Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Publication year range
1.
Mol Psychiatry ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609585

ABSTRACT

The hippocampus is crucial for acquiring and retrieving episodic and contextual memories. In previous studies, the inactivation of dentate gyrus (DG) neurons by chemogenetic- and optogenetic-mediated hyperpolarization led to opposing conclusions about DG's role in memory retrieval. One study used Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-mediated clozapine N-oxide (CNO)-induced hyperpolarization and reported that the previously formed memory was erased, thus concluding that denate gyrus is needed for memory maintenance. The other study used optogenetic with halorhodopsin induced hyperpolarization and reported and dentate gyrus is needed for memory retrieval. We hypothesized that this apparent discrepancy could be due to the length of hyperpolarization in previous studies; minutes by optogenetics and several hours by DREADD/CNO. Since hyperpolarization interferes with anterograde and retrograde neuronal signaling, it is possible that the memory engram in the dentate gyrus and the entorhinal to hippocampus trisynaptic circuit was erased by long-term, but not with short-term hyperpolarization. We developed and applied an advanced chemogenetic technology to selectively silence synaptic output by blocking neurotransmitter release without hyperpolarizing DG neurons to explore this apparent discrepancy. We performed in vivo electrophysiology during trace eyeblink in a rabbit model of associative learning. Our work shows that the DG output is required for memory retrieval. Based on previous and recent findings, we propose that the actively functional anterograde and retrograde neuronal signaling is necessary to preserve synaptic memory engrams along the entorhinal cortex to the hippocampal trisynaptic circuit.

2.
Hippocampus ; 34(3): 126-140, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38140716

ABSTRACT

The hippocampus has been implicated in temporal learning. Plasticity within the hippocampus requires NMDA receptor-dependent glutamatergic neurotransmission. We tested the prediction that hippocampal NMDA receptors are required for learning about time by testing mice that lack postembryonal NMDARs in the CA1 and dentate gyrus (DG) hippocampal subfields on three different appetitive temporal learning procedures. The conditional knockout mice (Grin1ΔDCA1 ) showed normal sensitivity to cue duration, responding at a higher level to a short duration cue than compared to a long duration cue. Knockout mice also showed normal precision and accuracy of response timing in the peak procedure in which reinforcement occurred after 10 s delay within a 30 s cue presentation. Mice were tested on the matching of response rates to reinforcement rates on instrumental conditioning with two levers reinforced on a concurrent variable interval schedule. Pressing on one lever was reinforced at a higher rate than the other lever. Grin1ΔDGCA1 mice showed normal sensitivity to the relative reinforcement rates of the levers. In contrast to the lack of effect of hippocampal NMDAR deletion on measures of temporal sensitivity, Grin1ΔDGCA1 mice showed increased baseline measures of magazine activity and lever pressing. Furthermore, reversal learning was enhanced when the reward contingencies were switched in the lever pressing task, but this was true only for mice trained with a large difference between relative reinforcement rates between the levers. The results failed to demonstrate a role for NMDARs in excitatory CA1 and DG neurons in learning about temporal information.


Subject(s)
Learning , Receptors, N-Methyl-D-Aspartate , Mice , Animals , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Mice, Knockout , Learning/physiology , Hippocampus/physiology , Dentate Gyrus/metabolism
3.
Mol Psychiatry ; 28(2): 579-587, 2023 02.
Article in English | MEDLINE | ID: mdl-36460723

ABSTRACT

Psychosis in disorders like schizophrenia is commonly associated with aberrant salience and elevated striatal dopamine. However, the underlying cause(s) of this hyper-dopaminergic state remain elusive. Various lines of evidence point to glutamatergic dysfunction and impairments in synaptic plasticity in the etiology of schizophrenia, including deficits associated with the GluA1 AMPAR subunit. GluA1 knockout (Gria1-/-) mice provide a model of impaired synaptic plasticity in schizophrenia and exhibit a selective deficit in a form of short-term memory which underlies short-term habituation. As such, these mice are unable to reduce attention to recently presented stimuli. In this study we used fast-scan cyclic voltammetry to measure phasic dopamine responses in the nucleus accumbens of Gria1-/- mice to determine whether this behavioral phenotype might be a key driver of a hyper-dopaminergic state. There was no effect of GluA1 deletion on electrically-evoked dopamine responses in anaesthetized mice, demonstrating normal endogenous release properties of dopamine neurons in Gria1-/- mice. Furthermore, dopamine signals were initially similar in Gria1-/- mice compared to controls in response to both sucrose rewards and neutral light stimuli. They were also equally sensitive to changes in the magnitude of delivered rewards. In contrast, however, these stimulus-evoked dopamine signals failed to habituate with repeated presentations in Gria1-/- mice, resulting in a task-relevant, hyper-dopaminergic phenotype. Thus, here we show that GluA1 dysfunction, resulting in impaired short-term habituation, is a key driver of enhanced striatal dopamine responses, which may be an important contributor to aberrant salience and psychosis in psychiatric disorders like schizophrenia.


Subject(s)
Dopamine , Habituation, Psychophysiologic , Mice , Animals , Mice, Knockout , Memory, Short-Term , Phenotype
4.
Glia ; 71(12): 2770-2781, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37564028

ABSTRACT

Astrocytes are intricately involved in the activity of neural circuits; however, their basic physiology of interacting with nearby neurons is not well established. Using two-photon imaging of neurons and astrocytes during higher frequency stimulation of hippocampal CA3-CA1 Schaffer collateral (Scc) excitatory synapses, we could show that increasing levels of released glutamate accelerated local astrocytic Ca2+ elevation. However, blockage of glutamate transporters did not abolish this astrocytic Ca2+ response, suggesting that astrocytic Ca2+ elevation is indirectly associated with an uptake of extracellular glutamate. However, during the astrocytic glutamate uptake, the Na+ /Ca2+ exchanger (NCX) reverse mode was activated, and mediated extracellular Ca2+ entry, thereby triggering the internal release of Ca2+ . In addition, extracellular Ca2+ entry via membrane P2X receptors further facilitated astrocytic Ca2+ elevation via ATP binding. These findings suggest a novel mechanism of activity induced Ca2+ permeability increases of astrocytic membranes, which drives astrocytic responses during neuronal stimulation of CA3-CA1 Scc excitatory synapses.


Subject(s)
Astrocytes , Neurons , Astrocytes/metabolism , Neurons/metabolism , Hippocampus/metabolism , Synapses/metabolism , Glutamic Acid/metabolism , Permeability , Calcium/metabolism
5.
Mol Psychiatry ; 26(11): 6482-6504, 2021 11.
Article in English | MEDLINE | ID: mdl-34021263

ABSTRACT

Mutations in SHANK genes play an undisputed role in neuropsychiatric disorders. Until now, research has focused on the postsynaptic function of SHANKs, and prominent postsynaptic alterations in glutamatergic signal transmission have been reported in Shank KO mouse models. Recent studies have also suggested a possible presynaptic function of SHANK proteins, but these remain poorly defined. In this study, we examined how SHANK2 can mediate electrophysiological, molecular, and behavioral effects by conditionally overexpressing either wild-type SHANK2A or the extrasynaptic SHANK2A(R462X) variant. SHANK2A overexpression affected pre- and postsynaptic targets and revealed a reversible, development-dependent autism spectrum disorder-like behavior. SHANK2A also mediated redistribution of Ca2+-permeable AMPA receptors between apical and basal hippocampal CA1 dendrites, leading to impaired synaptic plasticity in the basal dendrites. Moreover, SHANK2A overexpression reduced social interaction and increased the excitatory noise in the olfactory cortex during odor processing. In contrast, overexpression of the extrasynaptic SHANK2A(R462X) variant did not impair hippocampal synaptic plasticity, but still altered the expression of presynaptic/axonal signaling proteins. We also observed an attention-deficit/hyperactivity-like behavior and improved social interaction along with enhanced signal-to-noise ratio in cortical odor processing. Our results suggest that the disruption of pre- and postsynaptic SHANK2 functions caused by SHANK2 mutations has a strong impact on social behavior. These findings indicate that pre- and postsynaptic SHANK2 actions cooperate for normal neuronal function, and that an imbalance between these functions may lead to different neuropsychiatric disorders.


Subject(s)
Autism Spectrum Disorder , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Hippocampus/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Social Behavior
6.
EMBO J ; 36(18): 2770-2789, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28790178

ABSTRACT

Canonical transient receptor potential (TRPC) channels influence various neuronal functions. Using quantitative high-resolution mass spectrometry, we demonstrate that TRPC1, TRPC4, and TRPC5 assemble into heteromultimers with each other, but not with other TRP family members in the mouse brain and hippocampus. In hippocampal neurons from Trpc1/Trpc4/Trpc5-triple-knockout (Trpc1/4/5-/-) mice, lacking any TRPC1-, TRPC4-, or TRPC5-containing channels, action potential-triggered excitatory postsynaptic currents (EPSCs) were significantly reduced, whereas frequency, amplitude, and kinetics of quantal miniature EPSC signaling remained unchanged. Likewise, evoked postsynaptic responses in hippocampal slice recordings and transient potentiation after tetanic stimulation were decreased. In vivo, Trpc1/4/5-/- mice displayed impaired cross-frequency coupling in hippocampal networks and deficits in spatial working memory, while spatial reference memory was unaltered. Trpc1/4/5-/- animals also exhibited deficiencies in adapting to a new challenge in a relearning task. Our results indicate the contribution of heteromultimeric channels from TRPC1, TRPC4, and TRPC5 subunits to the regulation of mechanisms underlying spatial working memory and flexible relearning by facilitating proper synaptic transmission in hippocampal neurons.


Subject(s)
Hippocampus/physiology , Memory, Short-Term , Protein Multimerization , Synaptic Transmission , TRPC Cation Channels/metabolism , Animals , Gene Knockout Techniques , Hippocampus/metabolism , Mass Spectrometry , Mice , Mice, Knockout , TRPC Cation Channels/genetics
7.
Psychol Sci ; 32(2): 204-217, 2021 02.
Article in English | MEDLINE | ID: mdl-33395376

ABSTRACT

Theories of learning differ in whether they assume that learning reflects the strength of an association between memories or symbolic encoding of the statistical properties of events. We provide novel evidence for symbolic encoding of informational variables by demonstrating that sensitivity to time and number in learning is dissociable. Whereas responding in normal mice was dependent on reinforcement rate, responding in mice that lacked the GluA1 AMPA receptor subunit was insensitive to reinforcement rate and, instead, dependent on the number of times a cue had been paired with reinforcement. This suggests that GluA1 is necessary for weighting numeric information by temporal information in order to calculate reinforcement rate. Sample sizes per genotype varied between seven and 23 across six experiments and consisted of both male and female mice. The results provide evidence for explicit encoding of variables by animals rather than implicit encoding via variations in associative strength.


Subject(s)
Learning , Receptors, AMPA , Animals , Female , Male , Mice , Mice, Knockout , Receptors, AMPA/genetics , Reinforcement, Psychology
8.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374598

ABSTRACT

The correlation between dysfunction in the glutamatergic system and neuropsychiatric disorders, including schizophrenia and autism spectrum disorder, is undisputed. Both disorders are associated with molecular and ultrastructural alterations that affect synaptic plasticity and thus the molecular and physiological basis of learning and memory. Altered synaptic plasticity, accompanied by changes in protein synthesis and trafficking of postsynaptic proteins, as well as structural modifications of excitatory synapses, are critically involved in the postnatal development of the mammalian nervous system. In this review, we summarize glutamatergic alterations and ultrastructural changes in synapses in schizophrenia and autism spectrum disorder of genetic or drug-related origin, and briefly comment on the possible reversibility of these neuropsychiatric disorders in the light of findings in regular synaptic physiology.


Subject(s)
Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/metabolism , Glutamic Acid/metabolism , Receptors, Glutamate/metabolism , Schizophrenia/etiology , Schizophrenia/metabolism , Synapses/metabolism , Synapses/ultrastructure , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Humans , Mitochondria/metabolism , Neuronal Plasticity , Neurons/metabolism , Rodentia , Synapses/pathology
9.
Nat Rev Neurosci ; 15(3): 181-92, 2014 03.
Article in English | MEDLINE | ID: mdl-24552786

ABSTRACT

Recent studies using transgenic mice lacking NMDA receptors in the hippocampus challenge the long-standing hypothesis that hippocampal long-term potentiation-like mechanisms underlie the encoding and storage of associative long-term spatial memories. However, it may not be the synaptic plasticity-dependent memory hypothesis that is wrong; instead, it may be the role of the hippocampus that needs to be re-examined. We present an account of hippocampal function that explains its role in both memory and anxiety.


Subject(s)
Anxiety/physiopathology , Hippocampus/physiology , Memory/physiology , Neuronal Plasticity/physiology , Space Perception/physiology , Synapses/physiology , Animals , Behavior, Animal/physiology , Hippocampus/physiopathology , Mice , Mice, Knockout , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology
10.
Cereb Cortex ; 28(11): 4036-4048, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30169757

ABSTRACT

Epileptic seizures are associated with increased astrocytic Ca2+ signaling, but the fine spatiotemporal kinetics of the ictal astrocyte-neuron interplay remains elusive. By using 2-photon imaging of awake head-fixed mice with chronic hippocampal windows we demonstrate that astrocytic Ca2+ signals precede neuronal Ca2+ elevations during the initial bout of kainate-induced seizures. On average, astrocytic Ca2+ elevations preceded neuronal activity in CA1 by about 8 s. In subsequent bouts of epileptic seizures, astrocytes and neurons were activated simultaneously. The initial astrocytic Ca2+ elevation was abolished in mice lacking the type 2 inositol-1,4,5-trisphosphate-receptor (Itpr2-/-). Furthermore, we found that Itpr2-/- mice exhibited 60% less epileptiform activity compared with wild-type mice when assessed by telemetric EEG monitoring. In both genotypes we also demonstrate that spreading depression waves may play a part in seizure termination. Our findings imply a role for astrocytic Ca2+ signals in ictogenesis.


Subject(s)
Astrocytes/physiology , Calcium Signaling , Epilepsy/physiopathology , Hippocampus/physiopathology , Neurons/physiology , Seizures/physiopathology , Animals , Epilepsy/chemically induced , Excitatory Amino Acid Agonists/administration & dosage , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/physiology , Kainic Acid/administration & dosage , Male , Mice, Inbred C57BL , Mice, Knockout , Seizures/chemically induced
11.
Cereb Cortex ; 27(1): 24-33, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28365776

ABSTRACT

Cortical spreading depression (CSD) is a phenomenon that challenges the homeostatic mechanisms on which normal brain function so critically depends. Analyzing the sequence of events in CSD holds the potential of providing new insight in the physiological processes underlying normal brain function as well as the pathophysiology of neurological conditions characterized by ionic dyshomeostasis. Here, we have studied the sequential progression of CSD in awake wild-type mice and in mice lacking aquaporin-4 (AQP4) or inositol 1,4,5-triphosphate type 2 receptor (IP3R2). By the use of a novel combination of genetically encoded sensors that a novel combination - an unprecedented temporal and spatial resolution, we show that CSD leads to brisk Ca2+ signals in astrocytes and that the duration of these Ca2+ signals is shortened in the absence of AQP4 but not in the absence of IP3R2. The decrease of the astrocytic, AQP4-dependent Ca2+ signals, coincides in time and space with a decrease in the duration of extracellular glutamate overflow but not with the initial peak of the glutamate release suggesting that in CSD, extracellular glutamate accumulation is extended through AQP4-dependent glutamate release from astrocytes. The present data point to a salient glial contribution to CSD and identify AQP4 as a new target for therapy.


Subject(s)
Aquaporin 4/metabolism , Astrocytes/physiology , Cortical Spreading Depression/physiology , Extracellular Fluid/metabolism , Glutamic Acid/metabolism , Wakefulness/physiology , Animals , Aquaporin 4/genetics , Calcium Signaling/physiology , Down-Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
Eur J Neurosci ; 45(7): 912-921, 2017 04.
Article in English | MEDLINE | ID: mdl-28186680

ABSTRACT

Group II metabotropic glutamate receptor agonists have been suggested as potential anti-psychotics, at least in part, based on the observation that the agonist LY354740 appeared to rescue the cognitive deficits caused by non-competitive N-methyl-d-aspartate receptor (NMDAR) antagonists, including spatial working memory deficits in rodents. Here, we tested the ability of LY354740 to rescue spatial working memory performance in mice that lack the GluA1 subunit of the AMPA glutamate receptor, encoded by Gria1, a gene recently implicated in schizophrenia by genome-wide association studies. We found that LY354740 failed to rescue the spatial working memory deficit in Gria1-/- mice during rewarded alternation performance in the T-maze. In contrast, LY354740 did reduce the locomotor hyperactivity in these animals to a level that was similar to controls. A similar pattern was found with the dopamine receptor antagonist haloperidol, with no amelioration of the spatial working memory deficit in Gria1-/- mice, even though the same dose of haloperidol reduced their locomotor hyperactivity. These results with LY354740 contrast with the rescue of spatial working memory in models of glutamatergic hypofunction using non-competitive NMDAR antagonists. Future studies should determine whether group II mGluR agonists can rescue spatial working memory deficits with other NMDAR manipulations, including genetic models and other pharmacological manipulations of NMDAR function.


Subject(s)
Bridged Bicyclo Compounds/pharmacology , Dopamine Antagonists/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Haloperidol/pharmacology , Hyperkinesis/metabolism , Memory, Short-Term/drug effects , Receptors, AMPA/genetics , Animals , Bridged Bicyclo Compounds/therapeutic use , Dopamine Antagonists/therapeutic use , Excitatory Amino Acid Agonists/therapeutic use , Female , Haloperidol/therapeutic use , Hyperkinesis/drug therapy , Hyperkinesis/physiopathology , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism
14.
J Neurosci ; 35(7): 3016-21, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25698739

ABSTRACT

To date, it has been difficult to reveal physiological Ca(2+) events occurring within the fine astrocytic processes of mature animals. The objective of the study was to explore whether neuronal activity evokes astrocytic Ca(2+) signals at glutamatergic synapses of adult mice. We stimulated the Schaffer collateral/commissural fibers in acute hippocampal slices from adult mice transduced with the genetically encoded Ca(2+) indicator GCaMP5E driven by the glial fibrillary acidic protein promoter. Two-photon imaging revealed global stimulation-evoked astrocytic Ca(2+) signals with distinct latencies, rise rates, and amplitudes in fine processes and somata. Specifically, the Ca(2+) signals in the processes were faster and of higher amplitude than those in the somata. A combination of P2 purinergic and group I/II metabotropic glutamate receptor (mGluR) antagonists reduced the amplitude of the Ca(2+) transients by 30-40% in both astrocytic compartments. Blockage of the mGluRs alone only modestly reduced the magnitude of the stimulation-evoked Ca(2+) signals in processes and failed to affect the somatic Ca(2+) response. Local application of group I or I/II mGluR agonists or adenosine triphosphate (ATP) elicited global astrocytic Ca(2+) signals that mimicked the stimulation-evoked astrocytic Ca(2+) responses. We conclude that stimulation-evoked Ca(2+) signals in astrocytic processes at CA3-CA1 synapses of adult mice (1) differ from those in astrocytic somata and (2) are modulated by glutamate and ATP.


Subject(s)
Adenosine Triphosphate/pharmacology , Astrocytes/metabolism , Calcium Signaling/drug effects , Glutamic Acid/pharmacology , Hippocampus/cytology , Synapses/drug effects , Animals , Astrocytes/drug effects , Calcium/metabolism , Calcium Signaling/physiology , Calmodulin/genetics , Calmodulin/metabolism , Dioxolanes/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenylacetates/pharmacology , Purines/pharmacology , Synapses/physiology , Synapsins/genetics , Synapsins/metabolism , Time Factors
15.
Neurobiol Learn Mem ; 135: 83-90, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27417577

ABSTRACT

Spatial working memory (SWM) is an essential cognitive function important for survival in a competitive environment. In rodents SWM requires an intact hippocampus and SWM expression is impaired in mice lacking the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 (Gria1-/- mice). Here we used viral gene transfer to show that re-expression of GluA1 in the hippocampus can affect the behavioral performance of GluA1 deficient mice. We found that Gria1-/- mice with hippocampus-specific rescue of GluA1 expression (Gria1Hpc mice) are more anxious, less hyperactive and only partly impaired in SWM expression in the Y-maze spatial novelty preference paradigm compared to Gria1-/- mice. However, Gria1Hpc mice still express SWM performance deficits when tested in the rewarded alternation T-maze task. Thus, the restoration of hippocampal function affects several behaviors of GluA1 deficient mice - including SWM expression - in different tasks. The virus-mediated GluA1 expression in Gria1-/- mice is not sufficient for a comprehensive SWM restoration, suggesting that both hippocampal as well as extra-hippocampal GluA1-containing AMPA receptors contribute to SWM.


Subject(s)
Hippocampus/metabolism , Memory Disorders/metabolism , Memory Disorders/physiopathology , Memory, Short-Term/physiology , Receptors, AMPA/metabolism , Spatial Memory/physiology , Animals , Behavior, Animal/physiology , Gene Transfer Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, AMPA/deficiency
16.
Cell Mol Neurobiol ; 36(7): 1215-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26645823

ABSTRACT

The recently identified Cystine-knot containing AMPAR-associated protein (Ckamp44) represents a novel AMPAR-related protein that critically controls AMPAR-mediated currents and short-term plasticity. However, the effects of the lack of this protein at network level are not entirely understood. Here we used c-Fos brain mapping to analyse whether the excitatory/inhibitory balance is altered in the absence of the Ckamp44. We found that Ckamp44(-/-) mice treated with an NMDAR antagonist exhibited a very robust c-Fos expression pattern, similar with that seen in mice lacking the GluN2A subunit of NMDAR treated with the same compound. This finding is unexpected, in particular, since Ckamp44 expression is strongest in dentate gyrus granule cells and less abundant in the rest of the brain.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Genes, fos/physiology , Mice , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology
17.
Nature ; 468(7321): 277-82, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21068837

ABSTRACT

The central amygdala (CEA), a nucleus predominantly composed of GABAergic inhibitory neurons, is essential for fear conditioning. How the acquisition and expression of conditioned fear are encoded within CEA inhibitory circuits is not understood. Using in vivo electrophysiological, optogenetic and pharmacological approaches in mice, we show that neuronal activity in the lateral subdivision of the central amygdala (CEl) is required for fear acquisition, whereas conditioned fear responses are driven by output neurons in the medial subdivision (CEm). Functional circuit analysis revealed that inhibitory CEA microcircuits are highly organized and that cell-type-specific plasticity of phasic and tonic activity in the CEl to CEm pathway may gate fear expression and regulate fear generalization. Our results define the functional architecture of CEA microcircuits and their role in the acquisition and regulation of conditioned fear behaviour.


Subject(s)
Amygdala/physiology , Conditioning, Classical/physiology , Fear/physiology , Neural Inhibition/physiology , Neural Pathways/physiology , Action Potentials , Amygdala/anatomy & histology , Amygdala/cytology , Animals , Freezing Reaction, Cataleptic , Male , Mice , Mice, Inbred C57BL , Neural Pathways/cytology , Neuronal Plasticity/physiology , Neurons/physiology , gamma-Aminobutyric Acid/metabolism
18.
Cereb Cortex ; 25(11): 4469-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25840424

ABSTRACT

Cortical spreading depression is a slowly propagating wave of near-complete depolarization of brain cells followed by temporary suppression of neuronal activity. Accumulating evidence indicates that cortical spreading depression underlies the migraine aura and that similar waves promote tissue damage in stroke, trauma, and hemorrhage. Cortical spreading depression is characterized by neuronal swelling, profound elevation of extracellular potassium and glutamate, multiphasic blood flow changes, and drop in tissue oxygen tension. The slow speed of the cortical spreading depression wave implies that it is mediated by diffusion of a chemical substance, yet the identity of this substance and the pathway it follows are unknown. Intercellular spread between gap junction-coupled neurons or glial cells and interstitial diffusion of K(+) or glutamate have been proposed. Here we use extracellular direct current potential recordings, K(+)-sensitive microelectrodes, and 2-photon imaging with ultrasensitive Ca(2+) and glutamate fluorescent probes to elucidate the spatiotemporal dynamics of ionic shifts associated with the propagation of cortical spreading depression in the visual cortex of adult living mice. Our data argue against intercellular spread of Ca(2+) carrying the cortical spreading depression wavefront and are in favor of interstitial K(+) diffusion, rather than glutamate diffusion, as the leading event in cortical spreading depression.


Subject(s)
Cerebral Cortex/physiology , Cortical Spreading Depression/physiology , Ions/metabolism , Neurons/physiology , Nonlinear Dynamics , Analysis of Variance , Animals , Cortical Spreading Depression/drug effects , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Patch-Clamp Techniques , Potassium/pharmacology , Synapsins/genetics , Synapsins/metabolism , Transduction, Genetic
19.
Hippocampus ; 24(4): 424-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24339333

ABSTRACT

Adolescence is characterized by important molecular and anatomical changes with relevance for the maturation of brain circuitry and cognitive function. This time period is of critical importance in the emergence of several neuropsychiatric disorders accompanied by cognitive impairment, such as affective disorders and schizophrenia. The molecular mechanisms underlying these changes at neuronal level during this specific developmental stage remains however poorly understood. GluA1-containing AMPA receptors, which are located predominantly on hippocampal neurons, are the primary molecular determinants of synaptic plasticity. We investigated here the consequences of the inducible deletion of GluA1 AMPA receptors in glutamatergic neurons during late adolescence. We generated mutant mice with a tamoxifen-inducible deletion of GluA1 under the control of the CamKII promoter for temporally and spatially restricted gene manipulation. GluA1 ablation during late adolescence induced cognitive impairments, but also marked hyperlocomotion and sensorimotor gating deficits. Unlike the global genetic deletion of GluA1, inducible GluA1 ablation during late adolescence resulted in normal sociability. Deletion of GluA1 induced redistribution of GluA2 subunits, suggesting AMPA receptor trafficking deficits. Mutant animals showed increased hippocampal NMDA receptor expression and no change in striatal dopamine concentration. Our data provide new insight into the role of deficient AMPA receptors specifically during late adolescence in inducing several cognitive and behavioral alterations with possible relevance for neuropsychiatric disorders.


Subject(s)
Cognition Disorders/metabolism , Corpus Striatum/metabolism , Hippocampus/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Social Behavior , Animals , Corpus Striatum/growth & development , Dopamine/metabolism , Hippocampus/growth & development , Maze Learning/physiology , Memory, Short-Term , Mental Disorders , Mice , Mice, Transgenic , Motor Activity/physiology , Phenotype , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Sensory Gating/physiology
20.
Hum Mol Genet ; 21(2): 344-57, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21994763

ABSTRACT

Mutations in the postsynaptic scaffolding gene SHANK2 have recently been identified in individuals with autism spectrum disorder (ASD) and intellectual disability. However, the cellular and physiological consequences of these mutations in neurons remain unknown. We have analyzed the functional impact caused by two inherited and one de novo SHANK2 mutations from ASD individuals (L1008_P1009dup, T1127M, R462X). Although all three variants affect spine volume and have smaller SHANK2 cluster sizes, T1127M additionally fails to rescue spine volume in Shank2 knock-down neurons. R462X is not able to rescue spine volume and dendritic branching and lacks postsynaptic clustering, indicating the most severe dysfunction. To demonstrate that R462X when expressed in mouse can be linked to physiological effects, we analyzed synaptic transmission and behavior. Principal neurons of mice expressing rAAV-transduced SHANK2-R462X present a specific, long-lasting reduction in miniature postsynaptic AMPA receptor currents. This dominant negative effect translates into dose-dependent altered cognitive behavior of SHANK2-R462X-expressing mice, with an impact on the penetrance of ASD.


Subject(s)
Child Development Disorders, Pervasive/genetics , Morphogenesis , Nerve Tissue Proteins/genetics , Neurons/cytology , Animals , Child , Child Development Disorders, Pervasive/pathology , Gene Knockdown Techniques , Hippocampus/pathology , Humans , Mice , Mutation , Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL