Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Pharmacol Exp Ther ; 375(1): 59-68, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32873622

RESUMEN

Organophosphate (OP) exposure induces status epilepticus (SE), a medical emergency with high morbidity and mortality. Current standard medical countermeasures lose efficacy with time so that treatment delays, in the range of tens of minutes, result in increasingly poor outcomes. As part of the Countermeasures Against Chemical Threats Neurotherapeutics Screening Program, we previously developed a realistic model of delayed treatment of OP-induced SE using the OP diisopropyl fluorophosphate (DFP) to screen compounds for efficacy in the termination of SE and elimination of neuronal death. Male rats were implanted for electroencephalogram (EEG) recordings 7 days prior to experimentation. Rats were then exposed to DFP, and SE was induced for 60 minutes and then treated with midazolam (MDZ) plus one of three antiseizure drugs (ASDs)-phenobarbital (PHB), memantine (MEM), or dexmedetomidine (DMT)-in conjunction with antidotes. EEG was recorded for 24 hours, and brains were stained with Fluoro-Jade B for quantification of degenerating neurons. We found that PHB + MDZ induced a prolonged suppression of SE and reduced neuronal death. MEM + MDZ treatment exacerbated SE and increased mortality; however, surviving rats had fewer degenerating neurons. DMT + MDZ significantly suppressed SE with only a minimal reduction in neuronal death. These data demonstrate that delayed treatment of OP-induced SE with other ASDs, when added to MDZ, can achieve greater seizure suppression with additional reduction in degenerating neurons throughout the brain compared with MDZ alone. The effect of a drug on the severity of seizure activity did not necessarily determine the drug's effect on neuronal death under these conditions. SIGNIFICANCE STATEMENT: This study assesses the relative effectiveness of three different delayed-treatment regimens for the control of organophosphate-induced status epilepticus and reduction of subsequent neuronal death. The data demonstrate the potential for highly effective therapies despite significant treatment delay and a potential disconnect between seizure severity and neuronal death.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Dexmedetomidina/administración & dosificación , Isoflurofato/envenenamiento , Memantina/administración & dosificación , Fenobarbital/administración & dosificación , Estado Epiléptico/tratamiento farmacológico , Tiempo de Tratamiento , Animales , Anticonvulsivantes/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/patología , Muerte Celular/efectos de los fármacos , Dexmedetomidina/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Electroencefalografía , Masculino , Memantina/uso terapéutico , Neuronas/efectos de los fármacos , Neuronas/patología , Fenobarbital/uso terapéutico , Prohibitinas , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/patología , Resultado del Tratamiento
2.
Epilepsia ; 60(7): 1387-1398, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31125451

RESUMEN

OBJECTIVE: Exposure to organophosphates (OPs) and OP nerve agents (NAs) causes status epilepticus (SE) and irreversible brain damage. Rapid control of seizure activity is important to minimize neuronal injury and the resulting neurological and behavioral disorders; however, early treatment will not be possible after mass release of OPs or NAs. METHODS: We utilized a delayed-treatment model of OP exposure in adult rats by administration of diisopropyl fluorophosphate (DFP) to study the relationship between the antiseizure and neuroprotective effects of the "standard-of-care" benzodiazepine, midazolam (MDZ), when given at 30, 60, and 120 minutes after SE onset. After electroencephalography (EEG) recordings, neural damage in serial brain sections was studied with Fluoro-Jade B staining. RESULTS: MDZ-induced seizure suppression was equivalent in magnitude regardless of treatment delay (ie, seizure duration). When assessed globally (ie, normalized across 10 different brain regions) for each treatment delay, MDZ administration resulted in only nonsignificant reductions in neuronal death. However, when data for MDZ treatment were combined from all three delay times, a small but significant reduction in global neuronal death was detected when compared to vehicle treatment, which indicated that the substantive MDZ-induced seizure suppression led to only a small reduction in neuronal death. SIGNIFICANCE: In conclusion, MDZ significantly reduced DFP-induced SE intensity when treatment was delayed 30, 60, and even up to 120 minutes; however, this reduction in seizure intensity had no detectable effect on neuronal death at each individual delay time. These data show that although MDZ suppressed seizures, additional neuroprotective therapies are needed to mitigate the effects of OP exposure.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Isoflurofato/envenenamiento , Midazolam/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Intoxicación por Organofosfatos/tratamiento farmacológico , Convulsiones/inducido químicamente , Animales , Anticonvulsivantes/administración & dosificación , Modelos Animales de Enfermedad , Masculino , Midazolam/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Intoxicación por Organofosfatos/complicaciones , Ratas , Ratas Sprague-Dawley , Convulsiones/tratamiento farmacológico
3.
J Neurophysiol ; 119(5): 1693-1698, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29364072

RESUMEN

Macrocyclic lactones (MLs) are commonly used treatments for parasitic worm and insect infections in humans, livestock, and companion animals. MLs target the invertebrate glutamate-activated chloride channel that is not present in vertebrates. MLs are not entirely inert in vertebrates, though; they have been reported to have activity in heterologous expression systems consisting of ligand-gated ion channels that are present in the mammalian central nervous system (CNS). However, these compounds are typically not able to reach significant concentrations in the CNS because of the activity of the blood-brain barrier P-glycoprotein extrusion system. Despite this, these compounds are able to reach low levels in the CNS that may be useful in the design of novel "designer" ligand-receptor systems that can be used to directly investigate neuronal control of behavior in mammals and have potential for use in treating human neurological diseases. To determine whether MLs might affect neurons in intact brains, we investigated the activity of the ML moxidectin (MOX) at native GABA receptors. Specifically, we recorded tonic and phasic miniature inhibitory postsynaptic currents (mIPSCs) in ex vivo brain slices. Our data show that MOX potentiated tonic GABA currents in a dose-dependent manner but had no concomitant effects on phasic GABA currents (i.e., MOX had no effect on the amplitude, frequency, or decay kinetics of mIPSCs). These studies indicate that behavioral experiments that implement a ML-based novel ligand-receptor system should take care to control for potential effects of the ML on native tonic GABA receptors. NEW & NOTEWORTHY We have identified a novel mechanism of action in the mammalian central nervous system for the antihelminthic moxidectin, commonly prescribed to animals worldwide and currently being evaluated for use in humans. Specifically, moxidectin applied to rodent brain slices selectively enhanced the tonic GABA conductance of hippocampal pyramidal neurons.


Asunto(s)
Antihelmínticos/farmacología , Hipocampo/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Macrólidos/farmacología , Células Piramidales/efectos de los fármacos , Receptores de GABA/efectos de los fármacos , Animales , Antihelmínticos/administración & dosificación , Macrólidos/administración & dosificación , Ratones , Ratas , Ratas Sprague-Dawley
4.
Epilepsia ; 55(9): e94-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24995528

RESUMEN

Valnoctamide (VCD), a derivative of valproate, suppresses electrographic seizures in animal models of status epilepticus (SE), even when the seizures are resistant to benzodiazepines (BZDs). We therefore tested the effect of VCD on miniature inhibitory postsynaptic currents (mIPSCs) in CA1 pyramidal cells to determine if VCD acts directly on γ-aminobutyric acid (GABA)A receptors. Bath-applied VCD induced a specific, rapid, dose-dependent, and reversible slowing of the decay of mIPSCs (i.e., increased time constant) with no effect on their frequency or amplitude. This is similar to the effect of BZDs on mIPSCs, but the effect of VCD persisted in the presence of the BZD-binding site antagonist flumazenil, and was additive to the effect of the BZD, diazepam. These data suggest that VCD acts through a different binding site than that of BZDs, which likely accounts for its effect on BZD-refractory SE. A PowerPoint slide summarizing this article is available for download in the Supporting Information section here.


Asunto(s)
Amidas/farmacología , Anticonvulsivantes/farmacología , Región CA1 Hipocampal/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Animales , Animales Recién Nacidos , Benzodiazepinas/farmacocinética , Flumazenil/farmacología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Técnicas de Placa-Clamp , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
5.
Nat Commun ; 13(1): 5688, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36202854

RESUMEN

Human telencephalon is an evolutionarily advanced brain structure associated with many uniquely human behaviors and disorders. However, cell lineages and molecular pathways implicated in human telencephalic development remain largely unknown. We produce human telencephalic organoids from stem cell-derived single neural rosettes and investigate telencephalic development under normal and pathological conditions. We show that single neural rosette-derived organoids contain pallial and subpallial neural progenitors, excitatory and inhibitory neurons, as well as macroglial and periendothelial cells, and exhibit predictable organization and cytoarchitecture. We comprehensively characterize the properties of neurons in SNR-derived organoids and identify transcriptional programs associated with the specification of excitatory and inhibitory neural lineages from a common pool of NPs early in telencephalic development. We also demonstrate that neurons in organoids with a hemizygous deletion of an autism- and intellectual disability-associated gene SHANK3 exhibit intrinsic and excitatory synaptic deficits and impaired expression of several clustered protocadherins. Collectively, this study validates SNR-derived organoids as a reliable model for studying human telencephalic cortico-striatal development and identifies intrinsic, synaptic, and clustered protocadherin expression deficits in human telencephalic tissue with SHANK3 hemizygosity.


Asunto(s)
Trastorno Autístico , Trastorno Autístico/genética , Humanos , Proteínas del Tejido Nervioso/metabolismo , Organoides/metabolismo , Protocadherinas , Telencéfalo
6.
Neuroscience ; 463: 143-158, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33836243

RESUMEN

Benzodiazepines are the primary treatment option for organophosphate (OP)-induced status epilepticus (SE), but these antiseizure drugs (ASDs) lose efficacy as treatment is delayed. In the event of a mass civilian or military exposure, significant treatment delays are likely. New ASDs that combat benzodiazepine-resistant, OP-induced SE are critically needed, particularly if they can be efficacious after a long treatment delay. This study evaluated the efficacy of the Kv7 channel modulator, retigabine, as a novel therapy for OP-induced SE. Adult, male rats were exposed to soman or diisopropyl fluorophosphate (DFP) to elicit SE and monitored by electroencephalogram (EEG) recording. Retigabine was administered alone or adjunctive to midazolam (MDZ) at delays of 20- or 40-min in the soman model, and 60-min in the DFP model. Following EEG recordings, rats were euthanized and brain tissue was collected for Fluoro-Jade B (FJB) staining to quantify neuronal death. In the DFP model, MDZ + 15 mg/kg retigabine suppressed seizure activity and was neuroprotective. In the soman model, MDZ + 30 mg/kg retigabine suppressed seizures at 20- and 40-min delays. Without MDZ, 15 mg/kg retigabine provided partial antiseizure and neuroprotectant efficacy in the DFP model, while 30 mg/kg without MDZ failed to attenuate soman-induced SE. At 60 mg/kg, retigabine without MDZ strongly reduced seizure activity and neuronal degeneration against soman-induce SE. This study demonstrates the antiseizure and neuroprotective efficacy of retigabine against OP-induced SE. Our data suggest retigabine could be a useful adjunct to standard-of-care and has potential for use in the absence of MDZ.


Asunto(s)
Preparaciones Farmacéuticas , Estado Epiléptico , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Carbamatos , Humanos , Masculino , Organofosfatos/uso terapéutico , Fenilendiaminas , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Tiempo de Tratamiento
7.
Neuroscience ; 425: 280-300, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31783100

RESUMEN

Organophosphorus (OP) compounds are deadly chemicals that exert their intoxicating effects through the irreversible inhibition of acetylcholinesterase (AChE). In addition to an excess of peripheral ailments, OP intoxication induces status epilepticus (SE) which if left untreated may lead to permanent brain damage or death. Benzodiazepines are typically the primary therapies for OP-induced SE, but these drugs lose efficacy as treatment time is delayed. The CounterACT Neurotherapeutic Screening (CNS) Program was therefore established by the National Institutes of Health (NIH) to discover novel treatments that may be administered adjunctively with the currently approved medical countermeasures for OP-induced SE in a delayed treatment scenario. The CNS program utilizes in vivo EEG recordings and Fluoro-JadeB (FJB) histopathology in two established rat models of OP-induced SE, soman (GD) and diisopropylfluorophosphate (DFP), to evaluate the anticonvulsant and neuroprotectant efficacy of novel adjunct therapies when administered at 20 or 60 min after the induction of OP-induced SE. Here we report the results of multiple compounds that have previously shown anticonvulsant or neuroprotectant efficacy in other models of epilepsy or trauma. Drugs tested were ganaxolone, diazoxide, bumetanide, propylparaben, citicoline, MDL-28170, and chloroquine. EEG analysis revealed that ganaxolone demonstrated the most robust anticonvulsant activity, whereas all other drugs failed to attenuate ictal activity in both models of OP-induced SE. FJB staining demonstrated that none of the tested drugs had widespread neuroprotective abilities. Overall these data suggest that neurosteroids may represent the most promising anticonvulsant option for OP-induced SE out of the seven unique mechanisms tested here. Additionally, these results suggest that drugs that provide significant neuroprotection from OP-induced SE without some degree of anticonvulsant activity are elusive, which further highlights the necessity to continue screening novel adjunct treatments through the CNS program.


Asunto(s)
Anticonvulsivantes/farmacología , Epilepsia/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Convulsiones/tratamiento farmacológico , Animales , Benzodiazepinas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Inhibidores de la Colinesterasa/farmacología , Epilepsia/patología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Compuestos Organofosforados/farmacología , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
8.
Front Pharmacol ; 10: 560, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31178732

RESUMEN

Seizures induced by organophosphorus nerve agent exposure become refractory to treatment with benzodiazepines because these drugs engage synaptic γ-aminobutyric acid-A receptors (GABAARs) that rapidly internalize during status epilepticus (SE). Extrasynaptic GABAARs, such as those containing α4ß3δ subunits, are a putative pharmacological target to comprehensively manage nerve agent-induced seizures since they do not internalize during SE and are continuously available for activation. Neurosteroids related to allopregnanolone have been tested as a possible replacement for benzodiazepines because they target both synaptic and extrasynaptic GABAARs receptors. A longer effective treatment window, extended treatment efficacy, and enhanced neuroprotection represent significant advantages of neurosteroids over benzodiazepines. However, neurosteroid use is limited by poor physicochemical properties arising from the intrinsic requirement of the pregnane steroid core structure for efficacy rendering drug formulation problematic. We tested a non-steroidal enaminone GABAAR modulator that interacts with both synaptic and extrasynaptic GABAARs on a binding site distinct from neurosteroids or benzodiazepines for efficacy to control electrographic SE induced by diisopropyl fluorophosphate or soman intoxication in rats. Animals were treated with standard antidotes, and experimental therapeutic treatment was given following 1 h (diisopropyl fluorophosphate model) or 20 min (soman model) after SE onset. We found that the enaminone 2-261 had an extended duration of seizure termination (>10 h) in the diisopropyl fluorophosphate intoxication model in the presence or absence of midazolam (MDZ). 2-261 also moderately potentiated MDZ in the soman-induced seizure model but had limited efficacy as a stand-alone anticonvulsant treatment due to slow onset of action. 2-261 significantly reduced neuronal death in brain areas associated with either diisopropyl fluorophosphate- or soman-induced SE. 2-261 represents an alternate chemical template from neurosteroids for enhancing extrasynaptic α4ß3δ GABAAR activity to reverse SE from organophosphorous intoxication.

9.
eNeuro ; 4(4)2017.
Artículo en Inglés | MEDLINE | ID: mdl-28785726

RESUMEN

The death of GABAergic interneurons has long been hypothesized to contribute to acquired epilepsy. These experiments tested the hypothesis that focal interneuron lesions cause acute seizures [i.e., status epilepticus (SE)] and/or chronic epilepsy [i.e., persistent spontaneous recurrent seizures (SRSs)]. To selectively ablate interneurons, Gad2-ires-Cre mice were injected unilaterally in the CA1 area of the dorsal hippocampus with an adeno-associated virus containing the diphtheria toxin receptor (DTR). Simultaneously, an electrode, connected to a miniature telemetry device, was positioned at the injection site for chronic recordings of local field potentials (LFPs). Two weeks after virus transfection, intraperitoneal injection of DT consistently caused focal, specific, and extensive ablation of interneurons. Long-term, continuous monitoring revealed that all mice with DT-induced interneuron lesions had SRSs. Seizures lasted tens of seconds and interseizure intervals were several hours (or days); therefore, these interneuron lesions did not induce SE. The SRSs occurred 3-5 d after DT treatment, which is the estimated time required for DT-induced cell death; therefore, induction of SRSs occurred without the latent period typical of acquired epilepsy. In five of six DT-treated mice, SRSs stopped within days, suggesting that the DT-induced interneuron lesions did not usually cause epilepsy. In one mouse, however, SRSs occurred for ≥34 d after interneuron ablation, similar to epilepsy after experimental SE. Sham control mice had no detectable seizures, confirming that the SRSs were due to ablation of interneurons. These data show that selective interneuron ablation consistently caused SRSs but not SE; and, at least under the conditions used here, interneuron lesions rarely led to persistent SRSs (i.e., epilepsy).


Asunto(s)
Región CA1 Hipocampal/fisiopatología , Interneuronas/fisiología , Convulsiones/fisiopatología , Animales , Región CA1 Hipocampal/patología , Electrodos Implantados , Femenino , Vectores Genéticos , Inmunohistoquímica , Interneuronas/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Placa-Clamp , Prueba de Estudio Conceptual , Convulsiones/etiología , Convulsiones/patología , Potenciales Sinápticos/fisiología , Técnicas de Cultivo de Tejidos , Transfección , Ácido gamma-Aminobutírico/metabolismo
10.
Physiol Rep ; 4(1)2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26733246

RESUMEN

We have previously shown that in the basolateral amygdala (BLA), action potentials in one type of parvalbumin (PV)-expressing GABAergic interneuron can evoke a disynaptic feedback excitatory postsynaptic potential (fbEPSP) onto the same presynaptic interneuron. Here, using whole-cell recordings from PV-expressing interneurons in acute brain slices we expand on this finding to show that this response is first detectable at 2-week postnatal, and is most prevalent in animals beyond 3 weeks of age (>P21). This circuit has a very high fidelity, and single action potential evoked fbEPSPs display few failures. Reconstruction of filled neurons, and electron microscopy show that interneurons that receive feedback excitation make symmetrical synapses on both the axon initial segments (AIS), as well as the soma and proximal dendrites of local pyramidal neurons, suggesting fbEPSP interneurons are morphologically distinct from the highly specialized chandelier neurons that selectively target the axon initial segment of pyramidal neurons. Single PV interneurons could trigger very large (~ 1 nA) feedback excitatory postsynaptic currents (fbEPSCs) suggesting that these neurons are heavily reciprocally connected to local glutamatergic principal cells. We conclude that in the BLA, a subpopulation of PV interneurons forms a distinct neural circuit in which a single action potential can recruit multiple pyramidal neurons to discharge near simultaneously and feed back onto the presynaptic interneuron.


Asunto(s)
Complejo Nuclear Basolateral/metabolismo , Retroalimentación Fisiológica/fisiología , Neuronas GABAérgicas/fisiología , Interneuronas/metabolismo , Parvalbúminas/biosíntesis , Animales , Complejo Nuclear Basolateral/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Antagonistas del GABA/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Regulación de la Expresión Génica , Interneuronas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Técnicas de Cultivo de Órganos , Receptores de GABA/fisiología
11.
PLoS One ; 7(9): e41029, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22957010

RESUMEN

The dentate gyrus is a neurogenic zone where neurons continue to be born throughout life, mature and integrate into the local circuitry. In adults, this generation of new neurons is thought to contribute to learning and memory formation. As newborn neurons mature, they undergo a developmental sequence in which different stages of development are marked by expression of different proteins. Doublecortin (DCX) is an early marker that is expressed in immature granule cells that are beginning migration and dendritic growth but is turned off before neurons reach maturity. In the present study, we use a mouse strain in which enhanced green fluorescent protein (EGFP) is expressed under the control of the DCX promoter. We show that these neurons have high input resistances and some cells can discharge trains of action potentials. In mature granule cells, action potentials are followed by a slow afterhyperpolarization that is absent in EGFP-positive neurons. EGFP-positive neurons had a lower spine density than mature neurons and stimulation of either the medial or lateral perforant pathway activated dual component glutamatergic synapses that had both AMPA and NMDA receptors. NMDA receptors present at these synapses had slow kinetics and were blocked by ifenprodil, indicative of high GluN2B subunit content. These results show that EGFP-positive neurons in the DCX-EGFP mice are functionally immature both in their firing properties and excitatory synapses.


Asunto(s)
Giro Dentado/metabolismo , Regulación de la Expresión Génica , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Neuronas/metabolismo , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Potenciales de Acción , Animales , Animales Recién Nacidos , Calbindina 2 , Movimiento Celular , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Cinética , Masculino , Ratones , Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Neurogénesis , Piperidinas/farmacología , Regiones Promotoras Genéticas , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína G de Unión al Calcio S100/biosíntesis , Ácidos Siálicos/biosíntesis , Sinapsis/metabolismo
12.
Neuropharmacology ; 60(5): 765-73, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21093462

RESUMEN

The amygdala is a temporal lobe structure that is the center of emotion processing in the mammalian brain. Recent interest in the amygdala arises from its role in processing fear and the relationship of fear to human anxiety. The amygdaloid complex is divided into a number of subnuclei that have extensive intra and extra nuclear connections. In this review we discuss recent findings on the physiology and plasticity of inputs to interneurons in the basolateral amygdala, the primary input station. These interneurons are a heterogeneous group of cells that can be separated on immunohistochemical and electrophysiological grounds. Glutamatergic inputs to these interneurons form diverse types of excitatory synapses. This diversity is manifest in both the subunit composition of the underlying NMDA receptors as well as their ability to show plasticity. We discuss these differences and their relationship to fear learning. This article is part of a Special Issue entitled 'Synaptic Plasticity & Interneurons'.


Asunto(s)
Amígdala del Cerebelo/fisiología , Interneuronas/fisiología , Plasticidad Neuronal/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Axones/fisiología , Miedo/fisiología , Humanos , Receptores de N-Metil-D-Aspartato/fisiología
13.
J Neurophysiol ; 98(1): 96-104, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17475718

RESUMEN

Network activity in the 200- to 600-Hz range termed high-frequency oscillations (HFOs) has been detected in epileptic tissue from both humans and rodents and may underlie the mechanism of epileptogenesis in experimental rodent models. Slower network oscillations including theta and gamma oscillations as well as ripples are generated by the complex spike timing and interactions between interneurons and pyramidal cells of the hippocampus. We determined the activity of CA3 pyramidal cells, stratum oriens lacunosum-moleculare (O-LM) and s. radiatum lacunosum-moleculare (R-LM) interneurons during HFO in the in vitro low-Mg(2+) model of epileptiform activity in GIN mice. In these animals, interneurons can be identified prior to cell-attached recordings by the expression of green-fluorescent protein (GFP). Simultaneous local field potential recordings from s. pyramidale and on-cell recordings of individual interneurons and principal cells revealed three primary firing behaviors of the active cells: 36% of O-LM interneurons and 60% of pyramidal cells fired action potentials at high frequencies during the HFO. R-LM interneurons were biphasic in that they fired at high frequency at the beginning of the HFO but stopped firing before its end. When considering only the highest frequency component of the oscillations most pyramidal cells fired on the rising phase of the oscillation. These data provide evidence for functional distinction during HFOs within otherwise homogeneous groups of O-LM interneurons and pyramidal cells.


Asunto(s)
Potenciales de Acción/fisiología , Ventilación de Alta Frecuencia , Hipocampo/citología , Interneuronas/fisiología , Red Nerviosa/fisiología , Células Piramidales/fisiología , Animales , Estimulación Eléctrica/métodos , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Análisis Espectral , Factores de Tiempo
14.
J Neurophysiol ; 91(5): 2040-50, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14702334

RESUMEN

Generalized epilepsy with febrile seizures plus (GEFS+) is an autosomal dominant familial syndrome with a complex seizure phenotype. It is caused by mutations in one of 3 voltage-gated sodium channel subunit genes (SCN1B, SCN1A, and SCN2A) and the GABA(A) receptor gamma2 subunit gene (GBRG2). The biophysical characterization of 3 mutations (T875M, W1204R, and R1648H) in SCN1A, the gene encoding the CNS voltage-gated sodium channel alpha subunit Na(v)1.1, demonstrated a variety of functional effects. The T875M mutation enhanced slow inactivation, the W1204R mutation shifted the voltage dependency of activation and inactivation in the negative direction, and the R1648H mutation accelerated recovery from inactivation. To determine how these changes affect neuronal firing, we used the NEURON simulation software to design a computational model based on the experimentally determined properties of each GEFS+ mutant sodium channel and a delayed rectifier potassium channel. The model predicted that W1204R decreased the threshold, T875M increased the threshold, and R1648H did not affect the threshold for firing a single action potential. Despite the different effects on the threshold for firing a single action potential, all of the mutations resulted in an increased propensity to fire repetitive action potentials. In addition, each mutation was capable of driving repetitive firing in a mixed population of mutant and wild-type channels, consistent with the dominant nature of these mutations. These results suggest a common physiological mechanism for epileptogenesis resulting from sodium channel mutations that cause GEFS+.


Asunto(s)
Epilepsia Generalizada/fisiopatología , Fiebre/complicaciones , Fiebre/fisiopatología , Neuronas/fisiología , Convulsiones/etiología , Convulsiones/fisiopatología , Canales de Sodio/genética , Canales de Sodio/fisiología , Potenciales de Acción/efectos de los fármacos , Algoritmos , Animales , Simulación por Computador , Electrofisiología , Técnicas In Vitro , Cinética , Modelos Neurológicos , Mutación/fisiología , Oocitos/fisiología , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Programas Informáticos , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA