Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Cell Rep ; 42(12): 113488, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37995189

ABSTRACT

Response to threatening environmental stimuli requires detection and encoding of important environmental features that dictate threat. Aversive events are highly salient, which promotes associative learning about stimuli that signal this threat. The nucleus accumbens is uniquely positioned to process this salient, aversive information and promote motivated output, through plasticity on the major projection neurons in the brain area. We describe a nucleus accumbens core local circuit whereby excitatory plasticity facilitates learning and recall of discrete aversive cues. We demonstrate that putative nucleus accumbens substance P release and long-term excitatory plasticity on dopamine 2 receptor-expressing projection neurons are required for cue-dependent fear learning. Additionally, we find that fear learning and recall is dependent on distinct projection neuron subtypes. Our work demonstrates a critical role for nucleus accumbens substance P in cue-dependent aversive learning.


Subject(s)
Cues , Nucleus Accumbens , Nucleus Accumbens/physiology , Avoidance Learning , Substance P , Receptors, Dopamine
3.
Cell Rep ; 42(6): 112591, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37261953

ABSTRACT

Synapses in the brain exhibit cell-type-specific differences in basal synaptic transmission and plasticity. Here, we evaluated cell-type-specific specializations in the composition of glutamatergic synapses, identifying Btbd11 as an inhibitory interneuron-specific, synapse-enriched protein. Btbd11 is highly conserved across species and binds to core postsynaptic proteins, including Psd-95. Intriguingly, we show that Btbd11 can undergo liquid-liquid phase separation when expressed with Psd-95, supporting the idea that the glutamatergic postsynaptic density in synapses in inhibitory interneurons exists in a phase-separated state. Knockout of Btbd11 decreased glutamatergic signaling onto parvalbumin-positive interneurons. Further, both in vitro and in vivo, Btbd11 knockout disrupts network activity. At the behavioral level, Btbd11 knockout from interneurons alters exploratory behavior, measures of anxiety, and sensitizes mice to pharmacologically induced hyperactivity following NMDA receptor antagonist challenge. Our findings identify a cell-type-specific mechanism that supports glutamatergic synapse function in inhibitory interneurons-with implications for circuit function and animal behavior.


Subject(s)
Synapses , Synaptic Transmission , Animals , Mice , Disks Large Homolog 4 Protein/metabolism , Interneurons/metabolism , Mice, Knockout , Pyramidal Cells/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Transcription Factors/metabolism
4.
bioRxiv ; 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-36798245

ABSTRACT

Response to threatening environmental stimuli requires detection and encoding of important environmental features that dictate threat. Aversive events are highly salient which promotes associative learning about stimuli that signal this threat. The nucleus accumbens is uniquely positioned to process this salient, aversive information and promote motivated output, through plasticity on the major projection neurons in the brain area. We uncovered a nucleus accumbens core local circuit whereby excitatory plasticity facilitates learning and recall of discrete aversive cues. We demonstrate that putative nucleus accumbens substance P release and long-term excitatory plasticity on dopamine 2 receptor expressing projection neurons is required for cue-dependent fear learning. Additionally, we found fear learning and recall were dependent on distinct projection-neuron subtypes. Our work demonstrates a critical role for Nucleus Accumbens substance P in cue-dependent aversive learning.

5.
Curr Biol ; 31(12): 2652-2657.e3, 2021 06 21.
Article in English | MEDLINE | ID: mdl-33848459

ABSTRACT

Animals learn not only what is potentially useful but also what is meaningless and should be disregarded. How this is accomplished is a key but seldom explored question in psychology and neuroscience. Learning to ignore irrelevant cues is evident in latent inhibition-the ubiquitous phenomenon where presenting a cue several times without consequences leads to retardation of subsequent conditioning to that cue.1,2 Does learning to ignore these cues, because they predict nothing, involve the same neural circuits that are critical to learning to make predictions about other "real world" impending events? If so, the orbitofrontal cortex (OFC), as a key node in such networks, should be important.3 Specifically, the OFC has been hypothesized to participate in the recognition of hidden task states, which are not directly signaled by explicit outcomes.4 Evaluating its involvement in pre-exposure learning during latent inhibition would be an acid test for this hypothesis. Here, we report that selective chemogenetic inactivation of rat orbitofrontal cortex principal neurons during stimulus pre-exposure markedly reduces latent inhibition in subsequent conditioning. Inactivation only during pre-exposure ensured that the observed effects were due to an impact on the acquisition of information prior to its use in any sort of behavior, i.e., during latent learning. Further behavioral tests confirmed this, showing that the impact of OFC inactivation during pre-exposure was limited to the latent inhibition effect. These results demonstrate that the OFC is important for latent learning and the formation of associations even in the absence of explicit outcomes.


Subject(s)
Learning , Prefrontal Cortex , Animals , Cues , Neurons , Rats
6.
Neuropsychopharmacology ; 46(2): 277-278, 2021 01.
Article in English | MEDLINE | ID: mdl-33067544
7.
Neuron ; 103(3): 489-505.e7, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31204082

ABSTRACT

Despite a wealth of clinical and preclinical data implicating the serotonin (5-HT) system in fear-related affective disorders, a precise definition of this neuromodulator's role in fear remains elusive. Using convergent anatomical and functional approaches, we interrogate the contribution to fear of basal amygdala (BA) 5-HT inputs from the dorsal raphe nucleus (DRN). We show the DRN→BA 5-HT pathway is engaged during fear memory formation and retrieval, and activity of these projections facilitates fear and impairs extinction. The DRN→BA 5-HT pathway amplifies fear-associated BA neuronal firing and theta power and phase-locking. Although fear recruits 5-HT and VGluT3 co-expressing DRN neurons, the fear-potentiating influence of the DRN→BA 5-HT pathway requires signaling at BA 5-HT1A/2A receptors. Input-output mapping illustrates how the DRN→BA 5-HT pathway is anatomically distinct and connected with other brain regions that mediate fear. These findings reveal how a discrete 5-HT circuit orchestrates a broader neural network to calibrate aversive memory.


Subject(s)
Amygdala/physiology , Avoidance Learning/physiology , Dorsal Raphe Nucleus/physiology , Neural Pathways/physiology , Serotonin/physiology , Animals , Conditioning, Classical/physiology , Extinction, Psychological , Fear/physiology , Female , Genes, Reporter , Immobility Response, Tonic/physiology , Male , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Optogenetics , Synaptophysin/administration & dosage , Synaptophysin/analysis , Theta Rhythm/physiology
8.
Neuropsychopharmacology ; 44(6): 1177, 2019 05.
Article in English | MEDLINE | ID: mdl-30787427

ABSTRACT

The original version of this Article contained an error in the spelling of the author Anna K Radke, which was incorrectly given as Anna R Radke. This has now been corrected in both the PDF and HTML versions of the Article.

9.
Neuropsychopharmacology ; 44(6): 1163-1173, 2019 05.
Article in English | MEDLINE | ID: mdl-30622300

ABSTRACT

Obsessive-compulsive disorder (OCD) is a severe, chronic neuropsychiatric disorder with a strong genetic component. The SLC1A1 gene encoding the neuronal glutamate transporter EAAT3 has been proposed as a candidate gene for this disorder. Gene variants affecting SLC1A1 expression in human brain tissue have been associated with OCD. Several mouse models fully or partially lacking EAAT3 have shown no alterations in baseline anxiety-like or repetitive behaviors. We generated a transgenic mouse model (EAAT3glo) to achieve conditional, Cre-dependent EAAT3 overexpression and evaluated the overall impact of increased EAAT3 expression at behavioral and synaptic levels. Mice with EAAT3 overexpression driven by CaMKIIα-promoter (EAAT3glo/CMKII) displayed increased anxiety-like and repetitive behaviors that were both restored by chronic, but not acute, treatment with fluoxetine or clomipramine. EAAT3glo/CMKII mice also displayed greater spontaneous recovery of conditioned fear. Electrophysiological and biochemical analyses at corticostriatal synapses of EAAT3glo/CMKII mice revealed changes in NMDA receptor subunit composition and altered NMDA-dependent synaptic plasticity. By recapitulating relevant behavioral, neurophysiological, and psychopharmacological aspects, our results provide support for the glutamatergic hypothesis of OCD, particularly for the increased EAAT3 function, and provide a valuable animal model that may open novel therapeutic approaches to treat this devastating disorder.


Subject(s)
Anxiety/metabolism , Behavior, Animal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cerebral Cortex/metabolism , Excitatory Amino Acid Transporter 3/metabolism , Neostriatum/metabolism , Neuronal Plasticity/physiology , Obsessive-Compulsive Disorder/metabolism , Animals , Cell Line , Clomipramine/pharmacology , Disease Models, Animal , Excitatory Amino Acid Transporter 3/genetics , Fluoxetine/pharmacology , Gene Expression/genetics , Mice , Mice, Transgenic , Neuroblastoma , Patch-Clamp Techniques , Selective Serotonin Reuptake Inhibitors/pharmacology
10.
J Neurosci ; 37(7): 1785-1796, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087766

ABSTRACT

The serotonin (5-HT) system and the amygdala are key regulators of emotional behavior. Several lines of evidence suggest that 5-HT transmission in the amygdala is implicated in the susceptibility and drug treatment of mood disorders. Therefore, elucidating the physiological mechanisms through which midbrain 5-HT neurons modulate amygdala circuits could be pivotal in understanding emotional regulation in health and disease. To shed light on these mechanisms, we performed patch-clamp recordings from basal amygdala (BA) neurons in brain slices from mice with channelrhodopsin genetically targeted to 5-HT neurons. Optical stimulation of 5-HT terminals at low frequencies (≤1 Hz) evoked a short-latency excitation of BA interneurons (INs) that was depressed at higher frequencies. Pharmacological analysis revealed that this effect was mediated by glutamate and not 5-HT because it was abolished by ionotropic glutamate receptor antagonists. Optical stimulation of 5-HT terminals at higher frequencies (10-20 Hz) evoked both slow excitation and slow inhibition of INs. These effects were mediated by 5-HT because they were blocked by antagonists of 5-HT2A and 5-HT1A receptors, respectively. These fast glutamate- and slow 5-HT-mediated responses often coexisted in the same neuron. Interestingly, fast-spiking and non-fast-spiking INs displayed differential modulation by glutamate and 5-HT. Furthermore, optical stimulation of 5-HT terminals did not evoke glutamate release onto BA principal neurons, but inhibited these cells directly via activation of 5-HT1A receptors and indirectly via enhanced GABA release. Collectively, these findings suggest that 5-HT neurons exert a frequency-dependent, cell-type-specific control over BA circuitry via 5-HT and glutamate co-release to inhibit the BA output.SIGNIFICANCE STATEMENT The modulation of the amygdala by serotonin (5-HT) is important for emotional regulation and is implicated in the pathogenesis and treatment of affective disorders. Therefore, it is essential to determine the physiological mechanisms through which 5-HT neurons in the dorsal raphe nuclei modulate amygdala circuits. Here, we combined optogenetic, electrophysiological, and pharmacological approaches to study the effects of activation of 5-HT axons in the basal nucleus of the amygdala (BA). We found that 5-HT neurons co-release 5-HT and glutamate onto BA neurons in a cell-type-specific and frequency-dependent manner. Therefore, we suggest that theories on the contribution of 5-HT neurons to amygdala function should be revised to incorporate the concept of 5-HT/glutamate cotransmission.


Subject(s)
Amygdala/cytology , Glutamic Acid/metabolism , Nerve Net/physiology , Neurons/metabolism , Serotonin/metabolism , Animals , Animals, Newborn , Channelrhodopsins , Excitatory Amino Acid Agents/pharmacology , Female , GABA Antagonists/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/drug effects , Piperazines/pharmacology , Pyridines/pharmacology , Receptors, Serotonin/metabolism , Serotonin/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...