Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cereb Cortex ; 32(14): 2907-2923, 2022 07 12.
Article in English | MEDLINE | ID: mdl-34730179

ABSTRACT

Kainate receptors (KARs) are key regulators of synaptic circuits by acting at pre- and postsynaptic sites through either ionotropic or metabotropic actions. KARs can be activated by kainate, a potent neurotoxin, which induces acute convulsions. Here, we report that the acute convulsive effect of kainate mostly depends on GluK2/GluK5 containing KARs. By contrast, the acute convulsive activity of pilocarpine and pentylenetetrazol is not alleviated in the absence of KARs. Unexpectedly, the genetic inactivation of GluK2 rather confers increased susceptibility to acute pilocarpine-induced seizures. The mechanism involves an enhanced excitability of GluK2-/- CA3 pyramidal cells compared with controls upon pilocarpine application. Finally, we uncover that the absence of GluK2 increases pilocarpine modulation of Kv7/M currents. Taken together, our findings reveal that GluK2-containing KARs can control the excitability of hippocampal circuits through interaction with the neuromodulatory cholinergic system.


Subject(s)
Kainic Acid , Pilocarpine , Receptors, Kainic Acid , CA1 Region, Hippocampal/metabolism , Cholinergic Agents/pharmacology , Gene Deletion , Humans , Pilocarpine/toxicity , Pyramidal Cells/metabolism , Receptors, Kainic Acid/genetics , Seizures/chemically induced , Seizures/genetics , GluK2 Kainate Receptor
2.
J Neurosci ; 37(3): 587-598, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28100741

ABSTRACT

Despite extensive studies in hippocampal slices and incentive from computational theories, the synaptic mechanisms underlying information transfer at mossy fiber (mf) connections between the dentate gyrus (DG) and CA3 neurons in vivo are still elusive. Here we used an optogenetic approach in mice to selectively target and control the activity of DG granule cells (GCs) while performing whole-cell and juxtacellular recordings of CA3 neurons in vivo In CA3 pyramidal cells (PCs), mf-CA3 synaptic responses consisted predominantly of an IPSP at low stimulation frequency (0.05 Hz). Upon increasing the frequency of stimulation, a biphasic response was observed consisting of a brief mf EPSP followed by an inhibitory response lasting on the order of 100 ms. Spike transfer at DG-CA3 interneurons recorded in the juxtacellular mode was efficient at low presynaptic stimulation frequency and appeared insensitive to an increased frequency of GC activity. Overall, this resulted in a robust and slow feedforward inhibition of spike transfer at mf-CA3 pyramidal cell synapses. Short-term plasticity of EPSPs with increasing frequency of presynaptic activity allowed inhibition to be overcome to reach spike discharge in CA3 PCs. Whereas the activation of GABAA receptors was responsible for the direct inhibition of light-evoked spike responses, the slow inhibition of spiking activity required the activation of GABAB receptors in CA3 PCs. The slow inhibitory response defined an optimum frequency of presynaptic activity for spike transfer at ∼10 Hz. Altogether these properties define the temporal rules for efficient information transfer at DG-CA3 synaptic connections in the intact circuit. SIGNIFICANCE STATEMENT: Activity-dependent changes in synaptic strength constitute a basic mechanism for memory. Synapses from the dentate gyrus (DG) to the CA3 area of the hippocampus are distinctive for their prominent short-term plasticity, as studied in slices. Plasticity of DG-CA3 connections may assist in the encoding of precise memory in the CA3 network. Here we characterize DG-CA3 synaptic transmission in vivo using targeted optogenetic activation of DG granule cells while recording in whole-cell patch-clamp and juxtacellular configuration from CA3 pyramidal cells and interneurons. We show that, in vivo, short-term plasticity of excitatory inputs to CA3 pyramidal cells combines with robust feedforward inhibition mediated by both GABAA and GABAB receptors to control the efficacy and temporal rules for information transfer at DG-CA3 connections.


Subject(s)
Action Potentials/physiology , GABA Antagonists/pharmacology , Mossy Fibers, Hippocampal/physiology , Neural Inhibition/physiology , Receptors, GABA-A/physiology , Receptors, GABA-B/physiology , Action Potentials/drug effects , Animals , Male , Mice , Mice, Transgenic , Mossy Fibers, Hippocampal/drug effects , Neural Inhibition/drug effects
3.
J Physiol ; 594(13): 3489-500, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26969302

ABSTRACT

BK channels are large conductance potassium channels characterized by four pore-forming α subunits, often co-assembled with auxiliary ß and γ subunits to regulate Ca(2+) sensitivity, voltage dependence and gating properties. Abundantly expressed in the CNS, they have the peculiar characteristic of being activated by both voltage and intracellular calcium rise. The increase in intracellular calcium via voltage-dependent calcium channels (Cav ) during spiking triggers conformational changes and BK channel opening. This narrows the action potential and induces a fast after-hyperpolarization that shuts calcium channels. The tight coupling between BK and Cav channels at presynaptic active zones makes them particularly suitable for regulating calcium entry and neurotransmitter release. While in most synapses, BK channels exert a negative control on transmitter release under basal conditions, in others they do so only under pathological conditions, serving as an emergency brake to protect against hyperactivity. In particular cases, by interacting with other channels (i.e. limiting the activation of the delayed rectifier and the inactivation of Na(+) channels), BK channels induce spike shortening, increase in firing rate and transmitter release. Changes in transmitter release following BK channel dysfunction have been implicated in several neurological disorders including epilepsy, schizophrenia, fragile X syndrome, mental retardation and autism. In particular, two mutations, one in the α and one in the ß3 subunit, resulting in a gain of function have been associated with epilepsy. Hence, these discoveries have allowed identification of BK channels as new drug targets for therapeutic intervention.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Humans , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Neurotransmitter Agents/physiology , Presynaptic Terminals/physiology , Synaptic Transmission
4.
J Neurosci ; 33(3): 1044-9, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23325242

ABSTRACT

In the hippocampus, at excitatory synapses between principal cell and oriens/alveus (O/A) interneurons, a particular form of NMDA-independent long-term synaptic plasticity (LTP) has been described (Lamsa et al., 2007). This type of LTP occurs when presynaptic activation coincides with postsynaptic hyperpolarization. For this reason it has been named "anti-Hebbian" to distinguish from the classical Hebbian type of associative learning where presynaptic glutamate release coincides with postsynaptic depolarization. The different voltage dependency of LTP induction is thought to be mediated by calcium-permeable (CP) AMPA receptors that, due to polyamine-mediated rectification, favor calcium entry at hyperpolarized potentials. Here, we report that the induction of this form of LTP needs CP-α7 nicotinic acetylcholine receptors (nAChRs) that, like CP-AMPARs, exhibit a strong inward rectification because of polyamine block at depolarizing potentials. We found that high-frequency stimulation of afferent fibers elicits synaptic currents mediated by α7 nAChRs. Hence, LTP was prevented by α7 nAChR antagonists dihydro-ß-erythroidine and methyllycaconitine (MLA) and was absent in α7(-/-) mice. In addition, in agreement with previous observations (Le Duigou and Kullmann, 2011), in a minority of O/A interneurons in MLA-treated hippocampal slices from WT animals and α7(-/-) mice, a form of LTP probably dependent on the activation of group I metabotropic glutamate receptors was observed. These data indicate that, in O/A interneurons, anti-Hebbian LTP critically depends on cholinergic signaling via α7 nAChR. This may influence network oscillations and information processing.


Subject(s)
CA1 Region, Hippocampal/physiology , Cholinergic Neurons/physiology , Interneurons/physiology , Long-Term Potentiation/physiology , Receptors, Nicotinic/metabolism , Aconitine/analogs & derivatives , Aconitine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Dihydro-beta-Erythroidine/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Interneurons/drug effects , Interneurons/metabolism , Long-Term Potentiation/drug effects , Mice , Mice, Transgenic , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Receptors, Nicotinic/genetics , Synapses/drug effects , Synapses/physiology , alpha7 Nicotinic Acetylcholine Receptor
5.
Elife ; 122023 07 18.
Article in English | MEDLINE | ID: mdl-37462671

ABSTRACT

Oscillations occurring simultaneously in a given area represent a physiological unit of brain states. They allow for temporal segmentation of spikes and support distinct behaviors. To establish how multiple oscillatory components co-vary simultaneously and influence neuronal firing during sleep and wakefulness in mice, we describe a multivariate analytical framework for constructing the state space of hippocampal oscillations. Examining the co-occurrence patterns of oscillations on the state space, across species, uncovered the presence of network constraints and distinct set of cross-frequency interactions during wakefulness compared to sleep. We demonstrated how the state space can be used as a canvas to map the neural firing and found that distinct neurons during navigation were tuned to different sets of simultaneously occurring oscillations during sleep. This multivariate analytical framework provides a window to move beyond classical bivariate pipelines for investigating oscillations and neuronal firing, thereby allowing to factor-in the complexity of oscillation-population interactions.


Subject(s)
Hippocampus , Sleep , Mice , Animals , Hippocampus/physiology , Sleep/physiology , Neurons/physiology , Brain
6.
iScience ; 26(1): 105728, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36582822

ABSTRACT

In Neurodevelopmental Disorders, alterations of synaptic plasticity may trigger structural changes in neuronal circuits involved in cognitive functions. This hypothesis was tested in mice carrying the human R451C mutation of Nlgn3 gene (NLG3R451C KI), found in some families with autistic children. To this aim, the spike time dependent plasticity (STDP) protocol was applied to immature GABAergic Mossy Fibers (MF)-CA3 connections in hippocampal slices from NLG3R451C KI mice. These animals failed to exhibit STD-LTP, an effect that persisted in adulthood when these synapses became glutamatergic. Similar results were obtained in mice lacking the Nlgn3 gene (NLG3 KO mice), suggesting a loss of function. The loss of STD-LTP was associated with a premature shift of GABA from the depolarizing to the hyperpolarizing direction, a reduced BDNF availability and TrkB phosphorylation at potentiated synapses. These effects may constitute a general mechanism underlying cognitive deficits in those forms of Autism caused by synaptic dysfunctions.

7.
Front Cell Neurosci ; 17: 1332179, 2023.
Article in English | MEDLINE | ID: mdl-38298376

ABSTRACT

Autism spectrum disorders (ASDs) comprise developmental disabilities characterized by impairments of social interaction and repetitive behavior, often associated with cognitive deficits. There is no current treatment that can ameliorate most of the ASDs symptomatology; thus, identifying novel therapies is urgently needed. Here, we used the Neuroligin 3 knockout mouse (NLG3-/y), a model that recapitulates the social deficits reported in ASDs patients, to test the effects of systemic administration of IGF-2, a polypeptide that crosses the blood-brain barrier and acts as a cognitive enhancer. We show that systemic IGF-2 treatment reverses the typical defects in social interaction and social novelty discrimination reflective of ASDs-like phenotypes. This effect was not accompanied by any change in spontaneous glutamatergic synaptic transmission in CA2 hippocampal region, a mechanism found to be crucial for social novelty discrimination. However, in both NLG3+/y and NLG3-/y mice IGF-2 increased cell excitability. Although further investigation is needed to clarify the cellular and molecular mechanisms underpinning IGF-2 effect on social behavior, our findings highlight IGF-2 as a potential pharmacological tool for the treatment of social dysfunctions associated with ASDs.

8.
J Physiol ; 590(4): 655-66, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22124144

ABSTRACT

The hippocampal network comprises a large variety of locally connected GABAergic interneurons exerting a powerful control on network excitability and which are responsible for the oscillatory behaviour crucial for information processing. GABAergic interneurons receive an important cholinergic innervation from the medial septum-diagonal band complex of the basal forebrain and are endowed with a variety of muscarinic and nicotinic acetylcholine receptors (mAChRs and nAChRs) that regulate their activity. Deficits in the cholinergic system lead to the impairment of high cognitive functions, which are particularly relevant in neurodegenerative pathologies such as Alzheimer's and Parkinson's diseases as well as in schizophrenia. Here, we highlight some recent advances in the mechanisms by which cholinergic signalling via nAChRs regulates local inhibitory circuits in the hippocampus, early in postnatal life and in adulthood. We also discuss recent findings concerning the functional role of nAChRs in controlling short- and long-term modifications of synaptic efficacy. Insights into these processes may provide new targets for the therapeutic control of pathological conditions associated with cholinergic dysfunctions.


Subject(s)
Hippocampus/physiology , Receptors, Nicotinic/physiology , Animals , Interneurons/physiology , Neuronal Plasticity , gamma-Aminobutyric Acid/physiology
9.
Front Neural Circuits ; 16: 965172, 2022.
Article in English | MEDLINE | ID: mdl-36082110

ABSTRACT

Animal species are named social when they develop the capability of complex behaviors based on interactions with conspecifics that include communication, aggression, mating and parental behavior, crucial for well-being and survival. The underpinning of such complex behaviors is social memory, namely the capacity to discriminate between familiar and novel individuals. The Medial Septum (MS), a region localized in the basal forebrain, is part of the brain network involved in social memory formation. MS receives several cortical and subcortical synaptic and neuromodulatory inputs that make it an important hub in processing social information relevant for social memory. Particular attention is paid to synaptic inputs that control both the MS and the CA2 region of the hippocampus, one of the major MS output, that has been causally linked to social memory. In this review article, we will provide an overview of local and long range connectivity that allows MS to integrate and process social information. Furthermore, we will summarize previous strategies used to determine how MS controls social memory in different animal species. Finally, we will discuss the impact of an altered MS signaling on social memory in animal models and patients affected by neurodevelopmental and neurodegenerative disorders, including autism and Alzheimer's Disease.


Subject(s)
Alzheimer Disease , Hippocampus , Animals , Social Behavior
10.
J Neurosci ; 30(32): 10773-83, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20702707

ABSTRACT

In the brain, high cognitive functions are encoded by coherent network oscillations. Key players are inhibitory interneurons that, by releasing GABA into principal cells, pace targeted cells. Among these, oriens-lacunosum moleculare (O-LM) interneurons that provide a theta frequency patterned output to distal dendrites of pyramidal cells are endowed with HCN channels responsible for the slowly activating inwardly rectifying Ih current and their pacemaking activity. Here we show that, in transgenic mice expressing EGFP (enhanced green fluorescent protein) in a subset of stratum oriens somatostatin-containing interneurons that mostly comprise O-LM cells, nicotine, the active component of tobacco, reduced Ih and the oscillatory behavior of O-LM interneurons. In cells hyperpolarized at -90 mV, nicotine suppressed the theta resonance in the same way as ZD 7288 (4-ethylphenylamino-1,2-dimethyl-6-methylaminopyrimidinium chloride), a selective blocker of Ih. Nicotine blocked Ih in a concentration-dependent way with an EC50 of 62 nm. Similar effects were produced by epibatidine, a structural analog of nicotine. The effects of nicotine and epibatidine were independent on nicotinic ACh receptor (nAChR) activation because they persisted in the presence of nAChR antagonists. Furthermore, nicotine slowed down the interspike depolarizing slope and the firing rate, thus severely disrupting the oscillatory behavior of O-LM cells. Molecular modeling suggests that, similarly to ZD 7288, nicotine and epibatidine directly bind to the inner pore of the HCN channels. It is therefore likely that nicotine severely influences rhythmogenesis and high cognitive functions in smokers.


Subject(s)
Biological Clocks/drug effects , Cyclic Nucleotide-Gated Cation Channels/physiology , Interneurons/drug effects , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Potassium Channels/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Analysis of Variance , Animals , Cardiovascular Agents/pharmacology , Colforsin/pharmacology , Dihydro-beta-Erythroidine , Dose-Response Relationship, Drug , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Female , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , In Vitro Techniques , Male , Mecamylamine/pharmacology , Mice , Mice, Transgenic , Models, Biological , Models, Molecular , Neural Inhibition/drug effects , Neural Inhibition/physiology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques/methods , Pyrimidines/pharmacology , Somatostatin/metabolism , gamma-Aminobutyric Acid/metabolism
11.
Elife ; 102021 10 26.
Article in English | MEDLINE | ID: mdl-34696824

ABSTRACT

Acetylcholine (ACh), released in the hippocampus from fibers originating in the medial septum/diagonal band of Broca (MSDB) complex, is crucial for learning and memory. The CA2 region of the hippocampus has received increasing attention in the context of social memory. However, the contribution of ACh to this process remains unclear. Here, we show that in mice, ACh controls social memory. Specifically, MSDB cholinergic neurons inhibition impairs social novelty discrimination, meaning the propensity of a mouse to interact with a novel rather than a familiar conspecific. This effect is mimicked by a selective antagonist of nicotinic AChRs delivered in CA2. Ex vivo recordings from hippocampal slices provide insight into the underlying mechanism, as activation of nAChRs by nicotine increases the excitatory drive to CA2 principal cells via disinhibition. In line with this observation, optogenetic activation of cholinergic neurons in MSDB increases the firing of CA2 principal cells in vivo. These results point to nAChRs as essential players in social novelty discrimination by controlling inhibition in the CA2 region.


Subject(s)
Antipsychotic Agents/pharmacology , CA2 Region, Hippocampal/physiology , Cholinergic Neurons/physiology , Clozapine/analogs & derivatives , Exploratory Behavior/drug effects , Receptors, Nicotinic/metabolism , Social Interaction/drug effects , Animals , CA2 Region, Hippocampal/drug effects , Clozapine/pharmacology , Diagonal Band of Broca/drug effects , Diagonal Band of Broca/metabolism , Male , Mice , Social Behavior
12.
Neuroscience ; 439: 125-136, 2020 07 15.
Article in English | MEDLINE | ID: mdl-31356900

ABSTRACT

To be highly reliable, synaptic transmission needs postsynaptic receptors (Rs) in precise apposition to the presynaptic release sites. At inhibitory synapses, the postsynaptic protein gephyrin self-assembles to form a scaffold that anchors glycine and GABAARs to the cytoskeleton, thus ensuring the accurate accumulation of postsynaptic receptors at the right place. This protein undergoes several post-translational modifications which control protein-protein interaction and downstream signaling pathways. In addition, through the constant exchange of scaffolding elements and receptors in and out of synapses, gephyrin dynamically regulates synaptic strength and plasticity. The aim of the present review is to highlight recent findings on the functional role of gephyrin at GABAergic inhibitory synapses. We will discuss different approaches used to interfere with gephyrin in order to unveil its function. In addition, we will focus on the impact of gephyrin structure and distribution at the nanoscale level on the functional properties of inhibitory synapses as well as the implications of this scaffold protein in synaptic plasticity processes. Finally, we will emphasize how gephyrin genetic mutations or alterations in protein expression levels are implicated in several neuropathological disorders, including autism spectrum disorders, schizophrenia, temporal lobe epilepsy and Alzheimer's disease, all associated with severe deficits of GABAergic signaling. This article is part of a Special Issue entitled: Honoring Ricardo Miledi - outstanding neuroscientist of XX-XXI centuries.


Subject(s)
Membrane Proteins , Synapses , Carrier Proteins , Humans , Membrane Proteins/metabolism , Receptors, GABA-A , Synapses/metabolism
13.
iScience ; 23(5): 101078, 2020 May 22.
Article in English | MEDLINE | ID: mdl-32361506

ABSTRACT

Early in brain development, impaired neuronal signaling during time-sensitive windows triggers the onset of neurodevelopmental disorders. GABA, through its depolarizing and excitatory actions, drives early developmental events including neuronal circuit formation and refinement. BDNF/TrkB signaling cooperates with GABA actions. How these developmental processes influence the formation of neural circuits and affect adult brain function is unknown. Here, we show that early deletion of Ntrk2/Trkb from immature mouse hippocampal dentate granule cells (DGCs) affects the integration and maturation of newly formed DGCs in the hippocampal circuitry and drives a premature shift from depolarizing to hyperpolarizing GABAergic actions in the target of DGCs, the CA3 principal cells of the hippocampus, by reducing the expression of the cation-chloride importer Nkcc1. These changes lead to the disruption of early synchronized neuronal activity at the network level and impaired morphological maturation of CA3 pyramidal neurons, ultimately contributing to altered adult hippocampal synaptic plasticity and cognitive processes.

14.
Eur J Neurosci ; 30(6): 1011-22, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19735287

ABSTRACT

A large variety of distinct locally connected GABAergic cells are present in the hippocampus. By releasing GABA into principal cells and interneurons, they exert a powerful control on neuronal excitability and are responsible for network oscillations crucial for information processing in the brain. Here, whole-cell patch clamp recordings in current and voltage clamp mode were used to study the functional role of nicotinic acetylcholine receptors (nAChRs) on the firing properties of stratum oriens interneurons in hippocampal slices from transgenic mice expressing enhanced green fluorescent protein in a subpopulation of GABAergic cells containing somatostatin (GIN mice). Unexpectedly, activation of nAChRs by nicotine or endogenously released acetylcholine strongly enhanced spike frequency adaptation. This effect was blocked by apamin, suggesting the involvement of small calcium-dependent potassium channels (SK channels). Nicotine-induced reduction in firing frequency was dependent on intracellular calcium rise through calcium-permeable nAChRs and voltage-dependent calcium channels activated by the depolarizing action of nicotine. Calcium imaging experiments directly showed that nicotine effects on firing rate were correlated with large increases in intracellular calcium. Furthermore, blocking ryanodine receptors with ryanodine or sarcoplasmic-endoplasmic reticulum calcium ATPase with thapsygargin or cyclopiazonic acid fully prevented the effects of nicotine, suggesting that mobilization of calcium from the internal stores contributed to the observed effects. By regulating cell firing, cholinergic signalling through nAChRs would be instrumental for fine-tuning the output of stratum oriens interneurons and correlated activity at the network level.


Subject(s)
Calcium/metabolism , Hippocampus/physiology , Interneurons/physiology , Receptors, Nicotinic/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Synaptic Transmission/physiology , Acetylcholine/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Fluorescent Dyes , Fura-2 , Green Fluorescent Proteins/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Interneurons/drug effects , Interneurons/metabolism , Mice , Mice, Transgenic , Nerve Net/drug effects , Nerve Net/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Nicotine/pharmacology , Patch-Clamp Techniques , Signal Processing, Computer-Assisted , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
15.
Front Psychiatry ; 10: 513, 2019.
Article in English | MEDLINE | ID: mdl-31379628

ABSTRACT

Autism spectrum disorders (ASDs) comprise a heterogeneous group of neuro-developmental abnormalities with a strong genetic component, characterized by deficits in verbal and non-verbal communication, impaired social interactions, and stereotyped behaviors. In a small percentage of cases, ASDs are associated with alterations of genes involved in synaptic function. Among these, relatively frequent are mutations/deletions of genes encoding for neuroligins (NLGs). NLGs are postsynaptic adhesion molecules that, interacting with their presynaptic partners neurexins, ensure the cross talk between pre- and postsynaptic specializations and synaptic stabilization, a condition needed for maintaining a proper excitatory/inhibitory balance within local neuronal circuits. We have focused on mice lacking NLG3 (NLG3 knock-out mice), animal models of a non-syndromic form of autism, which exhibit deficits in social behavior reminiscent of those found in ASDs. Among different brain areas involved in social cognition, the CA2 region of the hippocampus has recently emerged as a central structure for social memory processing. Here, in vivo recordings from anesthetized animals and ex vivo recordings from hippocampal slices have been used to assess the dynamics of neuronal signaling in the CA2 hippocampal area. In vivo experiments from NLG3-deficient mice revealed a selective impairment of spike-related slow wave activity in the CA2 area and a significant reduction in oscillatory activity in the theta and gamma frequencies range in both CA2 and CA3 regions of the hippocampus. These network effects were associated with an increased neuronal excitability in the CA2 hippocampal area. Ex vivo recordings from CA2 principal cells in slices obtained from NLG3 knock-out animals unveiled a strong excitatory/inhibitory imbalance in this region accompanied by a strong reduction of perisomatic inhibition mediated by CCK-containing GABAergic interneurons. These data clearly suggest that the selective alterations in network dynamics and GABAergic signaling observed in the CA2 hippocampal region of NLG3 knock-out mice may account for deficits in social memory reminiscent of those observed in autistic patients.

16.
Front Cell Neurosci ; 11: 255, 2017.
Article in English | MEDLINE | ID: mdl-28878628

ABSTRACT

Synchronized neuronal activity occurring at different developmental stages in various brain structures represents a hallmark of developmental circuits. This activity, which differs in its specific patterns among animal species may play a crucial role in de novo formation and in shaping neuronal networks. In the rodent hippocampus in vitro, the so-called giant depolarizing potentials (GDPs) constitute a primordial form of neuronal synchrony preceding more organized forms of activity such as oscillations in the theta and gamma frequency range. GDPs are generated at the network level by the interaction of the neurotransmitters glutamate and GABA which, immediately after birth, exert both a depolarizing and excitatory action on their targets. GDPs are triggered by GABAergic interneurons, which in virtue of their extensive axonal branching operate as functional hubs to synchronize large ensembles of cells. Intrinsic bursting activity, driven by a persistent sodium conductance and facilitated by the low expression of Kv7.2 and Kv7.3 channel subunits, responsible for IM, exerts a permissive role in GDP generation. Here, we discuss how GDPs are generated in a probabilistic way when neuronal excitability within a local circuit reaches a certain threshold and how GDP-associated calcium transients act as coincident detectors for enhancing synaptic strength at emerging GABAergic and glutamatergic synapses. We discuss the possible in vivo correlate of this activity. Finally, we debate recent data showing how, in several animal models of neuropsychiatric disorders including autism, a GDPs dysfunction is associated to morphological alterations of neuronal circuits and behavioral deficits reminiscent of those observed in patients.

17.
J Neuroimmunol ; 179(1-2): 76-86, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16870269

ABSTRACT

We have recently shown that de novo formation of lymphoid structures resembling B-cell follicles occurs in the inflamed central nervous system (CNS) meninges in a subset of patients with secondary progressive multiple sclerosis and in SJL mice with relapsing-remitting experimental autoimmune encephalomyelitis (EAE). Because lymphotoxin (LT) alpha(1)beta(2) is essential for lymphoid tissue organization, we used real-time PCR to examine LTbeta and LTbeta receptor (LTbetaR) gene expression in the CNS of SJL mice immunized with PLP 139-151 peptide. Moreover, we used the decoy receptor LTbetaR-immunoglobulin fusion protein to block the interaction of lymphotoxin (LT) alpha(1)beta(2) with the LTbeta receptor (LTbetaR) in mice with established EAE and evaluate the effect of systemic and local treatments with the fusion protein on disease progression, CNS lymphocytic infiltration and formation of meningeal B-cell follicles. The present findings indicate that both LTbeta and LTbetaR are upregulated at EAE onset and during subsequent relapses and that systemic and local blockade of the LT pathway with LTbetaR-Ig results in protracted and transient inhibition of EAE clinical signs, respectively. LTbetaR-Ig treatment also reduces T- and B-cell infiltration and prevents the induction of the chemokines CXCL10 and CXCL13 and the formation of organized ectopic follicles in the EAE-affected CNS. Targeting of molecules involved in lymphoid organogenesis could represent a valid strategy to inhibit CNS inflammation and formation of ectopic follicles, which may play a role in maintaining an abnormal, intrathecal humoral immune response in CNS autoimmune disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunoglobulin G/administration & dosage , Inflammation/immunology , Lymphotoxin beta Receptor/metabolism , Meninges/immunology , Recombinant Fusion Proteins/administration & dosage , Animals , B-Lymphocytes/immunology , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Chemokine CXCL10 , Chemokine CXCL13 , Chemokines, CXC/biosynthesis , Chemokines, CXC/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression , Gene Expression Profiling , Humans , Immunoglobulin G/immunology , Immunohistochemistry , Inflammation/pathology , Injections, Intraventricular , Lymphotoxin beta Receptor/genetics , Lymphotoxin-beta/antagonists & inhibitors , Lymphotoxin-beta/genetics , Mice , Microscopy, Confocal , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , RNA, Messenger/analysis , Recombinant Fusion Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
18.
Mol Neurobiol ; 43(2): 97-106, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21042953

ABSTRACT

Early in postnatal life γ-aminobutyric acid (GABA), the primary inhibitory transmitter in adults, excites targeted neurons by an outwardly directed flux of chloride which results from the unbalance between the cation-chloride cotransporters NKCC1 and KCC2, involved in chloride uptake and extrusion, respectively. This effect contributes to generate synchronized network activity or giant depolarizing potentials (GDPs) in the developing hippocampus. Here, we review some recent data concerning the mechanisms by which GDPs are generated and their functional role in enhancing synaptic efficacy at poorly developed GABAergic and glutamatergic synapses. In adulthood, reshaping neuronal circuits due to changes in chloride homeostasis and to the shift of GABA from hyperpolarizing to depolarizing, has been implicated in several neurological disorders, including epilepsy. Evidence has been recently provided that in chronically nerve growth factor-deprived mice expressing a progressive age-dependent neurodegenerative pathology resembling that observed in patients with Alzheimer's disease, the reduced expression of mRNA encoding for the Kcc2 gene and the depolarizing action of GABA lead to the reorganization of the neuronal hippocampal network. This may represent a novel mechanism by which GABAergic signaling counterbalances the loss of synaptic activity in neurodegenerative diseases.


Subject(s)
Action Potentials/physiology , Hippocampus/embryology , Hippocampus/physiology , Nerve Net/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Glutamic Acid/metabolism , Humans , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL