Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Epilepsia ; 2024 May 08.
Article in English | MEDLINE | ID: mdl-38717560

ABSTRACT

OBJECTIVE: Genetic variations in proteins of the mechanistic target of rapamycin (mTOR) pathway cause a spectrum of neurodevelopmental disorders often associated with brain malformations and with intractable epilepsy. The mTORopathies are characterized by hyperactive mTOR pathway and comprise tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) type II. How hyperactive mTOR translates into abnormal neuronal activity and hypersynchronous network remains to be better understood. Previously, the role of upregulated GluN2C-containing glutamate-gated N-methyl-D-aspartate receptors (NMDARs) has been demonstrated for germline defects in the TSC genes. Here, we questioned whether this mechanism would expand to other mTORopathies in the different context of a somatic genetic variation of the MTOR protein recurrently found in FCD type II. METHODS: We used a rat model of FCD created by in utero electroporation of neural progenitors of dorsal telencephalon with expression vectors encoding either the wild-type or the pathogenic MTOR variant (p.S2215F). In this mosaic configuration, patch-clamp whole-cell recordings of the electroporated, spiny stellate neurons and extracellular recordings of the electroporated areas were performed in neocortical slices. Selective inhibitors were used to target mTOR activity and GluN2C-mediated currents. RESULTS: Neurons expressing the mutant protein displayed an excessive activation of GluN2C NMDAR-mediated spontaneous excitatory postsynaptic currents. GluN2C-dependent increase in spontaneous spiking activity was detected in the area of electroporated neurons in the mutant condition and was restricted to a critical time window between postnatal days P9 and P20. SIGNIFICANCE: Somatic MTOR pathogenic variant recurrently found in FCD type II resulted in overactivation of GluN2C-mediated neuronal NMDARs in neocortices of rat pups. The related and time-restricted local hyperexcitability was sensitive to subunit GluN2C-specific blockade. Our study suggests that GluN2C-related pathomechanisms might be shared in common by mTOR-related brain disorders.

2.
Elife ; 122023 10 16.
Article in English | MEDLINE | ID: mdl-37843188

ABSTRACT

The role of myelination for axonal conduction is well-established in projection neurons but little is known about its significance in GABAergic interneurons. Myelination is discontinuous along interneuron axons and the mechanisms controlling myelin patterning and segregation of ion channels at the nodes of Ranvier have not been elucidated. Protein 4.1B is implicated in the organization of the nodes of Ranvier as a linker between paranodal and juxtaparanodal membrane proteins to the spectrin cytoskeleton. In the present study, 4.1B KO mice are used as a genetic model to analyze the functional role of myelin in Lhx6-positive parvalbumin (PV) and somatostatin (SST) neurons, two major classes of GABAergic neurons in the hippocampus. We show that 4.1B-deficiency induces disruption of juxtaparanodal K+ channel clustering and mislocalization of nodal or heminodal Na+ channels. Strikingly, 4.1B-deficiency causes loss of myelin in GABAergic axons in the hippocampus. In particular, stratum oriens SST cells display severe axonal dysmyelination and a reduced excitability. This reduced excitability is associated with a decrease in occurrence probability of small amplitude synaptic inhibitory events on pyramidal cells. In contrast, stratum pyramidale fast-spiking PV cells do not appear affected. In conclusion, our results indicate a class-specific effect of dysmyelination on the excitability of hippocampal interneurons associated with a functional alteration of inhibitory drive.


Subject(s)
Hippocampus , Interneurons , Mice , Animals , Interneurons/physiology , Hippocampus/metabolism , Pyramidal Cells/metabolism , Axons/physiology , GABAergic Neurons/metabolism , Parvalbumins/metabolism
3.
Neurobiol Dis ; 185: 106260, 2023 09.
Article in English | MEDLINE | ID: mdl-37573957

ABSTRACT

Temporal Lobe Epilepsy (TLE) is the most common form of epilepsy in adults. In TLE, recurrent mossy fiber (rMF) sprouting from dentate gyrus granule cells (DGCs) forms an aberrant epileptogenic network between dentate granule cells (DGCs) that operates via ectopically expressed kainate receptors (KARs). It was previously shown that KARs expressed at the rMF-DGC synapses play a prominent role in epileptiform network events in TLE. However, it is not well understood how KARs influence neuronal network dynamics and contribute to the generation of epileptiform network activity in the dentate gyrus. To address this question, we monitored the activity of DGCs using single-cell resolution calcium imaging performed in a reliable in vitro model of TLE. Under our experimental conditions, the most prominent DGC activity patterns were interictal-like epileptiform network events, which were correlated with high levels of neuronal synchronization. The pharmacological blockade of KARs reduced the frequency as well as the number of neurons involved in these events, without altering their spatiotemporal dynamics. Analysis of the microstructure of synchrony showed that blockade of KARs diminished the fraction of neurons forming the main functional cluster. Therefore, we propose that KARs act as modulators in the epileptic network by facilitating the recruitment of neurons into coactive cell assemblies, thereby contributing to the occurrence of epileptiform network events.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Humans , Receptors, Kainic Acid , Neurons/metabolism , Dentate Gyrus/metabolism
4.
Ann Neurol ; 94(4): 745-761, 2023 10.
Article in English | MEDLINE | ID: mdl-37341588

ABSTRACT

OBJECTIVE: Temporal lobe epilepsy (TLE) is characterized by recurrent seizures generated in the limbic system, particularly in the hippocampus. In TLE, recurrent mossy fiber sprouting from dentate gyrus granule cells (DGCs) crea an aberrant epileptogenic network between DGCs which operates via ectopically expressed GluK2/GluK5-containing kainate receptors (KARs). TLE patients are often resistant to anti-seizure medications and suffer significant comorbidities; hence, there is an urgent need for novel therapies. Previously, we have shown that GluK2 knockout mice are protected from seizures. This study aims at providing evidence that downregulating KARs in the hippocampus using gene therapy reduces chronic epileptic discharges in TLE. METHODS: We combined molecular biology and electrophysiology in rodent models of TLE and in hippocampal slices surgically resected from patients with drug-resistant TLE. RESULTS: Here, we confirmed the translational potential of KAR suppression using a non-selective KAR antagonist that markedly attenuated interictal-like epileptiform discharges (IEDs) in TLE patient-derived hippocampal slices. An adeno-associated virus (AAV) serotype-9 vector expressing anti-grik2 miRNA was engineered to specifically downregulate GluK2 expression. Direct delivery of AAV9-anti grik2 miRNA into the hippocampus of TLE mice led to a marked reduction in seizure activity. Transduction of TLE patient hippocampal slices reduced levels of GluK2 protein and, most importantly, significantly reduced IEDs. INTERPRETATION: Our gene silencing strategy to knock down aberrant GluK2 expression demonstrates inhibition of chronic seizure in a mouse TLE model and IEDs in cultured slices derived from TLE patients. These results provide proof-of-concept for a gene therapy approach targeting GluK2 KARs for drug-resistant TLE patients. ANN NEUROL 2023;94:745-761.


Subject(s)
Drug Resistant Epilepsy , Epilepsy, Temporal Lobe , MicroRNAs , Humans , Mice , Animals , Epilepsy, Temporal Lobe/therapy , Temporal Lobe , Hippocampus , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/therapy , Seizures
5.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34740966

ABSTRACT

Cerebellar Purkinje neurons integrate information transmitted at excitatory synapses formed by granule cells. Although these synapses are considered essential sites for learning, most of them appear not to transmit any detectable electrical information and have been defined as silent. It has been proposed that silent synapses are required to maximize information storage capacity and ensure its reliability, and hence to optimize cerebellar operation. Such optimization is expected to occur once the cerebellar circuitry is in place, during its maturation and the natural and steady improvement of animal agility. We therefore investigated whether the proportion of silent synapses varies over this period, from the third to the sixth postnatal week in mice. Selective expression of a calcium indicator in granule cells enabled quantitative mapping of presynaptic activity, while postsynaptic responses were recorded by patch clamp in acute slices. Through this approach and the assessment of two anatomical features (the distance that separates adjacent planar Purkinje dendritic trees and the synapse density), we determined the average excitatory postsynaptic potential per synapse. Its value was four to eight times smaller than responses from paired recorded detectable connections, consistent with over 70% of synapses being silent. These figures remained remarkably stable across maturation stages. According to the proposed role for silent synapses, our results suggest that information storage capacity and reliability are optimized early during cerebellar maturation. Alternatively, silent synapses may have roles other than adjusting the information storage capacity and reliability.


Subject(s)
Cerebellum/growth & development , Animals , Calcium Signaling , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Purkinje Cells/physiology , Synapses/physiology
6.
Neurobiol Dis ; 161: 105547, 2021 12.
Article in English | MEDLINE | ID: mdl-34752924

ABSTRACT

Systemic pilocarpine treatment is one of the most reliable means of inducing temporal lobe epilepsy (TLE). However, the traditional pilocarpine injection protocol using mice was associated with a high death rate, possibly because of cardiorespiratory collapse following status epilepticus (SE). To prevent this, we developed a modified procedure of pilocarpine SE induction, which included a single injection of a moderate dose of caffeine during the induction phase. That new protocol was based on the use of young male mice as well as on a refined Racine's scale. Using that protocol, we report a substantially increased survival rate, thus enabling the generation of a large cohort of mice that exhibited cardinal histological (e.g., mossy fiber sprouting) and electrophysiological (e.g., chronic interictal events and ictal seizures) characteristics associated with TLE. In conclusion, our refined caffeine- and pilocarpine-based protocol substantially improves the outcome of the reliable pilocarpine mouse model of TLE.


Subject(s)
Epilepsy, Temporal Lobe , Status Epilepticus , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Humans , Male , Mice , Pilocarpine/toxicity , Seizures , Status Epilepticus/chemically induced , Status Epilepticus/pathology
7.
Neuropharmacology ; 197: 108699, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34246686

ABSTRACT

Kainate receptors (KARs) constitute a family of ionotropic glutamate receptors (iGluRs) with distinct physiological roles in synapses and neuronal circuits. Despite structural and biophysical commonalities with the other iGluRs, AMPA receptors and NMDA receptors, their role as post-synaptic receptors involved in shaping EPSCs to transmit signals across synapses is limited to a small number of synapses. On the other hand KARs regulate presynaptic release mechanisms and control ion channels and signaling pathways through non-canonical metabotropic actions. We review how these different KAR-dependent mechanisms concur to regulate the activity and plasticity of neuronal circuits in physiological conditions of activation of KARs by endogenous glutamate (as opposed to pharmacological activation by exogenous agonists). KARs have been implicated in neurological disorders, based on genetic association and on physiopathological studies. A well described example relates to temporal lobe epilepsy for which the aberrant recruitment of KARs at recurrent mossy fiber synapses takes part in epileptogenic neuronal activity. In conclusion, KARs certainly represent an underestimated actor in the regulation of neuronal circuits, and a potential therapeutic target awaiting more selective and efficient genetic tools and/or ligands. This article is part of the special Issue on 'Glutamate Receptors - Kainate receptors'.


Subject(s)
Nerve Net/physiology , Receptors, Kainic Acid/physiology , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Humans , Nerve Net/drug effects , Receptors, Kainic Acid/drug effects , Receptors, Kainic Acid/genetics , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism
8.
Front Cell Neurosci ; 14: 609123, 2020.
Article in English | MEDLINE | ID: mdl-33519383

ABSTRACT

The dentate granule cells (DGCs) play a crucial role in learning and memory. Many studies have described the role and physiological properties of these sparsely active neurons using different behavioral contexts. However, the morpho-functional features of DGCs recruited in mice maintained in their home cage (without training), considered as a baseline condition, have not yet been established. Using fosGFP transgenic mice, we observed ex vivo that DGCs recruited in animals maintained in the home cage condition are mature neurons that display a longer dendritic tree and lower excitability compared with non-activated cells. The higher GABAA receptor-mediated shunting inhibition contributes to the lower excitability of DGCs activated in the home environment by shifting the input resistance towards lower values. Remarkably, that shunting inhibition is neither observed in non-activated DGCs nor in DGCs activated during training in virtual reality. In short, our results suggest that strong shunting inhibition and reduced excitability could constitute a distinctive neural signature of mature DGCs recruited in the context of the home environment.

9.
Front Cell Neurosci ; 13: 222, 2019.
Article in English | MEDLINE | ID: mdl-31164806

ABSTRACT

In myelinated fibers, the voltage-gated sodium channels Nav1 are concentrated at the nodal gap to ensure the saltatory propagation of action potentials. The voltage-gated potassium channels Kv1 are segregated at the juxtaparanodes under the compact myelin sheath and may stabilize axonal conduction. It has been recently reported that hippocampal GABAergic neurons display high density of Nav1 channels remarkably in clusters along the axon before myelination (Freeman et al., 2015). In inhibitory neurons, the Nav1 channels are trapped by the ankyrinG scaffold at the axon initial segment (AIS) as observed in pyramidal and granule neurons, but are also forming "pre-nodes," which may accelerate conduction velocity in pre-myelinated axons. However, the distribution of the Kv1 channels along the pre-myelinated inhibitory axons is still unknown. In the present study, we show that two subtypes of hippocampal GABAergic neurons, namely the somatostatin and parvalbumin positive cells, display a selective high expression of Kv1 channels at the AIS and all along the unmyelinated axons. These inhibitory axons are also highly enriched in molecules belonging to the juxtaparanodal Kv1 complex, including the cell adhesion molecules (CAMs) TAG-1, Caspr2, and ADAM22 and the scaffolding protein 4.1B. Here, taking advantage of hippocampal cultures from 4.1B and TAG-1 knock-out mice, we observed that 4.1B is required for the proper positioning of Caspr2 and TAG-1 along the distal axon, and that TAG-1 deficiency induces alterations in the axonal distribution of Caspr2. However, the axonal expression of Kv1 channels and clustering of ankyrinG were not modified. In conclusion, this study allowed the analysis of the hierarchy between channels, CAMs and scaffolding proteins for their expression along hippocampal inhibitory axons before myelination. The early steps of channel compartmentalization preceding myelination may be crucial for stabilizing nerve impulses switching from a continuous to saltatory conduction during network development.

10.
Front Cell Neurosci ; 11: 60, 2017.
Article in English | MEDLINE | ID: mdl-28326015

ABSTRACT

The subiculum is the main output of the hippocampal formation. A high proportion of its principal neurons fire action potentials in bursts triggered by the activation of low threshold calcium currents. This firing pattern promotes synaptic release and regulates spike-timing-dependent plasticity. The subiculum receives a high density of fibers originating from the raphe nuclei, suggesting that serotonin (5-HT) modulates subicular neurons. Here we investigated if and how 5-HT modulates the firing pattern of bursting neurons. By combining electrophysiological analysis with pharmacology, optogenetics and calcium imaging, we demonstrate that 5-HT2C receptors reduce bursting activity by inhibiting a low-threshold calcium current mediated by T-type Ca2+ channels in principal cells of the subiculum. In addition, we show that the activation of this novel pathway decreases bursting activity and the occurrence of epileptiform discharges induced in in vitro models for temporal lobe epilepsy (TLE).

11.
Ann Neurol ; 81(2): 251-265, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28074534

ABSTRACT

OBJECTIVE: Rewiring of excitatory glutamatergic neuronal circuits is a major abnormality in epilepsy. Besides the rewiring of excitatory circuits, an abnormal depolarizing γ-aminobutyric acidergic (GABAergic) drive has been hypothesized to participate in the epileptogenic processes. However, a remaining clinically relevant question is whether early post-status epilepticus (SE) evoked chloride dysregulation is important for the remodeling of aberrant glutamatergic neuronal circuits. METHODS: Osmotic minipumps were used to infuse intracerebrally a specific inhibitor of depolarizing GABAergic transmission as well as a functionally blocking antibody toward the pan-neurotrophin receptor p75 (p75NTR ). The compounds were infused between 2 and 5 days after pilocarpine-induced SE. Immunohistochemistry for NKCC1, KCC2, and ectopic recurrent mossy fiber (rMF) sprouting as well as telemetric electroencephalographic and electrophysiological recordings were performed at day 5 and 2 months post-SE. RESULTS: Blockade of NKCC1 after SE with the specific inhibitor bumetanide restored NKCC1 and KCC2 expression, normalized chloride homeostasis, and significantly reduced the glutamatergic rMF sprouting within the dentate gyrus. This mechanism partially involves p75NTR signaling, as bumetanide application reduced SE-induced p75NTR expression and functional blockade of p75NTR decreased rMF sprouting. The early transient (3 days) post-SE infusion of bumetanide reduced rMF sprouting and recurrent seizures in the chronic epileptic phase. INTERPRETATION: Our findings show that early post-SE abnormal depolarizing GABA and p75NTR signaling fosters a long-lasting rearrangement of glutamatergic network that contributes to the epileptogenic process. This finding defines promising and novel targets to constrain reactive glutamatergic network rewiring in adult epilepsy. Ann Neurol 2017;81:251-265.


Subject(s)
Bumetanide/pharmacology , Mossy Fibers, Hippocampal/drug effects , Receptors, Nerve Growth Factor/drug effects , Signal Transduction/drug effects , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/drug effects , Status Epilepticus/metabolism , Symporters/drug effects , gamma-Aminobutyric Acid/drug effects , Animals , Bumetanide/administration & dosage , Male , Nerve Tissue Proteins , Rats , Rats, Wistar , Receptors, Growth Factor , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Status Epilepticus/drug therapy , Status Epilepticus/physiopathology , K Cl- Cotransporters
12.
eNeuro ; 3(3)2016.
Article in English | MEDLINE | ID: mdl-27257629

ABSTRACT

The dentate gyrus, a major entry point to the hippocampus, gates (or filters) incoming information from the cortex. During sleep or anesthesia, the slow-wave oscillation (SWO) orchestrates hippocampus-neocortex communication, which is important for memory formation. The dentate gate is altered in temporal lobe epilepsy (TLE) early during epileptogenesis, which favors the propagation of pathological activities. Yet, whether the gating of physiological SWO by dentate granule cells (DGCs) is altered in TLE has remained unexplored. We combined intracellular recordings of membrane potential (V m) of DGCs and local field potential recordings of the SWO in parietal cortex in anesthetized rats early during epileptogenesis [post-status epilepticus (SE) rats]. As expected, in control rats, the V m of DGCs weakly and rarely oscillated in the SWO frequency range. In contrast, in post-SE rats, the V m of DGCs displayed strong and long-lasting SWO. In these cells, clear UP and DOWN states, in phase with the neocortical SWO, led to a bimodal V m distribution. In post-SE rats, the firing of DGCs was increased and more temporally modulated by the neocortical SWO. We conclude that UP/DOWN state dynamics dominate the V m of DGCs and firing early during epileptogenesis. This abnormally strong neocortical influence on the dynamics of DGCs may profoundly modify the hippocampus-neocortex dialogue during sleep and associated cognitive functions.


Subject(s)
Dentate Gyrus/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Membrane Potentials/physiology , Neocortex/physiopathology , Neurons/physiology , Animals , Dentate Gyrus/pathology , Disease Models, Animal , Epilepsy, Temporal Lobe/pathology , Lithium Compounds , Male , Microelectrodes , Neocortex/pathology , Neural Pathways/pathology , Neural Pathways/physiopathology , Neurons/pathology , Patch-Clamp Techniques , Periodicity , Pilocarpine , Rats, Wistar
13.
Ann Neurol ; 77(4): 592-606, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25583290

ABSTRACT

OBJECTIVE: Patients with temporal lobe epilepsy often display cognitive comorbidity with recurrent seizures. However, the cellular mechanisms underlying the impairment of neuronal information processing remain poorly understood in temporal lobe epilepsy. Within the hippocampal formation neuronal networks undergo major reorganization, including the sprouting of mossy fibers in the dentate gyrus; they establish aberrant recurrent synapses between dentate granule cells and operate via postsynaptic kainate receptors. In this report, we tested the hypothesis that this aberrant local circuit alters information processing of perforant path inputs constituting the major excitatory afferent pathway from entorhinal cortex to dentate granule cells. METHODS: Experiments were performed in dentate granule cells from control rats and rats with temporal lobe epilepsy induced by pilocarpine hydrochloride treatment. Neurons were recorded in patch clamp in whole cell configuration in hippocampal slices. RESULTS: Our present data revealed that an aberrant readout of synaptic inputs by kainate receptors triggered a long-lasting impairment of the perforant path input-output operation in epileptic dentate granule cells. We demonstrated that this is due to the aberrant activity-dependent potentiation of the persistent sodium current altering intrinsic firing properties of dentate granule cells. INTERPRETATION: We propose that this aberrant activity-dependent intrinsic plasticity, which lastingly impairs the information processing of cortical inputs in dentate gyrus, may participate in hippocampal-related cognitive deficits, such as those reported in patients with epilepsy.


Subject(s)
Dentate Gyrus/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Neuronal Plasticity , Neurons , Animals , Excitatory Postsynaptic Potentials/physiology , Male , Neuronal Plasticity/physiology , Neurons/physiology , Organ Culture Techniques , Rats , Rats, Wistar
14.
Curr Opin Pharmacol ; 20: 83-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25506747

ABSTRACT

Kainate receptors (KARs) are tetrameric ionotropic glutamate receptors composed of the combinations of five subunits GluK1-GluK5. KARs are structurally related to AMPA receptors but they serve quite distinct functions by regulating the activity of synaptic circuits at presynaptic and postsynaptic sites, through either ionotropic or metabotropic actions. Although kainate is a potent neurotoxin known to induce acute seizures through activation of KARs, the actual role of KARs in the clinically-relevant chronic phase of temporal lobe epilepsy (TLE) has long been elusive. Recent evidences have described pathophysiological mechanisms of heteromeric GluK2/GluK5 KARs in generating recurrent seizures in chronic epilepsy. The role of the other major subunit GluK1 in epileptogenic activity is still a matter of debate. This review will present the current knowledge on the subtype-specific pharmacology of KARs and highlight recent results linking KARs to epileptic conditions.


Subject(s)
Epilepsy/physiopathology , Receptors, Kainic Acid/metabolism , Synapses/metabolism , Animals , Anticonvulsants/pharmacology , Epilepsy/drug therapy , Humans , Kainic Acid/toxicity , Neurotoxins/toxicity
15.
Cell Rep ; 8(2): 347-54, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25043179

ABSTRACT

Kainate is a potent neurotoxin known to induce acute seizures. However, whether kainate receptors (KARs) play any role in the pathophysiology of temporal lobe epilepsy (TLE) is not known. In TLE, recurrent mossy fiber (rMF) axons form abnormal excitatory synapses onto other dentate granule cells that operate via KARs. The present study explores the pathophysiological implications of KARs in generating recurrent seizures in chronic epilepsy. In an in vitro model of TLE, seizure-like activity was minimized in mice lacking the GluK2 subunit, which is a main component of aberrant synaptic KARs at rMF synapses. In vivo, the frequency of interictal spikes and ictal discharges was strongly reduced in GluK2(-/-) mice or in the presence of a GluK2/GluK5 receptor antagonist. Our data show that aberrant GluK2-containing KARs play a major role in the chronic seizures that characterize TLE and thus constitute a promising antiepileptic target.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Mossy Fibers, Hippocampal/physiology , Receptors, Kainic Acid/genetics , Seizures/metabolism , Animals , Epilepsy, Temporal Lobe/physiopathology , Excitatory Postsynaptic Potentials , Male , Mice , Mossy Fibers, Hippocampal/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Seizures/physiopathology , GluK2 Kainate Receptor
16.
Cereb Cortex ; 23(6): 1484-94, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22628459

ABSTRACT

Epileptic encephalopathies comprise a heterogeneous group of severe infantile disorders for which the pathophysiological basis of epilepsy is inaccurately clarified by genotype-phenotype analysis. Because a deficit of GABA neurons has been found in some of these syndromes, notably in patients with X-linked lissencephaly with abnormal genitalia, epilepsy was suggested to result from an imbalance in GABAergic inhibition, and the notion of "interneuronopathy" was proposed. Here, we studied the impact of a polyalanine expansion of aristaless-related homeobox (ARX) gene, a mutation notably found in West and Ohtahara syndromes. Analysis of Arx((GCG)7/Y) knock-in mice revealed that GABA neuron development is not affected. Moreover, pyramidal cell migration and cortical layering are unaltered in these mice. Interestingly, electrophysiological recordings show that hippocampal pyramidal neurons displayed a frequency of inhibitory postsynaptic currents similar to wild-type (WT) mice. However, these neurons show a dramatic increase in the frequency of excitatory inputs associated with a remodeling of their axonal arborization, suggesting that epilepsy in Arx((GCG)7/Y)mice would result from a glutamate network remodeling. We therefore propose that secondary alterations are instrumental for the development of disease-specific phenotypes and should be considered to explain the phenotypic diversity associated with epileptogenic mutations.


Subject(s)
GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/genetics , Glutamates/metabolism , Homeodomain Proteins/genetics , Peptides/genetics , Transcription Factors/genetics , gamma-Aminobutyric Acid/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/drug effects , Action Potentials/genetics , Age Factors , Animals , Animals, Newborn , Cell Movement/genetics , Doublecortin Protein , Electroporation , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Female , GABAergic Neurons/cytology , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , RNA, Small Interfering/genetics , Statistics, Nonparametric , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Transfection
17.
Cereb Cortex ; 23(2): 323-31, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22345355

ABSTRACT

Progress in understanding the roles of kainate receptors (KARs) in synaptic integration, synaptic networks, and higher brain function has been hampered by the lack of selective pharmacological tools. We have found that UBP310 and related willardiine derivatives, previously characterized as selective GluK1 and GluK3 KAR antagonists, block postsynaptic KARs at hippocampal mossy fiber (MF) CA3 synapses while sparing AMPA and NMDA receptors. We further show that UBP310 is an antagonist of recombinant GluK2/GluK5 receptors, the major population of KARs in the brain. Postsynaptic KAR receptor blockade at MF synapses significantly reduces the sustained depolarization, which builds up during repetitive activity, and impacts on spike transmission mediated by heterosynaptic signals. In addition, KARs present in aberrant MF synapses in the epileptic hippocampus were also blocked by UBP310. Our results support a specific role for postsynaptic KARs in synaptic integration of CA3 pyramidal cells and describe a tool that will be instrumental in understanding the physiopathological role of KARs in the brain.


Subject(s)
Epilepsy, Temporal Lobe/physiopathology , Mossy Fibers, Hippocampal/physiology , Receptors, Kainic Acid/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Animals , Disease Models, Animal , Epilepsy, Temporal Lobe/metabolism , Excitatory Postsynaptic Potentials/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Rats , Rats, Wistar
18.
J Physiol ; 591(4): 799-805, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23027823

ABSTRACT

Cerebral cortex is a highly sophisticated computing machine, feeding on information provided by the senses, which is integrated with other, internally generated patterns of neural activity, to trigger behavioural outputs. Bit by bit, we are coming to understand how this may occur, but still, the nature of the 'cortical code' remains one of the greatest challenges in science. As with other great scientific challenges of the past, fresh insights have come from a coalescence of different experimental and theoretical approaches. These theoretical considerations are typically reserved for cortical function rather than cortical pathology. This approach, though, may also shed light on cortical dysfunction. The particular focus of this review is epilepsy; we will argue that the information capacity of different brain states provides a means of understanding, and even assessing, the impact and locality of the epileptic pathology. Epileptic discharges, on account of their all-consuming and stereotyped nature, represent instances where the information capacity of the network is massively compromised. These intense discharges also prevent normal processing in surrounding territories, but in a different way, through enhanced inhibition in these territories. Information processing is further compromised during the period of post-ictal suppression, during interictal bursts, and also at other times, through more subtle changes in synaptic function. We also comment on information processing in other more physiological brain states.


Subject(s)
Brain/physiology , Epilepsy/physiopathology , Animals , Humans , Neurons/physiology , gamma-Aminobutyric Acid/physiology
19.
J Neurosci ; 31(30): 10811-8, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21795533

ABSTRACT

Dentate granule cells, at the gate of the hippocampus, use coincidence detection of synaptic inputs to code afferent information under a sparse firing regime. In both human patients and animal models of temporal lobe epilepsy, mossy fibers sprout to form an aberrant glutamatergic network between dentate granule cells. These new synapses operate via long-lasting kainate receptor-mediated events, which are not present in the naive condition. Here, we report that in chronic epileptic rat, aberrant kainate receptors in interplay with the persistent sodium current dramatically expand the temporal window for synaptic integration. This introduces a multiplicative gain change in the input-output operation of dentate granule cells. As a result, their sparse firing is switched to an abnormal sustained and rhythmic mode. We conclude that synaptic kainate receptors dramatically alter the fundamental coding properties of dentate granule cells in temporal lobe epilepsy.


Subject(s)
Action Potentials/physiology , Dentate Gyrus/pathology , Epilepsy, Temporal Lobe/pathology , Neurons/physiology , Receptors, Kainic Acid/metabolism , Sodium Channels/physiology , Synapses/physiology , Action Potentials/drug effects , Animals , Biophysics , Disease Models, Animal , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Male , Neurons/drug effects , Patch-Clamp Techniques/methods , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Tetrodotoxin/pharmacology
20.
Cereb Cortex ; 20(4): 898-911, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19684246

ABSTRACT

Spike timing precision is a fundamental aspect of neuronal information processing in the brain. Here we examined the temporal precision of input-output operation of dentate granule cells (DGCs) in an animal model of temporal lobe epilepsy (TLE). In TLE, mossy fibers sprout and establish recurrent synapses on DGCs that generate aberrant slow kainate receptor-mediated excitatory postsynaptic potentials (EPSP(KA)) not observed in controls. We report that, in contrast to time-locked spikes generated by EPSP(AMPA) in control DGCs, aberrant EPSP(KA) are associated with long-lasting plateaus and jittered spikes during single-spike mode firing. This is mediated by a selective voltage-dependent amplification of EPSP(KA) through persistent sodium current (I(NaP)) activation. In control DGCs, a current injection of a waveform mimicking the slow shape of EPSP(KA) activates I(NaP) and generates jittered spikes. Conversely in epileptic rats, blockade of EPSP(KA) or I(NaP) restores the temporal precision of EPSP-spike coupling. Importantly, EPSP(KA) not only decrease spike timing precision at recurrent mossy fiber synapses but also at perforant path synapses during synaptic integration through I(NaP) activation. We conclude that a selective interplay between aberrant EPSP(KA) and I(NaP) severely alters the temporal precision of EPSP-spike coupling in DGCs of chronic epileptic rats.


Subject(s)
Dentate Gyrus/pathology , Epilepsy/pathology , Excitatory Postsynaptic Potentials/physiology , Neurons/physiology , Receptors, Kainic Acid/metabolism , Sodium Channels/physiology , Action Potentials/physiology , Animals , Biophysics , Computer Simulation , Disease Models, Animal , Electric Stimulation/methods , Epilepsy/chemically induced , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Male , Models, Neurological , Mossy Fibers, Hippocampal/physiopathology , Patch-Clamp Techniques , Pilocarpine , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...