Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
eNeuro ; 7(4)2020.
Article in English | MEDLINE | ID: mdl-32601096

ABSTRACT

Both the basal amygdala (BA) and the bed nucleus of the stria terminalis (BNST) can participate in contextual fear, but it is unclear whether contextual fear engrams involve a direct interaction between these two brain regions. To determine whether dorsal BNST (dBNST)-projecting neurons in the BA participate in contextual fear engrams, we combined the TetTag mouse with a retrograde tracer to label dBNST-projecting cells in the BA. We identified a population of neurons located in the anterior subdivision of the BA (aBA) that was activated during fear conditioning and reactivated during retrieval but that did not project to the dBNST. In contrast, dBNST-projecting neurons located in the posterior BA (pBA) were activated during contextual fear conditioning but were not reactivated during retrieval. Similarly, we found neurons in the oval BNST subdivision (ovBNST) that were activated during contextual fear conditioning without being reactivated during retrieval. However, the anterodorsal BNST (adBNST) subdivision was not activated during either contextual fear conditioning or retrieval, underscoring the divergent functionality of these two dBNST subdivisions. Finally, we found that the ovBNST receives a monosynaptic projection from neurons located in the BA. Our results indicate that aBA neurons that do not project to the dBNST participate in contextual fear engrams. In contrast, dBNST-projecting neurons in the BA do not appear to participate in contextual fear engrams, but might instead contain a BA → ovBNST pathway that is active during the initial encoding of contextual fear memories.


Subject(s)
Basolateral Nuclear Complex , Septal Nuclei , Animals , Fear , Memory , Mice , Neurons
2.
Brain Res Bull ; 141: 44-49, 2018 07.
Article in English | MEDLINE | ID: mdl-29269319

ABSTRACT

Great progress has been made in our understanding of how so-called memory engrams in the brain enable the storage and retrieval of memories. This has led to the realization that across the lifetime of an animal, the spatial and temporal properties of a memory engram are not fixed, but instead are subjected to dynamic modifications that can be both dependent and independent on additional experiences. The dynamic nature of engrams is especially relevant in the case of fear memories, whose contributions to an animal's evolutionary fitness depend on a delicate balance of stability and flexibility. Though fear memories have the potential to last a lifetime, their expression also needs to be properly tuned to prevent maladaptive behavior, such as seen in patients with post-traumatic stress disorder. To achieve this balance, fear engrams are subjected to complex spatiotemporal dynamics, making them informative examples of the "dynamic engram". In this review, we discuss the current understanding of the dynamic nature of fear engrams in the basolateral amygdala, a brain region that plays a central role in fear memory encoding and expression. We propose that this understanding can be further advanced by studying how fast dynamics, such as oscillatory circuit activity, support the storage and retrieval of fear engrams that can be stable over long time intervals.


Subject(s)
Basolateral Nuclear Complex/physiology , Fear/physiology , Memory/physiology , Animals , Basolateral Nuclear Complex/physiopathology , Humans , Models, Neurological , Models, Psychological
3.
Nat Neurosci ; 20(11): 1624-1633, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28967909

ABSTRACT

The mammalian brain contains dedicated circuits for both the learned expression and suppression of fear. These circuits require precise coordination to facilitate the appropriate expression of fear behavior, but the mechanisms underlying this coordination remain unclear. Using a combination of chemogenetics, activity-based neuronal-ensemble labeling and in vivo electrophysiology, we found that fear extinction learning confers on parvalbumin-expressing (PV) interneurons in the basolateral amygdala (BLA) a dedicated role in the selective suppression of a previously encoded fear memory and BLA fear-encoding neurons. In addition, following extinction learning, PV interneurons enable a competing interaction between a 6-12 Hz oscillation and a fear-associated 3-6 Hz oscillation within the BLA. Loss of this competition increases a 3-6 Hz oscillatory signature, with BLA→medial prefrontal cortex directionality signaling the recurrence of fear expression. The discovery of cellular and oscillatory substrates of fear extinction learning that critically depend on BLA PV interneurons could inform therapies aimed at preventing the pathological recurrence of fear following extinction learning.


Subject(s)
Basolateral Nuclear Complex/physiology , Biological Clocks/physiology , Extinction, Psychological/physiology , Fear/physiology , Learning/physiology , Prefrontal Cortex/physiology , Animals , Basolateral Nuclear Complex/cytology , Fear/psychology , Female , Male , Mice , Mice, 129 Strain , Mice, Transgenic , Prefrontal Cortex/cytology
4.
Neuron ; 96(2): 248-249, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29024648

ABSTRACT

The brain processes information and generates behavior by employing a wide array of different cell types. In this issue of Neuron, Wu et al. (2017) report a novel method that enables the efficient identification of molecularly defined cells that participate in a specific brain function.


Subject(s)
Brain Mapping , Neurons , Brain , Humans , Nervous System Physiological Phenomena
5.
Nat Neurosci ; 19(4): 564-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26900924

ABSTRACT

The hippocampus provides the brain's memory system with a subset of neurons holding a map-like representation of each environment experienced. We found in mice that optogenetic silencing those neurons active in an environment unmasked a subset of quiet neurons, enabling the emergence of an alternative map. When applied in a cocaine-paired environment, this intervention neutralized an otherwise long-lasting drug-place preference, showing that recoding a spatial memory engram can alleviate associated maladaptive behavior.


Subject(s)
Cocaine/administration & dosage , Conditioning, Psychological/physiology , Hippocampus/physiology , Memory/physiology , Optogenetics/methods , Action Potentials/drug effects , Action Potentials/physiology , Animals , Conditioning, Psychological/drug effects , Hippocampus/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Front Mol Neurosci ; 8: 56, 2015.
Article in English | MEDLINE | ID: mdl-26557053

ABSTRACT

The brain contains a large variety of projection neurons with different functional properties. The functional properties of projection neurons arise from their connectivity with other neurons and their molecular composition. We describe a novel tool for obtaining the gene expression profiles of projection neurons that are anatomically defined by the location of their soma and axon terminals. Our tool utilizes adeno-associated virus serotype 9 (AAV9), which we found to retrogradely transduce projection neurons after injection at the site of the axon terminals. We used AAV9 to express Enhanced Green Fluorescent Protein (EGFP)-tagged ribosomal protein L10a (EGFP-L10a), which enables the immunoprecipitation of EGFP-tagged ribosomes and associated mRNA with a method known as Translating Ribosome Affinity Purification (TRAP). To achieve high expression of the EGFP-L10a protein in projection neurons, we placed its expression under control of a 1.3 kb alpha-calcium/calmodulin-dependent protein kinase II (Camk2a) promoter. We injected the AAV9-Camk2a-TRAP virus in either the hippocampus or the bed nucleus of the stria terminalis (BNST) of the mouse brain. In both brain regions the 1.3 kb Camk2a promoter did not confer complete cell-type specificity around the site of injection, as EGFP-L10a expression was observed in Camk2a-expressing neurons as well as in neuronal and non-neuronal cells that did not express Camk2a. In contrast, cell-type specific expression was observed in Camk2a-positive projection neurons that were retrogradely transduced by AAV9-Camk2a-TRAP. Injection of AAV9-Camk2a-TRAP into the BNST enabled the use of TRAP to collect ribosome-bound mRNA from basal amygdala projection neurons that innervate the BNST. AAV9-Camk2a-TRAP provides a single-virus system that can be used for the molecular profiling of anatomically defined projection neurons in mice and other mammalian model organisms. In addition, AAV9-Camk2a-TRAP may enable the discovery of protein synthesis events that support information storage in projection neurons.

7.
Front Mol Neurosci ; 7: 82, 2014.
Article in English | MEDLINE | ID: mdl-25400545

ABSTRACT

Acquiring the gene expression profiles of specific neuronal cell-types is important for understanding their molecular identities. Genome-wide gene expression profiles of genetically defined cell-types can be acquired by collecting and sequencing mRNA that is bound to epitope-tagged ribosomes (TRAP; translating ribosome affinity purification). Here, we introduce a transgenic mouse model that combines the TRAP technique with the tetracycline transactivator (tTA) system by expressing EGFP-tagged ribosomal protein L10a (EGFP-L10a) under control of the tetracycline response element (tetO-TRAP). This allows both spatial control of EGFP-L10a expression through cell-type specific tTA expression, as well as temporal regulation by inhibiting transgene expression through the administration of doxycycline. We show that crossing tetO-TRAP mice with transgenic mice expressing tTA under the Camk2a promoter (Camk2a-tTA) results in offspring with cell-type specific expression of EGFP-L10a in CA1 pyramidal neurons and medium spiny neurons in the striatum. Co-immunoprecipitation confirmed that EGFP-L10a integrates into a functional ribosomal complex. In addition, collection of ribosome-bound mRNA from the hippocampus yielded the expected enrichment of genes expressed in CA1 pyramidal neurons, as well as a depletion of genes expressed in other hippocampal cell-types. Finally, we show that crossing tetO-TRAP mice with transgenic Fos-tTA mice enables the expression of EGFP-L10a in CA1 pyramidal neurons that are activated during a fear conditioning trial. The tetO-TRAP mouse can be combined with other tTA mouse lines to enable gene expression profiling of a variety of different cell-types.

8.
Nat Commun ; 5: 4510, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25072471

ABSTRACT

The subcellular localization and translation of messenger RNA (mRNA) supports functional differentiation between cellular compartments. In neuronal dendrites, local translation of mRNA provides a rapid and specific mechanism for synaptic plasticity and memory formation, and might be involved in the pathophysiology of certain brain disorders. Despite the importance of dendritic mRNA translation, little is known about which mRNAs can be translated in dendrites in vivo and when their translation occurs. Here we collect ribosome-bound mRNA from the dendrites of CA1 pyramidal neurons in the adult mouse hippocampus. We find that dendritic mRNA rapidly associates with ribosomes following a novel experience consisting of a contextual fear conditioning trial. High throughput RNA sequencing followed by machine learning classification reveals an unexpected breadth of ribosome-bound dendritic mRNAs, including mRNAs expected to be entirely somatic. Our findings are in agreement with a mechanism of synaptic plasticity that engages the acute local translation of functionally diverse dendritic mRNAs.


Subject(s)
Dendrites/physiology , Models, Neurological , Neuronal Plasticity/physiology , Protein Biosynthesis/physiology , Pyramidal Cells/physiology , RNA, Messenger/metabolism , Ribosomes/metabolism , Animals , Base Sequence , Conditioning, Psychological , Dendrites/metabolism , Fear , High-Throughput Nucleotide Sequencing , Hippocampus/cytology , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment
9.
PLoS Biol ; 11(11): e1001703, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24348200

ABSTRACT

Genome-wide studies of circadian transcription or mRNA translation have been hindered by the presence of heterogeneous cell populations in complex tissues such as the nervous system. We describe here the use of a Drosophila cell-specific translational profiling approach to document the rhythmic "translatome" of neural clock cells for the first time in any organism. Unexpectedly, translation of most clock-regulated transcripts--as assayed by mRNA ribosome association--occurs at one of two predominant circadian phases, midday or mid-night, times of behavioral quiescence; mRNAs encoding similar cellular functions are translated at the same time of day. Our analysis also indicates that fundamental cellular processes--metabolism, energy production, redox state (e.g., the thioredoxin system), cell growth, signaling and others--are rhythmically modulated within clock cells via synchronized protein synthesis. Our approach is validated by the identification of mRNAs known to exhibit circadian changes in abundance and the discovery of hundreds of novel mRNAs that show translational rhythms. This includes Tdc2, encoding a neurotransmitter synthetic enzyme, which we demonstrate is required within clock neurons for normal circadian locomotor activity.


Subject(s)
CLOCK Proteins/physiology , Circadian Rhythm , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Protein Biosynthesis , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Gene Expression Regulation , Motor Activity , NADP/metabolism , Nervous System/cytology , Nervous System/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Proteome/genetics , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome , Tyrosine Decarboxylase/genetics , Tyrosine Decarboxylase/metabolism
10.
Neuron ; 80(4): 1054-65, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24183705

ABSTRACT

A more complete understanding of how fear extinction alters neuronal activity and connectivity within fear circuits may aid in the development of strategies to treat human fear disorders. Using a c-fos-based transgenic mouse, we found that contextual fear extinction silenced basal amygdala (BA) excitatory neurons that had been previously activated during fear conditioning. We hypothesized that the silencing of BA fear neurons was caused by an action of extinction on BA inhibitory synapses. In support of this hypothesis, we found extinction-induced target-specific remodeling of BA perisomatic inhibitory synapses originating from parvalbumin and cholecystokinin-positive interneurons. Interestingly, the predicted changes in the balance of perisomatic inhibition matched the silent and active states of the target BA fear neurons. These observations suggest that target-specific changes in perisomatic inhibitory synapses represent a mechanism through which experience can sculpt the activation patterns within a neural circuit.


Subject(s)
Extinction, Psychological/physiology , Fear/psychology , Synapses/physiology , Amygdala/physiology , Animals , Behavior, Animal/physiology , Cholecystokinin/metabolism , Electroshock , Image Processing, Computer-Assisted , Immunohistochemistry , Interneurons/physiology , Learning/physiology , Limbic System/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Neural Pathways/physiology , Neurons/physiology , Parvalbumins/metabolism , Proto-Oncogene Proteins c-fos/physiology , Receptor, Cannabinoid, CB1/metabolism
11.
Curr Biol ; 23(2): 99-106, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-23246402

ABSTRACT

BACKGROUND: Episodic memories are encoded within hippocampal and neocortical circuits. Retrieving these memories is assumed to involve reactivation of neural ensembles that were established during learning. Although it has been possible to follow the activity of individual neurons shortly after learning, it has not been possible to examine their activity weeks later during retrieval. We addressed this issue by using a stable form of GFP (H2B-GFP) to permanently tag neurons that are active during contextual fear conditioning. RESULTS: H2B-GFP expression in transgenic mice was increased by learning and could be regulated by doxycycline (DOX). Using this system, we found a large network of neurons in the hippocampus, amygdala, and neocortex that were active during context fear conditioning and subsequent memory retrieval 2 days later. Reactivation was contingent on memory retrieval and was not observed when animals were trained and tested in different environments. When memory was retrieved several weeks after learning, reactivation was altered in the hippocampus and amygdala but remained unchanged in the cortex. CONCLUSIONS: Retrieving a recently formed context fear memory reactivates neurons in the hippocampus, amygdala, and cortex. Several weeks after learning, the degree of reactivation is altered in hippocampal and amygdala networks but remains stable in the cortex.


Subject(s)
Amygdala/physiology , Hippocampus/physiology , Mental Recall/physiology , Neocortex/physiology , Animals , Conditioning, Psychological , Female , Green Fluorescent Proteins , Male , Mice , Mice, Transgenic , Nerve Net/physiology
12.
PLoS One ; 7(1): e29940, 2012.
Article in English | MEDLINE | ID: mdl-22238679

ABSTRACT

Memory loss is one of the hallmark symptoms of Alzheimer's disease (AD). It has been proposed that soluble amyloid-beta (Abeta) oligomers acutely impair neuronal function and thereby memory. We here report that natural Abeta oligomers acutely impair contextual fear memory in mice. A natural Abeta oligomer solution containing Abeta monomers, dimers, trimers, and tetramers was derived from the conditioned medium of 7PA2 cells, a cell line that expresses human amyloid precursor protein containing the Val717Phe familial AD mutation. As a control we used 7PA2 conditioned medium from which Abeta oligomers were removed through immunodepletion. Separate groups of mice were injected with Abeta and control solutions through a cannula into the lateral brain ventricle, and subjected to fear conditioning using two tone-shock pairings. One day after fear conditioning, mice were tested for contextual fear memory and tone fear memory in separate retrieval trials. Three experiments were performed. For experiment 1, mice were injected three times: 1 hour before and 3 hours after fear conditioning, and 1 hour before context retrieval. For experiments 2 and 3, mice were injected a single time at 1 hour and 2 hours before fear conditioning respectively. In all three experiments there was no effect on tone fear memory. Injection of Abeta 1 hour before fear conditioning, but not 2 hours before fear conditioning, impaired the formation of a contextual fear memory. In future studies, the acute effect of natural Abeta oligomers on contextual fear memory can be used to identify potential mechanisms and treatments of AD associated memory loss.


Subject(s)
Amyloid beta-Peptides/pharmacology , Avoidance Learning/drug effects , Fear/drug effects , Fear/psychology , Memory/drug effects , Algorithms , Amyloid beta-Peptides/metabolism , Animals , Avoidance Learning/physiology , CHO Cells , Cricetinae , Cricetulus , Fear/physiology , Male , Memory/physiology , Memory Disorders/chemically induced , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Physical Conditioning, Animal , Protein Multimerization/physiology , Recognition, Psychology/drug effects , Recognition, Psychology/physiology
13.
Science ; 317(5842): 1230-3, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17761885

ABSTRACT

Do learning and retrieval of a memory activate the same neurons? Does the number of reactivated neurons correlate with memory strength? We developed a transgenic mouse that enables the long-lasting genetic tagging of c-fos-active neurons. We found neurons in the basolateral amygdala that are activated during Pavlovian fear conditioning and are reactivated during memory retrieval. The number of reactivated neurons correlated positively with the behavioral expression of the fear memory, indicating a stable neural correlate of associative memory. The ability to manipulate these neurons genetically should allow a more precise dissection of the molecular mechanisms of memory encoding within a distributed neuronal network.


Subject(s)
Amygdala/physiology , Memory , Mental Recall , Nerve Net/physiology , Neurons/physiology , Amygdala/cytology , Animals , Brain Mapping , Conditioning, Psychological , Extinction, Psychological , Fear , Genes, fos , Learning , Mice , Mice, Transgenic , Neuronal Plasticity
14.
Learn Mem ; 13(2): 143-9, 2006.
Article in English | MEDLINE | ID: mdl-16585790

ABSTRACT

Targeted mutagenesis in mice has shown that genes from a wide variety of gene families are involved in memory formation. The efficient identification of genes involved in learning and memory could be achieved by random mutagenesis combined with high-throughput phenotyping. Here, we provide the first report of a mutagenesis screen that has generated memory mutants in the mouse. We tested a group of N-ethyl-N-nitrosourea (ENU) mutagenized mice in the conditioned fear paradigm. We screened for both dominant and recessive mutations that caused impairments in contextual or tone fear conditioning. Heritability testing confirmed three fear conditioning mutants, i.e., Forgetful, Slowlearner, and Scatterbrain. All three have a learning or short-term memory deficit in contextual fear conditioning. Forgetful was further characterized and showed a highly specific phenotype. The contextual fear-conditioning deficit was apparent when Forgetful was trained with tone-shock pairings, but not when trained with shock alone. The deficit was not due to changes in shock sensitivity or anxiety. Forgetful was not impaired in two other memory tests (hidden platform version of Morris water maze and object recognition). Our data show that a mutagenesis screen can generate mutant mice with highly specific memory phenotypes that can supplement existing mice with targeted mutations. Mapping of Slowlearner found linkage to a region of chromosome 12 (LOD score of 6.5 close to D12Mit171), which suggests that ENU mutants should enable the positional cloning of genes involved in memory formation.


Subject(s)
Behavior, Animal/physiology , Conditioning, Classical/physiology , Fear/physiology , Genetic Testing , Mice, Mutant Strains , Animals , Environment , Female , Genetics, Behavioral/methods , Male , Memory/physiology , Memory Disorders/genetics , Methylnitrosourea , Mice , Mice, Inbred C57BL , Mutagenesis , Phenotype , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...