Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2635, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38528004

ABSTRACT

High levels of proinflammatory cytokines induce neurotoxicity and catalyze inflammation-driven neurodegeneration, but the specific release mechanisms from microglia remain elusive. Here we show that secretory autophagy (SA), a non-lytic modality of autophagy for secretion of vesicular cargo, regulates neuroinflammation-mediated neurodegeneration via SKA2 and FKBP5 signaling. SKA2 inhibits SA-dependent IL-1ß release by counteracting FKBP5 function. Hippocampal Ska2 knockdown in male mice hyperactivates SA resulting in neuroinflammation, subsequent neurodegeneration and complete hippocampal atrophy within six weeks. The hyperactivation of SA increases IL-1ß release, contributing to an inflammatory feed-forward vicious cycle including NLRP3-inflammasome activation and Gasdermin D-mediated neurotoxicity, which ultimately drives neurodegeneration. Results from protein expression and co-immunoprecipitation analyses of male and female postmortem human brains demonstrate that SA is hyperactivated in Alzheimer's disease. Overall, our findings suggest that SKA2-regulated, hyperactive SA facilitates neuroinflammation and is linked to Alzheimer's disease, providing mechanistic insight into the biology of neuroinflammation.


Subject(s)
Alzheimer Disease , Autophagy , Chromosomal Proteins, Non-Histone , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Animals , Female , Humans , Male , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Autophagy/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cytokines/metabolism , Inflammasomes/metabolism , Microglia/metabolism , Neuroinflammatory Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
2.
Nat Commun ; 15(1): 878, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38296993

ABSTRACT

In brain, the striatum is a heterogenous region involved in reward and goal-directed behaviors. Striatal dysfunction is linked to psychiatric disorders, including opioid use disorder (OUD). Striatal subregions are divided based on neuroanatomy, each with unique roles in OUD. In OUD, the dorsal striatum is involved in altered reward processing, formation of habits, and development of negative affect during withdrawal. Using single nuclei RNA-sequencing, we identified both canonical (e.g., dopamine receptor subtype) and less abundant cell populations (e.g., interneurons) in human dorsal striatum. Pathways related to neurodegeneration, interferon response, and DNA damage were significantly enriched in striatal neurons of individuals with OUD. DNA damage markers were also elevated in striatal neurons of opioid-exposed rhesus macaques. Sex-specific molecular differences in glial cell subtypes associated with chronic stress were found in OUD, particularly female individuals. Together, we describe different cell types in human dorsal striatum and identify cell type-specific alterations in OUD.


Subject(s)
Corpus Striatum , Opioid-Related Disorders , Male , Animals , Humans , Female , Macaca mulatta , Corpus Striatum/metabolism , Neurons/metabolism , Opioid-Related Disorders/genetics , Opioid-Related Disorders/metabolism , Gene Expression Profiling
3.
Am J Psychiatry ; 180(10): 739-754, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37491937

ABSTRACT

OBJECTIVE: Multidisciplinary studies of posttraumatic stress disorder (PTSD) and major depressive disorder (MDD) implicate the dorsolateral prefrontal cortex (DLPFC) in disease risk and pathophysiology. Postmortem brain studies have relied on bulk-tissue RNA sequencing (RNA-seq), but single-cell RNA-seq is needed to dissect cell-type-specific mechanisms. The authors conducted the first single-nucleus RNA-seq postmortem brain study in PTSD to elucidate disease transcriptomic pathology with cell-type-specific resolution. METHOD: Profiling of 32 DLPFC samples from 11 individuals with PTSD, 10 with MDD, and 11 control subjects was conducted (∼415K nuclei; >13K cells per sample). A replication sample included 15 DLPFC samples (∼160K nuclei; >11K cells per sample). RESULTS: Differential gene expression analyses identified significant single-nucleus RNA-seq differentially expressed genes (snDEGs) in excitatory (EX) and inhibitory (IN) neurons and astrocytes, but not in other cell types or bulk tissue. MDD samples had more false discovery rate-corrected significant snDEGs, and PTSD samples had a greater replication rate. In EX and IN neurons, biological pathways that were differentially enriched in PTSD compared with MDD included glucocorticoid signaling. Furthermore, glucocorticoid signaling in induced pluripotent stem cell (iPSC)-derived cortical neurons demonstrated greater relevance in PTSD and opposite direction of regulation compared with MDD, especially in EX neurons. Many snDEGs were from the 17q21.31 locus and are particularly interesting given causal roles in disease pathogenesis and DLPFC-based neuroimaging (PTSD: ARL17B, LINC02210-CRHR1, and LRRC37A2; MDD: LRRC37A and LRP4), while others were regulated by glucocorticoids in iPSC-derived neurons (PTSD: SLC16A6, TAF1C; MDD: CDH3). CONCLUSIONS: The study findings point to cell-type-specific mechanisms of brain stress response in PTSD and MDD, highlighting the importance of examining cell-type-specific gene expression and indicating promising novel biomarkers and therapeutic targets.


Subject(s)
Depressive Disorder, Major , Stress Disorders, Post-Traumatic , Humans , Dorsolateral Prefrontal Cortex , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Stress Disorders, Post-Traumatic/genetics , Glucocorticoids/metabolism , Gene Expression Profiling , Transcriptome/genetics , Neurons/metabolism , Prefrontal Cortex/metabolism
4.
bioRxiv ; 2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37066393

ABSTRACT

High levels of proinflammatory cytokines induce neurotoxicity and catalyze inflammation-driven neurodegeneration, but the specific release mechanisms from microglia remain elusive. We demonstrate that secretory autophagy (SA), a non-lytic modality of autophagy for secretion of vesicular cargo, regulates neuroinflammation-mediated neurodegeneration via SKA2 and FKBP5 signaling. SKA2 inhibits SA-dependent IL-1ß release by counteracting FKBP5 function. Hippocampal Ska2 knockdown in mice hyperactivates SA resulting in neuroinflammation, subsequent neurodegeneration and complete hippocampal atrophy within six weeks. The hyperactivation of SA increases IL-1ß release, initiating an inflammatory feed-forward vicious cycle including NLRP3-inflammasome activation and Gasdermin D (GSDMD)-mediated neurotoxicity, which ultimately drives neurodegeneration. Results from protein expression and co-immunoprecipitation analyses of postmortem brains demonstrate that SA is hyperactivated in Alzheimer's disease. Overall, our findings suggest that SKA2-regulated, hyperactive SA facilitates neuroinflammation and is linked to Alzheimer's disease, providing new mechanistic insight into the biology of neuroinflammation.

5.
bioRxiv ; 2023 Mar 24.
Article in English | MEDLINE | ID: mdl-36993188

ABSTRACT

Stress produces profound effects on behavior, including persistent alterations in sleep patterns. Here we examined the effects of two prototypical stress peptides, pituitary adenylate cyclase-activating polypeptide (PACAP) and corticotropin-releasing factor (CRF), on sleep architecture and other translationally-relevant endpoints. Male and female mice were implanted with subcutaneous transmitters enabling continuous measurement of electroencephalography (EEG) and electromyography (EMG), as well as body temperature and locomotor activity, without tethering that restricts free movement, body posture, or head orientation during sleep. At baseline, females spent more time awake (AW) and less time in slow wave sleep (SWS) than males. Mice then received intracerebral infusions of PACAP or CRF at doses producing equivalent increases in anxiety-like behavior. The effects of PACAP on sleep architecture were similar in both sexes and resembled those reported in male mice after chronic stress exposure. Compared to vehicle infusions, PACAP infusions decreased time in AW, increased time in SWS, and increased rapid eye movement sleep (REM) time and bouts on the day following treatment. In addition, PACAP effects on REM time remained detectable a week after treatment. PACAP infusions also reduced body temperature and locomotor activity. Under the same experimental conditions, CRF infusions had minimal effects on sleep architecture in either sex, causing only transient increases in SWS during the dark phase, with no effects on temperature or activity. These findings suggest that PACAP and CRF have fundamentally different effects on sleep-related metrics, and provide new insights into the mechanisms by which stress disrupts sleep.

6.
Transl Psychiatry ; 12(1): 423, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36192377

ABSTRACT

Exposure to stress triggers biological changes throughout the body. Accumulating evidence indicates that alterations in immune system function are associated with the development of stress-associated illnesses such as major depressive disorder and post-traumatic stress disorder, increasing interest in identifying immune markers that provide insight into mental health. Recombination events during T-cell receptor rearrangement and T-cell maturation in the thymus produce circular DNA fragments called T-cell receptor excision circles (TRECs) that can be utilized as indicators of thymic function and numbers of newly emigrating T-cells. Given data suggesting that stress affects thymus function, we examined whether blood levels of TRECs might serve as a quantitative peripheral index of cumulative stress exposure and its physiological correlates. We hypothesized that chronic stress exposure would compromise thymus function and produce corresponding decreases in levels of TRECs. In male mice, exposure to chronic social defeat stress (CSDS) produced thymic involution, adrenal hypertrophy, and decreased levels of TRECs in blood. Extending these studies to humans revealed robust inverse correlations between levels of circulating TRECs and childhood emotional and physical abuse. Cell-type specific analyses also revealed associations between TREC levels and blood cell composition, as well as cell-type specific methylation changes in CD4T + and CD8T + cells. Additionally, TREC levels correlated with epigenetic age acceleration, a common biomarker of stress exposure. Our findings demonstrate alignment between findings in mice and humans and suggest that blood-borne TRECs are a translationally-relevant biomarker that correlates with, and provides insight into, the cumulative physiological and immune-related impacts of stress exposure in mammals.


Subject(s)
Depressive Disorder, Major , Receptors, Antigen, T-Cell , Animals , Biomarkers/analysis , Child , DNA, Circular , Depressive Disorder, Major/genetics , Humans , Male , Mammals/genetics , Mice , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes
7.
Nature ; 608(7923): 586-592, 2022 08.
Article in English | MEDLINE | ID: mdl-35859170

ABSTRACT

The ability to associate temporally segregated information and assign positive or negative valence to environmental cues is paramount for survival. Studies have shown that different projections from the basolateral amygdala (BLA) are potentiated following reward or punishment learning1-7. However, we do not yet understand how valence-specific information is routed to the BLA neurons with the appropriate downstream projections, nor do we understand how to reconcile the sub-second timescales of synaptic plasticity8-11 with the longer timescales separating the predictive cues from their outcomes. Here we demonstrate that neurotensin (NT)-expressing neurons in the paraventricular nucleus of the thalamus (PVT) projecting to the BLA (PVT-BLA:NT) mediate valence assignment by exerting NT concentration-dependent modulation in BLA during associative learning. We found that optogenetic activation of the PVT-BLA:NT projection promotes reward learning, whereas PVT-BLA projection-specific knockout of the NT gene (Nts) augments punishment learning. Using genetically encoded calcium and NT sensors, we further revealed that both calcium dynamics within the PVT-BLA:NT projection and NT concentrations in the BLA are enhanced after reward learning and reduced after punishment learning. Finally, we showed that CRISPR-mediated knockout of the Nts gene in the PVT-BLA pathway blunts BLA neural dynamics and attenuates the preference for active behavioural strategies to reward and punishment predictive cues. In sum, we have identified NT as a neuropeptide that signals valence in the BLA, and showed that NT is a critical neuromodulator that orchestrates positive and negative valence assignment in amygdala neurons by extending valence-specific plasticity to behaviourally relevant timescales.


Subject(s)
Basolateral Nuclear Complex , Learning , Neural Pathways , Neurotensin , Punishment , Reward , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/physiology , Calcium/metabolism , Cues , Neuronal Plasticity , Neurotensin/metabolism , Optogenetics , Thalamic Nuclei/cytology , Thalamic Nuclei/physiology
8.
Cell Rep ; 35(9): 109185, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34077736

ABSTRACT

Responding to different dynamic levels of stress is critical for mammalian survival. Disruption of mineralocorticoid receptor (MR) and glucocorticoid receptor (GR) signaling is proposed to underlie hypothalamic-pituitary-adrenal (HPA) axis dysregulation observed in stress-related psychiatric disorders. In this study, we show that FK506-binding protein 51 (FKBP5) plays a critical role in fine-tuning MR:GR balance in the hippocampus. Biotinylated-oligonucleotide immunoprecipitation in primary hippocampal neurons reveals that MR binding, rather than GR binding, to the Fkbp5 gene regulates FKBP5 expression during baseline activity of glucocorticoids. Notably, FKBP5 and MR exhibit similar hippocampal expression patterns in mice and humans, which are distinct from that of the GR. Pharmacological inhibition and region- and cell type-specific receptor deletion in mice further demonstrate that lack of MR decreases hippocampal Fkbp5 levels and dampens the stress-induced increase in glucocorticoid levels. Overall, our findings demonstrate that MR-dependent changes in baseline Fkbp5 expression modify GR sensitivity to glucocorticoids, providing insight into mechanisms of stress homeostasis.


Subject(s)
Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Stress, Physiological , Tacrolimus Binding Proteins/metabolism , Animals , Cells, Cultured , Gene Deletion , Gene Expression Regulation , Hippocampus/metabolism , Humans , Male , Mice, Inbred C57BL , Models, Biological , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Tacrolimus Binding Proteins/genetics
9.
Biol Psychiatry ; 89(12): 1138-1149, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33715826

ABSTRACT

BACKGROUND: Stress is implicated in the pathophysiology of major depression and posttraumatic stress disorder. These conditions share core features, including motivational deficits, heighted anxiety, and sleep dysregulation. Chronic stress produces these same features in rodents, with some individuals being susceptible or resilient, as seen in humans. While stress-induced neuroadaptations within the nucleus accumbens are implicated in susceptibility-related dysregulation of motivational and emotional behaviors, their effects on sleep are unclear. METHODS: We used chemogenetics (DREADDs [designer receptors exclusively activated by designer drugs]) to examine the effects of selective alterations in activity of nucleus accumbens medium spiny neurons expressing dopamine D1 receptors (D1-MSNs) or dopamine D2 receptors (D2-MSNs) on sleep-related end points. Mice were implanted with wireless transmitters enabling continuous collection of data to quantify vigilance states over a 20-day test period. Parallel cohorts were examined in behavioral tests assessing stress susceptibility. RESULTS: D1- and D2-MSNs play dissociable roles in sleep regulation. Stimulation of inhibitory or excitatory DREADDs expressed in D1-MSNs exclusively affects rapid eye movement sleep, whereas targeting D2-MSNs affects slow wave sleep. The combined effects of D1-MSN inhibition and D2-MSN activation on sleep resemble those of chronic social defeat stress. Alterations in D1-MSN function also affect stress susceptibility in social behavior tests. Elevation of CREB (cAMP response element-binding protein) within D1-MSNs is sufficient to produce stress-like effects on rapid eye movement sleep. CONCLUSIONS: In addition to regulation of motivational and emotional behaviors, the nucleus accumbens also influences sleep, an end point with high translational relevance. These findings provide a neural basis for comorbidity in key features of stress-related illness.


Subject(s)
Nucleus Accumbens , Receptors, Dopamine D1 , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Sleep
10.
11.
Nat Commun ; 11(1): 5180, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057013

ABSTRACT

Fear and extinction learning are adaptive processes caused by molecular changes in specific neural circuits. Neurons expressing the corticotropin-releasing hormone gene (Crh) in central amygdala (CeA) are implicated in threat regulation, yet little is known of cell type-specific gene pathways mediating adaptive learning. We translationally profiled the transcriptome of CeA Crh-expressing cells (Crh neurons) after fear conditioning or extinction in mice using translating ribosome affinity purification (TRAP) and RNAseq. Differential gene expression and co-expression network analyses identified diverse networks activated or inhibited by fear vs extinction. Upstream regulator analysis demonstrated that extinction associates with reduced CREB expression, and viral vector-induced increased CREB expression in Crh neurons increased fear expression and inhibited extinction. These findings suggest that CREB, within CeA Crh neurons, may function as a molecular switch that regulates expression of fear and its extinction. Cell-type specific translational analyses may suggest targets useful for understanding and treating stress-related psychiatric illness.


Subject(s)
Central Amygdaloid Nucleus/physiology , Conditioning, Psychological/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Extinction, Psychological/physiology , Fear/physiology , Animals , Behavior, Animal , Central Amygdaloid Nucleus/cytology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Female , Male , Mice , Mice, Transgenic , Models, Animal , Neurons/metabolism , RNA-Seq
12.
Mol Psychiatry ; 25(10): 2330-2341, 2020 10.
Article in English | MEDLINE | ID: mdl-30610201

ABSTRACT

Immune activation during pregnancy via infection or autoimmune disease is a risk factor for neuropsychiatric illness. Mouse models of prenatal immune activation often involve maternal administration of agents that activate toll-like receptors (TLRs), a class of pattern recognition receptors that initiate innate immune responses. Such studies have focused primarily on activating the TLR3 or TLR4 subtypes, to mimic immune responses to viral or bacterial infections, respectively. Here, we characterize the effects of prenatal activation of TLR7, which is implicated in the pathogenesis of autoimmune disease. Prenatal TLR7 activation via administration of the selective agonist imiquimod (5.0 mg/kg) induces a phenotype in offspring characterized by reduced anxiety-like behavior, fragmented social behavior, and altered ultrasonic vocalization patterns at 6-12 weeks of age. The characteristics of this phenotype are readily distinguishable from-and in some ways opposite to-those seen following prenatal activation of TLR3 and/or TLR4. Prenatal TLR7-activated mice have normal baseline locomotor activity, but are hyperresponsive to stimuli including social partners, circadian cues, and gonadal hormone fluctuations. These alterations are accompanied by decreases in microglia density but increases in ramifications. RNA-sequencing of dorsal striatum, a region showing profound changes in microglial markers, indicates that prenatal TLR7 activation induces differential expression of hundreds of genes at 13 weeks of age, with virtually no overlap in differentially expressed genes between males and females. Our findings demonstrate that prenatal immune activation can promote a wide range of developmental trajectories, depending on the type and/or pattern of TLR activation and the sex of the offspring.


Subject(s)
Fetus , Immunity, Innate , Membrane Glycoproteins , Sex Characteristics , Toll-Like Receptor 7 , Animals , Cytokines , Female , Fetus/immunology , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Toll-Like Receptor 7/immunology
13.
Transl Psychiatry ; 8(1): 164, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135420

ABSTRACT

Behavioral and molecular characterization of cell-type-specific populations governing fear learning and behavior is a promising avenue for the rational identification of potential therapeutics for fear-related disorders. Examining cell-type-specific changes in neuronal translation following fear learning allows for targeted pharmacological intervention during fear extinction learning, mirroring possible treatment strategies in humans. Here we identify the central amygdala (CeA) Drd2-expressing population as a novel fear-supporting neuronal population that is molecularly distinct from other, previously identified, fear-supporting CeA populations. Sequencing of actively translating transcripts of Drd2 neurons using translating ribosome affinity purification (TRAP) technology identifies mRNAs that are differentially regulated following fear learning. Differentially expressed transcripts with potentially targetable gene products include Npy5r, Rxrg, Adora2a, Sst5r, Fgf3, Erbb4, Fkbp14, Dlk1, and Ssh3. Direct pharmacological manipulation of NPY5R, RXR, and ADORA2A confirms the importance of this cell population and these cell-type-specific receptors in fear behavior. Furthermore, these findings validate the use of functionally identified specific cell populations to predict novel pharmacological targets for the modulation of emotional learning.


Subject(s)
Fear/physiology , Learning/physiology , Neurons/physiology , Receptors, Dopamine D2/genetics , Animals , Behavior, Animal , Central Amygdaloid Nucleus/cytology , Conditioning, Psychological/physiology , Extinction, Psychological/physiology , Male , Mice, Inbred C57BL , Sequence Analysis, RNA
14.
eNeuro ; 5(1)2018.
Article in English | MEDLINE | ID: mdl-29445764

ABSTRACT

Molecular identification and characterization of fear controlling circuitries is a promising path towards developing targeted treatments of fear-related disorders. Three-color in situ hybridization analysis was used to determine whether somatostatin (SOM, Sst), neurotensin (NTS, Nts), corticotropin-releasing factor (CRF, Crf), tachykinin 2 (TAC2, Tac2), protein kinase c-δ (PKC-δ, Prkcd), and dopamine receptor 2 (DRD2, Drd2) mRNA colocalize in male mouse amygdala neurons. Expression and colocalization was examined across capsular (CeC), lateral (CeL), and medial (CeM) compartments of the central amygdala. The greatest expression of Prkcd and Drd2 were found in CeC and CeL. Crf was expressed primarily in CeL, while Sst-, Nts-, and Tac2-expressing neurons were distributed between CeL and CeM. High levels of colocalization were identified between Sst, Nts, Crf, and Tac2 within the CeL, while little colocalization was detected between any mRNAs within the CeM. These findings provide a more detailed understanding of the molecular mechanisms that regulate the development and maintenance of fear and anxiety behaviors.


Subject(s)
Central Amygdaloid Nucleus/metabolism , Fear/physiology , Neurons/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Male , Mice, Inbred C57BL , Neurotensin/metabolism , Protein Kinase C-delta/metabolism , Protein Precursors/metabolism , RNA, Messenger/metabolism , Receptors, Dopamine D2/metabolism , Somatostatin/metabolism , Tachykinins/metabolism
15.
Nat Commun ; 7: 13149, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27767183

ABSTRACT

Molecular characterization of neuron populations, particularly those controlling threat responses, is essential for understanding the cellular basis of behaviour and identifying pharmacological agents acting selectively on fear-controlling circuitry. Here we demonstrate a comprehensive workflow for identification of pharmacologically tractable markers of behaviourally characterized cell populations. Thy1-eNpHR-, Thy1-Cre- and Thy1-eYFP-labelled neurons of the BLA consistently act as fear inhibiting or 'Fear-Off' neurons during behaviour. We use cell-type-specific optogenetics and chemogenetics (DREADDs) to modulate activity in this population during behaviour to block or enhance fear extinction. Dissociated Thy1-eYFP neurons are isolated using FACS. RNA sequencing identifies genes strongly upregulated in RNA of this population, including Ntsr2, Dkk3, Rspo2 and Wnt7a. Pharmacological manipulation of neurotensin receptor 2 confirms behavioural effects observed in optogenetic and chemogenetic experiments. These experiments identify and validate Ntsr2-expressing neurons within the BLA, as a putative 'Fear-Off' population.


Subject(s)
Basolateral Nuclear Complex/metabolism , Extinction, Psychological/physiology , Fear/physiology , Memory/physiology , Thy-1 Antigens/genetics , Adaptor Proteins, Signal Transducing , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/drug effects , Clozapine/analogs & derivatives , Clozapine/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Gene Expression Regulation , Genes, Reporter , Injections, Intraventricular , Integrases/genetics , Integrases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Memory/drug effects , Mice , Oligopeptides/pharmacology , Optogenetics/methods , Piperidines/pharmacology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sequence Analysis, RNA , Stereotaxic Techniques , Thrombospondins/genetics , Thrombospondins/metabolism , Thy-1 Antigens/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
16.
J Neurosci ; 33(25): 10396-404, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785152

ABSTRACT

Research has identified distinct neuronal circuits within the basolateral amygdala (BLA) that differentially mediate fear expression versus inhibition; however, molecular markers of these populations remain unknown. Here we examine whether optogenetic activation of a cellular subpopulation, which may correlate with the physiologically identified extinction neurons in the BLA, would differentially support fear conditioning versus fear inhibition/extinction. We first molecularly characterized Thy1-channelrhodopsin-2 (Thy1-ChR2-EYFP)-expressing neurons as a subpopulation of glutamatergic pyramidal neurons within the BLA. Optogenetic stimulation of these neurons inhibited a subpopulation of medial central amygdala neurons and shunted excitation from the lateral amygdala. Brief activation of these neurons during fear training disrupted later fear memory in male mice. Optogenetic activation during unreinforced stimulus exposure enhanced extinction retention, but had no effect on fear expression, locomotion, or open-field behavior. Together, these data suggest that the Thy1-expressing subpopulation of BLA pyramidal neurons provide an important molecular and pharmacological target for inhibiting fear and enhancing extinction and for furthering our understanding of the molecular mechanisms of fear processing.


Subject(s)
Amygdala/physiology , Fear/physiology , Inhibition, Psychological , Neurons/physiology , Thy-1 Antigens/physiology , Amygdala/cytology , Animals , Channelrhodopsins , Conditioning, Psychological , Cues , Extinction, Psychological , Glutamic Acid/physiology , Immunohistochemistry , Lasers , Locomotion/physiology , Mice , Motor Activity/physiology , Patch-Clamp Techniques , Photic Stimulation , Thy-1 Antigens/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...