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











Publication year range
1.
Trends Neurosci ; 47(4): 239-240, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38514350

ABSTRACT

A recent study by Cheung, Pauler, Koppensteiner et al. combining lineage tracing with single-cell RNA sequencing (scRNA-seq) has revealed unexpected features of the developing superior colliculus (SC). Extremely multipotent individual progenitors generate all types of SC neurons and glial cells that were found to localize in a non-predetermined pattern, demonstrating a remarkable degree of unpredictability in SC development.


Subject(s)
Neurons , Superior Colliculi , Humans , Superior Colliculi/physiology , Neurons/physiology , Neuroglia , Neurogenesis
2.
PLoS Biol ; 21(11): e3002386, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37983249

ABSTRACT

Defensive responses to visually threatening stimuli represent an essential fear-related survival instinct, widely detected across species. The neural circuitry mediating visually triggered defensive responses has been delineated in the midbrain. However, the molecular mechanisms regulating the development and function of these circuits remain unresolved. Here, we show that midbrain-specific deletion of the transcription factor Brn3b causes a loss of neurons projecting to the lateral posterior nucleus of the thalamus. Brn3b deletion also down-regulates the expression of the neuropeptide tachykinin 2 (Tac2). Furthermore, Brn3b mutant mice display impaired defensive freezing responses to visual threat precipitated by social isolation. This behavioral phenotype could be ameliorated by overexpressing Tac2, suggesting that Tac2 acts downstream of Brn3b in regulating defensive responses to threat. Together, our experiments identify specific genetic components critical for the functional organization of midbrain fear-related visual circuits. Similar mechanisms may contribute to the development and function of additional long-range brain circuits underlying fear-associated behavior.


Subject(s)
Fear , Mesencephalon , Animals , Mice , Fear/physiology , Mesencephalon/physiology , Neurons/physiology , Thalamus
3.
Development ; 146(14)2019 07 22.
Article in English | MEDLINE | ID: mdl-31332038

ABSTRACT

Developmental control of long-range neuronal connections in the mammalian midbrain remains unclear. We explored the mechanisms regulating target selection of the developing superior colliculus (SC). The SC is a midbrain center that directs orienting behaviors and defense responses. We discovered that a transcription factor, Rorß, controls establishment of axonal projections from the SC to two thalamic nuclei: the dorsal lateral geniculate nucleus (dLGN) and the lateral posterior nucleus (LP). A genetic strategy used to visualize SC circuits revealed that in control animals Rorß+ neurons abundantly innervate the dLGN but barely innervate the LP. The opposite phenotype was observed in global and conditional Rorb mutants: projections to the dLGN were strongly decreased, and projections to the LP were increased. Furthermore, overexpression of Rorb in the wild type showed increased projections to the dLGN and decreased projections to the LP. In summary, we identified Rorß as a key developmental mediator of colliculo-thalamic innervation. Such regulation could represent a general mechanism orchestrating long-range neuronal connections in the mammalian brain.


Subject(s)
Axons/physiology , Mesencephalon/embryology , Mesencephalon/growth & development , Neurons/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 2/physiology , Visual Pathways/metabolism , Animals , Animals, Newborn , Embryo, Mammalian , Female , Geniculate Bodies/physiology , HEK293 Cells , Humans , Male , Mesencephalon/cytology , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 2/genetics , Nuclear Receptor Subfamily 1, Group F, Member 2/metabolism , Pregnancy , Superior Colliculi/physiology
4.
Nature ; 562(7725): E1, 2018 10.
Article in English | MEDLINE | ID: mdl-30046102

ABSTRACT

Change history: In this Article, Extended Data Fig. 8 and Extended Data Table 1 contained errors, which have been corrected online.

5.
Cell ; 169(6): 1029-1041.e16, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575667

ABSTRACT

We report a noninvasive strategy for electrically stimulating neurons at depth. By delivering to the brain multiple electric fields at frequencies too high to recruit neural firing, but which differ by a frequency within the dynamic range of neural firing, we can electrically stimulate neurons throughout a region where interference between the multiple fields results in a prominent electric field envelope modulated at the difference frequency. We validated this temporal interference (TI) concept via modeling and physics experiments, and verified that neurons in the living mouse brain could follow the electric field envelope. We demonstrate the utility of TI stimulation by stimulating neurons in the hippocampus of living mice without recruiting neurons of the overlying cortex. Finally, we show that by altering the currents delivered to a set of immobile electrodes, we can steerably evoke different motor patterns in living mice.


Subject(s)
Deep Brain Stimulation/methods , Transcranial Direct Current Stimulation/methods , Animals , Deep Brain Stimulation/adverse effects , Deep Brain Stimulation/instrumentation , Electrodes , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Transcranial Direct Current Stimulation/adverse effects , Transcranial Direct Current Stimulation/instrumentation
6.
Elife ; 52016 12 21.
Article in English | MEDLINE | ID: mdl-28001126

ABSTRACT

The SNARE-mediated vesicular transport pathway plays major roles in synaptic remodeling associated with formation of long-term memories, but the mechanisms that regulate this pathway during memory acquisition are not fully understood. Here we identify miRNAs that are up-regulated in the rodent hippocampus upon contextual fear-conditioning and identify the vesicular transport and synaptogenesis pathways as the major targets of the fear-induced miRNAs. We demonstrate that miR-153, a member of this group, inhibits the expression of key components of the vesicular transport machinery, and down-regulates Glutamate receptor A1 trafficking and neurotransmitter release. MiR-153 expression is specifically induced during LTP induction in hippocampal slices and its knockdown in the hippocampus of adult mice results in enhanced fear memory. Our results suggest that miR-153, and possibly other fear-induced miRNAs, act as components of a negative feedback loop that blocks neuronal hyperactivity at least partly through the inhibition of the vesicular transport pathway.


Subject(s)
Fear , Feedback, Physiological , Hippocampus/physiology , Memory , MicroRNAs/metabolism , Neurons/physiology , Synaptic Vesicles/metabolism , Animals , Mice , Neurotransmitter Agents/metabolism , Receptors, Glutamate/metabolism
7.
Nature ; 540(7632): 230-235, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27929004

ABSTRACT

Changes in gamma oscillations (20-50 Hz) have been observed in several neurological disorders. However, the relationship between gamma oscillations and cellular pathologies is unclear. Here we show reduced, behaviourally driven gamma oscillations before the onset of plaque formation or cognitive decline in a mouse model of Alzheimer's disease. Optogenetically driving fast-spiking parvalbumin-positive (FS-PV)-interneurons at gamma (40 Hz), but not other frequencies, reduces levels of amyloid-ß (Aß)1-40 and Aß 1-42 isoforms. Gene expression profiling revealed induction of genes associated with morphological transformation of microglia, and histological analysis confirmed increased microglia co-localization with Aß. Subsequently, we designed a non-invasive 40 Hz light-flickering regime that reduced Aß1-40 and Aß1-42 levels in the visual cortex of pre-depositing mice and mitigated plaque load in aged, depositing mice. Our findings uncover a previously unappreciated function of gamma rhythms in recruiting both neuronal and glial responses to attenuate Alzheimer's-disease-associated pathology.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Gamma Rhythm , Microglia/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/prevention & control , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Cell Shape/drug effects , Disease Models, Animal , Female , Gamma Rhythm/radiation effects , Interneurons/metabolism , Interneurons/radiation effects , Light , Male , Mice , Microglia/cytology , Microglia/radiation effects , Optogenetics , Parvalbumins/metabolism , Peptide Fragments/metabolism , Plaque, Amyloid/therapy , Transcriptome , Visual Cortex/physiology , Visual Cortex/radiation effects
8.
Nat Neurosci ; 19(9): 1190-1, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27571198
10.
BMC Biol ; 14: 40, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27197636

ABSTRACT

The mechanism of memory remains one of the great unsolved problems of biology. Grappling with the question more than a hundred years ago, the German zoologist Richard Semon formulated the concept of the engram, lasting connections in the brain that result from simultaneous "excitations", whose precise physical nature and consequences were out of reach of the biology of his day. Neuroscientists now have the knowledge and tools to tackle this question, however, and this Forum brings together leading contemporary views on the mechanisms of memory and what the engram means today.


Subject(s)
Brain/physiology , Memory/physiology , Animals , Epigenomics , Hippocampus/physiology , Humans , Models, Animal , Neurons/physiology , Spine/physiology , Synapses/physiology
11.
Proc Natl Acad Sci U S A ; 112(23): 7291-6, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25995364

ABSTRACT

Repeated stress has been suggested to underlie learning and memory deficits via the basolateral amygdala (BLA) and the hippocampus; however, the functional contribution of BLA inputs to the hippocampus and their molecular repercussions are not well understood. Here we show that repeated stress is accompanied by generation of the Cdk5 (cyclin-dependent kinase 5)-activator p25, up-regulation and phosphorylation of glucocorticoid receptors, increased HDAC2 expression, and reduced expression of memory-related genes in the hippocampus. A combination of optogenetic and pharmacosynthetic approaches shows that BLA activation is both necessary and sufficient for stress-associated molecular changes and memory impairments. Furthermore, we show that this effect relies on direct glutamatergic projections from the BLA to the dorsal hippocampus. Finally, we show that p25 generation is necessary for the stress-induced memory dysfunction. Taken together, our data provide a neural circuit model for stress-induced hippocampal memory deficits through BLA activity-dependent p25 generation.


Subject(s)
Basolateral Nuclear Complex/physiopathology , Cyclin-Dependent Kinase 5/metabolism , Hippocampus/physiopathology , Learning Disabilities/physiopathology , Memory Disorders/physiopathology , Animals , Basolateral Nuclear Complex/radiation effects , Hippocampus/radiation effects , Light , Mice , Stress, Physiological
12.
Nat Neurosci ; 18(7): 1008-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26005852

ABSTRACT

Noncoding variants in the human MIR137 gene locus increase schizophrenia risk with genome-wide significance. However, the functional consequence of these risk alleles is unknown. Here we examined induced human neurons harboring the minor alleles of four disease-associated single nucleotide polymorphisms in MIR137. We observed increased MIR137 levels compared to those in major allele-carrying cells. microRNA-137 gain of function caused downregulation of the presynaptic target genes complexin-1 (Cplx1), Nsf and synaptotagmin-1 (Syt1), leading to impaired vesicle release. In vivo, miR-137 gain of function resulted in changes in synaptic vesicle pool distribution, impaired induction of mossy fiber long-term potentiation and deficits in hippocampus-dependent learning and memory. By sequestering endogenous miR-137, we were able to ameliorate the synaptic phenotypes. Moreover, reinstatement of Syt1 expression partially restored synaptic plasticity, demonstrating the importance of Syt1 as a miR-137 target. Our data provide new insight into the mechanism by which miR-137 dysregulation can impair synaptic plasticity in the hippocampus.


Subject(s)
Gene Expression Regulation/genetics , MicroRNAs/metabolism , Mossy Fibers, Hippocampal/metabolism , Neuronal Plasticity/genetics , Schizophrenia/genetics , Synaptic Vesicles/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Alleles , Animals , Behavior, Animal/physiology , Disease Models, Animal , Fibroblasts , Genetic Loci , HEK293 Cells , Humans , Learning/physiology , Long-Term Potentiation , Mice , Mice, Inbred C57BL , N-Ethylmaleimide-Sensitive Proteins/metabolism , Nerve Tissue Proteins/metabolism , Polymorphism, Single Nucleotide , Synaptotagmin I/metabolism
13.
J Neurosci ; 35(6): 2372-83, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673832

ABSTRACT

Perturbations in fast-spiking parvalbumin (PV) interneurons are hypothesized to be a major component of various neuropsychiatric disorders; however, the mechanisms regulating PV interneurons remain mostly unknown. Recently, cyclin-dependent kinase 5 (Cdk5) has been shown to function as a major regulator of synaptic plasticity. Here, we demonstrate that genetic ablation of Cdk5 in PV interneurons in mouse brain leads to an increase in GABAergic neurotransmission and impaired synaptic plasticity. PVCre;fCdk5 mice display a range of behavioral abnormalities, including decreased anxiety and memory impairment. Our results reveal a central role of Cdk5 expressed in PV interneurons in gating inhibitory neurotransmission and underscore the importance of such regulation during behavioral tasks. Our findings suggest that Cdk5 can be considered a promising therapeutic target in a variety of conditions attributed to inhibitory interneuronal dysfunction, such as epilepsy, anxiety disorders, and schizophrenia.


Subject(s)
Anxiety/psychology , Cyclin-Dependent Kinase 5/genetics , Inhibition, Psychological , Interneurons/metabolism , Memory Disorders/psychology , Parvalbumins/metabolism , Animals , Anxiety/genetics , Behavior, Animal/physiology , Interneurons/enzymology , Maze Learning/physiology , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Synaptic Vesicles/ultrastructure , gamma-Aminobutyric Acid/metabolism
14.
Cell ; 157(2): 486-498, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725413

ABSTRACT

Cyclin-dependent kinase 5 regulates numerous neuronal functions with its activator, p35. Under neurotoxic conditions, p35 undergoes proteolytic cleavage to liberate p25, which has been implicated in various neurodegenerative diseases. Here, we show that p25 is generated following neuronal activity under physiological conditions in a GluN2B- and CaMKIIα-dependent manner. Moreover, we developed a knockin mouse model in which endogenous p35 is replaced with a calpain-resistant mutant p35 (Δp35KI) to prevent p25 generation. The Δp35KI mice exhibit impaired long-term depression and defective memory extinction, likely mediated through persistent GluA1 phosphorylation at Ser845. Finally, crossing the Δp35KI mice with the 5XFAD mouse model of Alzheimer's disease (AD) resulted in an amelioration of ß-amyloid (Aß)-induced synaptic depression and cognitive impairment. Together, these results reveal a physiological role of p25 production in synaptic plasticity and memory and provide new insights into the function of p25 in Aß-associated neurotoxicity and AD-like pathology.


Subject(s)
Alzheimer Disease/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Peptides/metabolism , Animals , Calpain/metabolism , Cell Cycle Proteins/metabolism , Cognition , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Endocytosis , Gene Knock-In Techniques , Hippocampus/metabolism , Humans , Long-Term Potentiation , Long-Term Synaptic Depression , Mice , Nerve Tissue Proteins/genetics , Phosphotransferases , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses
15.
Neuropharmacology ; 80: 70-82, 2014 May.
Article in English | MEDLINE | ID: mdl-24495398

ABSTRACT

Epigenetic regulation has been long considered to be a critical mechanism in the control of key aspects of cellular functions such as cell division, growth, and cell fate determination. Exciting recent developments have demonstrated that epigenetic mechanisms can also play necessary roles in the nervous system by regulating, for example, neuronal gene expression, DNA damage, and genome stability. Despite the fact that postmitotic neurons are developmentally less active then dividing cells, epigenetic regulation appears to provide means of both long-lasting and very dynamic regulation of neuronal function. Growing evidence indicates that epigenetic mechanisms in the central nervous system (CNS) are important for regulating not only specific aspects of individual neuronal metabolism but also for maintaining function of neuronal circuits and regulating their behavioral outputs. Multiple reports demonstrated that higher-level cognitive behaviors, such as learning and memory, are subject to a sophisticated epigenetic control, which includes interplay between multiple mechanisms of neuronal chromatin modification. Experiments with animal models have demonstrated that various epigenetic manipulations can affect cognition in different ways, from severe dysfunction to substantial improvement. In humans, epigenetic dysregulation has been known to underlie a number of disorders that are accompanied by mental impairment. Here, we review some of the epigenetic mechanisms that regulate cognition and how their disruption may contribute to cognitive dysfunctions. Due to the fact that histone acetylation and DNA methylation are some of the best-studied and critically important epigenomic modifications our research team has particularly strong expertise in, in this review, we are going to concentrate on histone acetylation, as well as DNA methylation/hydroxymethylation, in the mammalian CNS. Additional epigenetic modifications, not surveyed here, are being discussed in depth in the other review articles in this issue of Neuropharmacology.


Subject(s)
Central Nervous System/metabolism , Cognition Disorders/metabolism , Cognition , Epigenesis, Genetic , Models, Biological , Neurons/metabolism , 5-Methylcytosine/analogs & derivatives , Acetylation , Animals , Central Nervous System/enzymology , Cognition Disorders/enzymology , Cognition Disorders/etiology , Cytosine/analogs & derivatives , Cytosine/metabolism , DNA Methylation , Gene Expression Regulation , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Learning , Memory , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Protein Processing, Post-Translational , Rett Syndrome/enzymology , Rett Syndrome/metabolism , Rett Syndrome/physiopathology , Rubinstein-Taybi Syndrome/enzymology , Rubinstein-Taybi Syndrome/metabolism , Rubinstein-Taybi Syndrome/physiopathology
16.
Neuron ; 79(6): 1109-1122, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24050401

ABSTRACT

The ten-eleven translocation (Tet) family of methylcytosine dioxygenases catalyze oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) and promote DNA demethylation. Despite the abundance of 5hmC and Tet proteins in the brain, little is known about the functions of the neuronal Tet enzymes. Here, we analyzed Tet1 knockout mice (Tet1KO) and found downregulation of multiple neuronal activity-regulated genes, including Npas4, c-Fos, and Arc. Furthermore, Tet1KO animals exhibited abnormal hippocampal long-term depression and impaired memory extinction. Analysis of the key regulatory gene, Npas4, indicated that its promoter region, containing multiple CpG dinucleotides, is hypermethylated in both naive Tet1KO mice and after extinction training. Such hypermethylation may account for the diminished expression of Npas4 itself and its downstream targets, impairing transcriptional programs underlying cognitive processes. In summary, we show that neuronal Tet1 regulates normal DNA methylation levels, expression of activity-regulated genes, synaptic plasticity, and memory extinction.


Subject(s)
DNA-Binding Proteins/physiology , Extinction, Psychological/physiology , Gene Expression Regulation/genetics , Neurons/physiology , Proto-Oncogene Proteins/physiology , Age Factors , Analysis of Variance , Animals , Anxiety/genetics , Anxiety/physiopathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebral Cortex/cytology , Conditioning, Classical/physiology , DNA-Binding Proteins/deficiency , Depression/genetics , Depression/metabolism , Disease Models, Animal , Exploratory Behavior/physiology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hippocampus/cytology , In Vitro Techniques , Locomotion/genetics , Long-Term Synaptic Depression/genetics , Male , Maze Learning , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins c-fos/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/genetics , Synapsins/metabolism
17.
Neurobiol Learn Mem ; 105: 54-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23850563

ABSTRACT

Cyclin-dependent kinase 5 (Cdk5) is associated with synaptic plasticity and cognitive function. Previous reports have demonstrated that Cdk5 is necessary for memory formation, although others have reported Cdk5 conditional knockout mouse models exhibiting enhanced learning and memory. Furthermore, how Cdk5 acts in specific cell populations to affect behavior and cognitive outcomes remains unclear. Here we conduct a behavioral characterization of a forebrain-specific Cdk5 conditional knockout mouse model under the αCaMKII promoter, in which Cdk5 is ablated in excitatory pyramidal neurons of the forebrain. The Cdk5 conditional knockouts exhibit hyperactivity in the open field, reduced anxiety, and reduced behavioral despair. Moreover, the Cdk5 conditional knockouts also display impaired spatial learning in the Morris water maze and are severely impaired in contextual fear memory, which correspond to deficits in synaptic transmission. Remarkably, the hyperactivity of the Cdk5 conditional knockouts can be ameliorated by the administration of lithium chloride, an inhibitor of GSK3ß signaling. Collectively, our data reveal that Cdk5 ablation from forebrain excitatory neurons results in deleterious effects on emotional and cognitive behavior and highlight a key role for Cdk5 in regulating the GSK3ß signaling pathway.


Subject(s)
Cognition , Cyclin-Dependent Kinase 5/metabolism , Hyperkinesis/metabolism , Prosencephalon/metabolism , Pyramidal Cells/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Neuron ; 75(4): 675-87, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22920258

ABSTRACT

N-type voltage-gated calcium channels localize to presynaptic nerve terminals and mediate key events including synaptogenesis and neurotransmission. While several kinases have been implicated in the modulation of calcium channels, their impact on presynaptic functions remains unclear. Here we report that the N-type calcium channel is a substrate for cyclin-dependent kinase 5 (Cdk5). The pore-forming α(1) subunit of the N-type calcium channel is phosphorylated in the C-terminal domain, and phosphorylation results in enhanced calcium influx due to increased channel open probability. Phosphorylation of the N-type calcium channel by Cdk5 facilitates neurotransmitter release and alters presynaptic plasticity by increasing the number of docked vesicles at the synaptic cleft. These effects are mediated by an altered interaction between N-type calcium channels and RIM1, which tethers presynaptic calcium channels to the active zone. Collectively, our results highlight a molecular mechanism by which N-type calcium channels are regulated by Cdk5 to affect presynaptic function.


Subject(s)
Calcium Channels, N-Type/physiology , Cyclin-Dependent Kinase 5/metabolism , Ion Channel Gating/physiology , Neurons/cytology , Phosphotransferases/metabolism , Presynaptic Terminals/physiology , Amiodarone , Analysis of Variance , Animals , Biophysics , Biotinylation , Calcium Channels, N-Type/genetics , Cells, Cultured , Cerebral Cortex/cytology , Cyclin-Dependent Kinase 5/genetics , Electric Stimulation , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Humans , Immunoprecipitation , In Vitro Techniques , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Models, Molecular , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques , Phosphorylation , Phosphotransferases/genetics , Presynaptic Terminals/drug effects , Presynaptic Terminals/ultrastructure , Radioimmunoassay , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL