning/complications , Organophosphate Poisoning/drug therapy , Organophosphate Poisoning/mortality , Pralidoxime Compounds/metabolism , Pralidoxime Compounds/pharmacokinetics , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Tritium/pharmacokinetics
18.
J Neurosci Res ; 94(6): 548-67, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26586374

ABSTRACT

The brain comprises an excitatory/inhibitory neuronal network that maintains a finely tuned balance of activity critical for normal functioning. Excitatory activity in the basolateral amygdala (BLA), a brain region that plays a central role in emotion and motivational processing, is tightly regulated by a relatively small population of γ-aminobutyric acid (GABA) inhibitory neurons. Disruption in GABAergic inhibition in the BLA can occur when there is a loss of local GABAergic interneurons, an alteration in GABAA receptor activation, or a dysregulation of mechanisms that modulate BLA GABAergic inhibition. Disruptions in GABAergic control of the BLA emerge during development, in aging populations, or after trauma, ultimately resulting in hyperexcitability. BLA hyperexcitability manifests behaviorally as an increase in anxiety, emotional dysregulation, or development of seizure activity. This Review discusses the anatomy, development, and physiology of the GABAergic system in the BLA and circuits that modulate GABAergic inhibition, including the dopaminergic, serotonergic, noradrenergic, and cholinergic systems. We highlight how alterations in various neurotransmitter receptors, including the acid-sensing ion channel 1a, cannabinoid receptor 1, and glutamate receptor subtypes, expressed on BLA interneurons, modulate GABAergic transmission and how defects of these systems affect inhibitory tonus within the BLA. Finally, we discuss alterations in the BLA GABAergic system in neurodevelopmental (autism/fragile X syndrome) and neurodegenerative (Alzheimer's disease) diseases and after the development of epilepsy, anxiety, and traumatic brain injury. A more complete understanding of the intrinsic excitatory/inhibitory circuit balance of the amygdala and how imbalances in inhibitory control contribute to excessive BLA excitability will guide the development of novel therapeutic approaches in neuropsychiatric diseases.


Subject(s)
Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/pathology , Brain Diseases/complications , Brain Diseases/pathology , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism , Humans , Interneurons/metabolism , Synaptic Transmission/physiology
19.
J Mol Neurosci ; 57(2): 282-303, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26319264

ABSTRACT

We have previously demonstrated that mild controlled cortical impact (mCCI) injury to rat cortex causes indirect, concussive injury to underlying hippocampus and other brain regions, providing a reproducible model for mild traumatic brain injury (mTBI) and its neurochemical, synaptic, and behavioral sequelae. Here, we extend a preliminary gene expression study of the hippocampus-specific events occurring after mCCI and identify 193 transcripts significantly upregulated, and 21 transcripts significantly downregulated, 24 h after mCCI. Fifty-three percent of genes altered by mCCI within 24 h of injury are predicted to be expressed only in the non-neuronal/glial cellular compartment, with only 13% predicted to be expressed only in neurons. The set of upregulated genes following mCCI was interrogated using Ingenuity Pathway Analysis (IPA) augmented with manual curation of the literature (190 transcripts accepted for analysis), revealing a core group of 15 first messengers, mostly inflammatory cytokines, predicted to account for >99% of the transcript upregulation occurring 24 h after mCCI. Convergent analysis of predicted transcription factors (TFs) regulating the mCCI target genes, carried out in IPA relative to the entire Affymetrix-curated transcriptome, revealed a high concordance with TFs regulated by the cohort of 15 cytokines/cytokine-like messengers independently accounting for upregulation of the mCCI transcript cohort. TFs predicted to regulate transcription of the 193-gene mCCI cohort also displayed a high degree of overlap with TFs predicted to regulate glia-, rather than neuron-specific genes in cortical tissue. We conclude that mCCI predominantly affects transcription of non-neuronal genes within the first 24 h after insult. This finding suggests that early non-neuronal events trigger later permanent neuronal changes after mTBI, and that early intervention after mTBI could potentially affect the neurochemical cascade leading to later reported synaptic and behavioral dysfunction.


Subject(s)
Brain Injuries/metabolism , Hippocampus/metabolism , Transcriptome , Animals , Brain Injuries/pathology , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Cytokines/genetics , Cytokines/metabolism , Male , Neuroglia/metabolism , Neurons/metabolism , Organ Specificity , Rats , Rats, Sprague-Dawley
20.
Exp Neurol ; 273: 11-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26238734

ABSTRACT

Patients that suffer mild traumatic brain injuries (mTBI) often develop cognitive impairments, including memory and learning deficits. The hippocampus shows a high susceptibility to mTBI-induced damage due to its anatomical localization and has been implicated in cognitive and neurological impairments after mTBI. However, it remains unknown whether mTBI cognitive impairments are a result of morphological and pathophysiological alterations occurring in the CA1 hippocampal region. We investigated whether mTBI induces morphological and pathophysiological alterations in the CA1 using the controlled cortical impact (CCI) model. Seven days after CCI, animals subjected to mTBI showed cognitive impairment in the passive avoidance test and deficits to long-term potentiation (LTP) of synaptic transmission. Deficiencies in inducing or maintaining LTP were likely due to an observed reduction in the activation of NMDA but not AMPA receptors. Significant reductions in the frequency and amplitude of spontaneous and miniature GABAA-receptor mediated inhibitory postsynaptic currents (IPSCs) were also observed 7 days after CCI. Design-based stereology revealed that although the total number of neurons was unaltered, the number of GABAergic interneurons is significantly reduced in the CA1 region 7 days after CCI. Additionally, the surface expression of α1, ß2/3, and γ2 subunits of the GABAA receptor were reduced, contributing to a reduced mIPSC frequency and amplitude, respectively. Together, these results suggest that mTBI causes a significant reduction in GABAergic inhibitory transmission and deficits to NMDA receptor mediated currents in the CA1, which may contribute to changes in hippocampal excitability and subsequent cognitive impairments after mTBI.


Subject(s)
Brain Injuries/pathology , CA1 Region, Hippocampal/pathology , GABAergic Neurons/pathology , Inhibitory Postsynaptic Potentials/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Avoidance Learning/physiology , Brain Injuries/complications , Disease Models, Animal , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Glucose Transporter Type 1/metabolism , Glutamate Decarboxylase/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/pathology , Male , Memory Disorders/etiology , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Receptors, GABA-A/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...