Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Neurobiol Dis ; 200: 106633, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39117119

ABSTRACT

The triggers of status epilepticus (SE) in non-epileptic patients can vary widely, from idiopathic causes to exposure to chemoconvulsants. Regardless of its etiology, prolonged SE can cause significant brain damage, commonly resulting in the development of epilepsy, which is often accompanied by increased anxiety. GABAA receptor (GABAAR)-mediated inhibition has a central role among the mechanisms underlying brain damage and the ensuing epilepsy and anxiety. During SE, calcium influx primarily via ionotropic glutamate receptors activates signaling cascades which trigger a rapid internalization of synaptic GABAARs; this weakens inhibition, exacerbating seizures and excitotoxicity. GABAergic interneurons are more susceptible to excitotoxic death than principal neurons. During the latent period of epileptogenesis, the aberrant reorganization in synaptic interactions that follow interneuronal loss in injured brain regions, leads to the formation of hyperexcitable, seizurogenic neuronal circuits, along with disturbances in brain oscillatory rhythms. Reduction in the spontaneous, rhythmic "bursts" of IPSCs in basolateral amygdala neurons is likely to play a central role in anxiogenesis. Protecting interneurons during SE is key to preventing both epilepsy and anxiety. Antiglutamatergic treatments, including antagonism of calcium-permeable AMPA receptors, can be expected to control seizures and reduce excitotoxicity not only by directly suppressing hyperexcitation, but also by counteracting the internalization of synaptic GABAARs. Benzodiazepines, as delayed treatment of SE, have low efficacy due to the reduction and dispersion of their targets (the synaptic GABAARs), but also because themselves contribute to further reduction of available GABAARs at the synapse; furthermore, benzodiazepines may be completely ineffective in the immature brain.


Subject(s)
Anxiety , Receptors, GABA-A , Status Epilepticus , Status Epilepticus/metabolism , Receptors, GABA-A/metabolism , Animals , Humans , Anxiety/metabolism , Neural Inhibition/physiology
2.
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
3.
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.

4.
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.

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.
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
7.
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
8.
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
9.
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
10.
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
11.
Neurotoxicology ; 44: 335-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25150775

ABSTRACT

The recent sarin attack in Syria killed 1429 people, including 426 children, and left countless more to deal with the health consequences of the exposure. Prior to the Syrian chemical assault, nerve agent attacks in Japan left many victims suffering from neuropsychiatric illnesses, particularly anxiety disorders, more than a decade later. Uncovering the neuro-pathophysiological mechanisms underlying the development of anxiety after nerve agent exposure is necessary for successful treatment. Anxiety is associated with hyperexcitability of the basolateral amygdala (BLA). The present study sought to determine the nature of the nerve agent-induced alterations in the BLA, which could explain the development of anxiety. Rats were exposed to soman, at a dose that induced prolonged status epilepticus. Twenty-four hours and 14-days after exposure, neurons from the BLA were recorded using whole-cell patch-clamp techniques. At both the 24h and 14-day post-exposure time-points, the frequency and amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) in the BLA were reduced, along with reduction in the frequency but not amplitude of miniature IPSCs. In addition, activation of α7-nicotinic acetylcholine receptors, a cholinergic receptor that participates in the regulation of BLA excitability and is involved in anxiety, increased spontaneous excitatory postsynaptic currents (sEPSCs) in both soman-exposed rats and controls, but was less effective in increasing sIPSCs in soman-exposed rats. Despite the loss of both interneurons and principal cells after soman-induced status epilepticus, the frequency of sEPSCs was increased in the soman-exposed rats. Impaired function and cholinergic modulation of GABAergic inhibition in the BLA may underlie anxiety disorders that develop after nerve agent exposure.


Subject(s)
Anxiety/chemically induced , Anxiety/physiopathology , Basolateral Nuclear Complex/drug effects , Chemical Warfare Agents/toxicity , Inhibitory Postsynaptic Potentials/drug effects , Receptors, GABA-A/metabolism , Soman/toxicity , Animals , Basolateral Nuclear Complex/physiopathology , Male , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Synaptic Transmission/drug effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism
12.
J Neurosci ; 34(9): 3130-41, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24573273

ABSTRACT

The discovery that even small changes in extracellular acidity can alter the excitability of neuronal networks via activation of acid-sensing ion channels (ASICs) could have therapeutic application in a host of neurological and psychiatric illnesses. Recent evidence suggests that activation of ASIC1a, a subtype of ASICs that is widely distributed in the brain, is necessary for the expression of fear and anxiety. Antagonists of ASIC1a, therefore, have been proposed as a potential treatment for anxiety. The basolateral amygdala (BLA) is central to fear generation, and anxiety disorders are characterized by BLA hyperexcitability. To better understand the role of ASIC1a in anxiety, we attempted to provide a direct assessment of whether ASIC1a activation increases BLA excitability. In rat BLA slices, activation of ASIC1a by low pH or ammonium elicited inward currents in both interneurons and principal neurons, and increased spontaneous IPSCs recorded from principal cells significantly more than spontaneous EPSCs. Epileptiform activity induced by high potassium and low magnesium was suppressed by ammonium. Antagonism of ASIC1a decreased spontaneous IPSCs more than EPSCs, and increased the excitability of the BLA network, as reflected by the pronounced increase of evoked field potentials, suggesting that ASIC1a channels are active in the basal state. In vivo activation or blockade of ASIC1a in the BLA suppressed or increased, respectively, anxiety-like behavior. Thus, in the rat BLA, ASIC1a has an inhibitory and anxiolytic function. The discovery of positive ASIC1a modulators may hold promise for the treatment of anxiety disorders.


Subject(s)
Acid Sensing Ion Channels/metabolism , Amygdala/metabolism , Anxiety/metabolism , Anxiety/pathology , Action Potentials/drug effects , Action Potentials/genetics , Ammonium Compounds/pharmacology , Amygdala/cytology , Amygdala/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anxiety/drug therapy , Dark Adaptation/drug effects , Dark Adaptation/physiology , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Flurbiprofen/pharmacology , GABA-A Receptor Antagonists/pharmacology , Hydrogen-Ion Concentration , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Male , Neurons/classification , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacology
13.
J Neurophysiol ; 110(10): 2358-69, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24004528

ABSTRACT

The basolateral amygdala (BLA) plays a key role in fear-related learning and memory, in the modulation of cognitive functions, and in the overall regulation of emotional behavior. Pathophysiological alterations involving hyperexcitability in this brain region underlie anxiety and other emotional disorders as well as some forms of epilepsy. GABAergic interneurons exert a tight inhibitory control over the BLA network; understanding the mechanisms that regulate their activity is necessary for understanding physiological and disordered BLA functions. The BLA receives dense cholinergic input from the basal forebrain, affecting both normal functions and dysfunctions of the amygdala, but the mechanisms involved in the cholinergic regulation of inhibitory activity in the BLA are unclear. Using whole cell recordings in rat amygdala slices, here we demonstrate that the α(7)-containing nicotinic acetylcholine receptors (α(7)-nAChRs) are present on somatic or somatodendritic regions of BLA interneurons. These receptors are active in the basal state enhancing GABAergic inhibition, and their further, exogenous activation produces a transient but dramatic increase of spontaneous inhibitory postsynaptic currents in principal BLA neurons. In the absence of AMPA/kainate receptor antagonists, activation of α(7)-nAChRs in the BLA network increases both GABAergic and glutamatergic spontaneous currents in BLA principal cells, but the inhibitory currents are enhanced significantly more than the excitatory currents, reducing overall excitability. The anxiolytic effects of nicotine as well as the role of the α(7)-nAChRs in seizure activity involving the amygdala and in mental illnesses, such as schizophrenia and Alzheimer's disease, may be better understood in light of the present findings.


Subject(s)
Amygdala/physiology , Interneurons/physiology , Nerve Net/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Glutamic Acid/metabolism , Male , Neural Inhibition , Rats , Rats, Sprague-Dawley , Synaptic Potentials , gamma-Aminobutyric Acid/metabolism
14.
J Neurosci ; 30(18): 6443-53, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20445070

ABSTRACT

The drug addiction process shares many commonalities with normal learning and memory. Addictive drugs subvert normal synaptic plasticity mechanisms, and the consequent synaptic changes underlie long-lasting modifications in behavior that accrue during the progression from drug use to addiction. Supporting this hypothesis, it was recently shown that nicotine administered to freely moving mice induces long-term synaptic potentiation of the perforant path connection to granule cells of the dentate gyrus. The perforant path carries place and spatial information that links the environment to drug taking. An example of that association is the nicotine-induced synaptic potentiation of the perforant path that was found to underlie nicotine-conditioned place preference. The present study examines the influence of nicotine over local GABAergic inhibition within the dentate gyrus during the drug-induced synaptic potentiation. In vivo recordings from freely moving mice suggested that both feedforward and feedback inhibition onto granules cells were diminished by nicotine during the induction of synaptic potentiation. In vitro brain slice studies indicated that nicotine altered local circuit inhibition within the dentate gyrus leading to disinhibition of granule cells. These changes in local inhibition contributed to nicotine-induced in vivo synaptic potentiation, thus, likely contributed to drug-associated memories. Through this learning process, environmental features become cues that motivate conditioned drug-seeking and drug-taking behaviors.


Subject(s)
Dentate Gyrus/drug effects , Illicit Drugs/pharmacology , Long-Term Potentiation/drug effects , Neural Inhibition/drug effects , Nicotine/pharmacology , Perforant Pathway/drug effects , Animals , Dentate Gyrus/physiology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Neural Inhibition/physiology , Neurons/physiology , Perforant Pathway/physiology , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , gamma-Aminobutyric Acid/physiology
15.
Proc Natl Acad Sci U S A ; 103(30): 11376-80, 2006 Jul 25.
Article in English | MEDLINE | ID: mdl-16847263

ABSTRACT

Acid-sensitive ion channels (ASICs) are proton-gated and belong to the family of degenerin channels. In the mammalian nervous system, ASICs are most well known in sensory neurons, where they are involved in nociception, occurring when injury or inflammation causes acidification. ASICs also are widely expressed in the CNS, and some synaptic roles have been revealed. Because neuronal activity can produce pH changes, ASICs may respond to local acidic transients and alter the excitability of neuronal circuits more widely than is presently appreciated. Furthermore, ASICs have been found to underlie calcium transients that contribute to neuronal death. Degeneration of midbrain dopamine neurons is characteristic of advanced idiopathic Parkinson's disease. Therefore, we tested for functional ASICs in midbrain dopamine neurons of the ventral tegmental area and substantia nigra compacta. Patch-clamp electrophysiology applied to murine midbrain slices revealed abundant acid-sensitive channels. The ASICs were gated and desensitized by extracellular application of millimolar concentrations of NH(4)Cl. Although the NH(4)Cl solution contains micromolar concentrations of NH(3) at pH 7.4, our evidence indicates that NH(4)(+) gates the ASICs. The proton-gated and the ammonium-gated currents were inhibited by tarantula venom (psalmotoxin), which is specific for the ASIC1a subtype. The results show that acid-sensitive channels are expressed in midbrain dopamine neurons and suggest that ammonium sensitivity is a widely distributed ASIC characteristic in the CNS, including the hippocampus. The ammonium sensitivity suggests a role for ASIC1s in hepatic encephalopathy, cirrhosis, and other neuronal disorders that are associated with hyperammonemia.


Subject(s)
Dopamine/metabolism , Hyperammonemia/pathology , Membrane Proteins/physiology , Mesencephalon/metabolism , Nerve Tissue Proteins/physiology , Neurons/metabolism , Quaternary Ammonium Compounds/pharmacology , Sodium Channels/physiology , Acid Sensing Ion Channels , Ammonia/pharmacology , Animals , Arachnida , Central Nervous System/pathology , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Neurons, Afferent/metabolism , Spider Venoms/metabolism
16.
J Neurosci Methods ; 142(1): 55-66, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15652617

ABSTRACT

When studying in vitro brain slices, rapidly applying multiple agonists, antagonists, drugs, or modulatory compounds is a significant technical problem. There are three major ways that multiple compounds are applied to slices: by bath, via a U-tube device, or by pressure application using a "puffer" pipette. Each of these methods has advantages and disadvantages, making each more appropriate for particular purposes. Because puffer pipettes have a small, sharp tip, they are best suited to apply a small quantity of a compound to a well-defined location within the slice. When used in this way, puffer pipettes have two shortcomings. Solution leaking from the tip of the pipette can contaminate the signal, and it is difficult to apply more than one test solution to exactly the same local area of the slice. We describe methods and newly designed devices aimed at overcoming those limitations. Relatively inexpensive approaches are described to apply eight different solutions to the same exact location deep within a brain slice. The validity of the approach is verified by measuring ligand-gated channel currents activated by glutamate (Glu), acetylcholine (ACh), and gamma-amino butyric acid (GABA).


Subject(s)
Brain/drug effects , Brain/physiology , Drug Delivery Systems/instrumentation , Microinjections/instrumentation , Neuropharmacology/instrumentation , Animals , Brain/anatomy & histology , Catheterization/standards , Cholinergic Agonists/pharmacology , Drug Delivery Systems/methods , Excitatory Amino Acid Agonists/pharmacology , GABA Agonists/pharmacology , Ion Channels/agonists , Ion Channels/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microelectrodes/standards , Microelectrodes/trends , Microinjections/methods , Neuropharmacology/methods , Organ Culture Techniques/instrumentation , Organ Culture Techniques/methods , Receptors, Neurotransmitter/agonists , Receptors, Neurotransmitter/physiology
17.
Learn Mem ; 11(1): 60-9, 2004.
Article in English | MEDLINE | ID: mdl-14747518

ABSTRACT

Tobacco use is a major health problem that is estimated to cause 4 million deaths a year worldwide. Nicotine is the main addictive component of tobacco. It acts as an agonist to activate and desensitize nicotinic acetylcholine receptors (nAChRs). A component of nicotine's addictive power is attributable to actions on the mesolimbic dopaminergic system, which serves a fundamental role in the acquisition of behaviors that are inappropriately reinforced by addictive drugs. Here we show that nicotine, in the same concentration and time ranges as obtained from tobacco, has three main actions that regulate the activity of midbrain dopamine (DA) neurons. Nicotine first activates and then desensitizes nAChRs on the DA neurons. This process directly excites the DA neurons for a short period of time before the nAChRs desensitize. Nicotine also enhances glutamatergic excitation and decreases GABAergic inhibition onto DA neurons. These events increase the probability for synaptic plasticity, such as long-term potentiation. The short-lived direct excitation of the DA neurons coupled with the enhanced glutamatergic afferent activity provides the presynaptic and postsynaptic coincidence necessary to initiate synaptic potentiation. In total, these synaptic events lead to a relatively long-lasting heightened activity of midbrain DA neurons. Consistent with other summarized studies, this work indicates that the synaptic changes normally associated with learning and memory can be influenced and commandeered during the nicotine addiction process.


Subject(s)
Acetylcholine/metabolism , Cholinergic Fibers/metabolism , Dopamine/metabolism , Nicotine/adverse effects , Receptors, Nicotinic/metabolism , Substance-Related Disorders/metabolism , Synaptic Transmission , Ventral Tegmental Area/metabolism , Afferent Pathways/physiopathology , Animals , Electrophysiology , Extracellular Space , Glutamic Acid/metabolism , Immunohistochemistry , Microdialysis , Neuronal Plasticity , Nicotine/pharmacology , Nucleus Accumbens/physiopathology , Rats , Rats, Sprague-Dawley , Substance-Related Disorders/physiopathology , Time Factors , Ventral Tegmental Area/physiopathology , gamma-Aminobutyric Acid/metabolism
18.
J Neurosci ; 23(8): 3176-85, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12716925

ABSTRACT

Although many psychopharmacological factors contribute to nicotine addiction, midbrain dopaminergic systems have received much attention because of their roles in reinforcement and associative learning. It is generally thought that the mesocorticolimbic dopaminergic system is important for the acquisition of behaviors that are reinforced by the salient drives of the environment or by the inappropriate stimuli of addictive drugs. Nicotine, as obtained from tobacco, can activate nicotinic acetylcholine receptors (nAChRs) and excite midbrain neurons of the mesocorticolimbic system. Using midbrain slices from rats, wild-type mice, and genetically engineered mice, we have found differences in the nAChR currents from the ventral tegmental area (VTA) and the substantia nigra compacta (SNc). Nicotinic AChRs containing the alpha7 subunit (alpha7* nAChRs) have a low expression density. Electrophysiological analysis of nAChR currents, autoradiography of [125I]-alpha-bungarotoxin binding, and in situ hybridization revealed that alpha7* nAChRs are more highly expressed in the VTA than the SNc. In contrast, beta2* nAChRs are move evenly distributed at a higher density in both the VTA and SNc. At the concentration of nicotine obtained by tobacco smokers, the slow components of current (mainly mediated by beta2* nAChRs) become essentially desensitized. However, the minority alpha7* component of the current in the VTA/SNc is not significantly desensitized by nicotine in the range < or =100 nm. These results suggest that nicotine, as obtained from tobacco, can have multiple effects on the midbrain areas by differentially influencing dopamine neurons of the VTA and SNc and differentially desensitizing alpha7* and non-alpha7 nAChRs.


Subject(s)
Dopamine/metabolism , Mesencephalon/metabolism , Neurons/metabolism , Nicotine/pharmacology , Receptors, Nicotinic/metabolism , Animals , In Vitro Techniques , Mesencephalon/cytology , Mesencephalon/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Neurons/drug effects , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL