Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 42(4): 112291, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36952344

ABSTRACT

Multiple brain regions are engaged in classical fear conditioning. Despite evidence for cerebellar involvement in fear conditioning, the mechanisms by which cerebellar outputs modulate fear learning and memory remain unclear. We identify a population of deep cerebellar nucleus (DCN) neurons with monosynaptic glutamatergic projections to the lateral parabrachial nucleus (lPBN) (DCN→lPBN neurons) in mice. While optogenetic suppression of DCN→lPBN neurons impairs auditory fear memory, activation of DCN→lPBN neurons elicits freezing behavior only after auditory fear conditioning. Moreover, auditory fear conditioning potentiates DCN-lPBN synapses, and subsequently, auditory cue activates lPBN neurons after fear conditioning. Furthermore, DCN→lPBN neuron activation can replace the auditory cue but not footshock in fear conditioning. These findings demonstrate that cerebellar nuclei modulate auditory fear conditioning via transmitting conditioned stimuli signals to the lPBN. Collectively, our findings suggest that the DCN-lPBN circuit is a part of neuronal substrates within interconnected brain regions underscoring auditory fear memory.


Subject(s)
Cerebellar Nuclei , Parabrachial Nucleus , Mice , Animals , Cerebellar Nuclei/physiology , Parabrachial Nucleus/physiology , Neurons/physiology , Conditioning, Classical/physiology , Fear/physiology
2.
Neuron ; 111(3): 418-429.e4, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36460007

ABSTRACT

Observational fear, a form of emotional contagion, is thought to be a basic form of affective empathy. However, the neural process engaged at the specific moment when socially acquired information provokes an emotional response remains elusive. Here, we show that reciprocal projections between the anterior cingulate cortex (ACC) and basolateral amygdala (BLA) in the right hemisphere are essential for observational fear, and 5-7 Hz neural oscillations were selectively increased in those areas at the onset of observational freezing. A closed-loop disruption demonstrated the causal relationship between 5-7 Hz oscillations in the cingulo-amygdala circuit and observational fear responses. The increase/decrease in theta power induced by optogenetic manipulation of the hippocampal theta rhythm bi-directionally modulated observational fear. Together, these results indicate that hippocampus-dependent 5-7 Hz oscillations in the cingulo-amygdala circuit in the right hemisphere are the essential component of the cognitive process that drives empathic fear, but not freezing, in general.


Subject(s)
Basolateral Nuclear Complex , Empathy , Mice , Animals , Amygdala/physiology , Basolateral Nuclear Complex/physiology , Gyrus Cinguli/physiology , Fear/physiology
3.
Neurobiol Learn Mem ; 169: 107171, 2020 03.
Article in English | MEDLINE | ID: mdl-31978552

ABSTRACT

Memory is stored in our brains over a temporally graded transition. With time, recently formed memories are transformed into remote memories for permanent storage; multiple brain regions, such as the hippocampus and neocortex, participate in this process. In this study, we aimed to understand the molecular mechanism of systems consolidation of memory and to investigate the brain regions that contribute to this regulation. We first carried out a contextual fear memory test using a transgenic mouse line, which expressed exogenously-derived Aplysia octopamine receptors in the forebrain region, such that, in response to octopamine treatment, cyclic adenosine monophosphate (cAMP) levels could be transiently elevated. From this experiment, we revealed that transient elevation of cAMP levels in the forebrain during systems consolidation led to an enhancement in remote fear memory and increased miniature excitatory synaptic currents in layer II/III of the anterior cingulate cortex (ACC). Furthermore, using an adeno-associated-virus-driven DREADD system, we investigated the specific regions in the forebrain that contribute to the regulation of memory transfer into long-term associations. Our results implied that transient elevation of cAMP levels was induced chemogenetically in the ACC, but not in the hippocampus, and showed a significant enhancement of remote memory. This finding suggests that neuronal activation during systems consolidation through the elevation of cAMP levels in the ACC contributes to remote memory enhancement.


Subject(s)
Cyclic AMP/physiology , Fear/physiology , Gyrus Cinguli/physiology , Hippocampus/physiology , Memory Consolidation/physiology , Memory, Long-Term/physiology , Neurons/physiology , Animals , Male , Mice, Inbred C57BL , Mice, Transgenic
4.
Nature ; 566(7744): 339-343, 2019 02.
Article in English | MEDLINE | ID: mdl-30760920

ABSTRACT

A psychotherapeutic regimen that uses alternating bilateral sensory stimulation (ABS) has been used to treat post-traumatic stress disorder. However, the neural basis that underlies the long-lasting effect of this treatment-described as eye movement desensitization and reprocessing-has not been identified. Here we describe a neuronal pathway driven by the superior colliculus (SC) that mediates persistent attenuation of fear. We successfully induced a lasting reduction in fear in mice by pairing visual ABS with conditioned stimuli during fear extinction. Among the types of visual stimulation tested, ABS provided the strongest fear-reducing effect and yielded sustained increases in the activities of the SC and mediodorsal thalamus (MD). Optogenetic manipulation revealed that the SC-MD circuit was necessary and sufficient to prevent the return of fear. ABS suppressed the activity of fear-encoding cells and stabilized inhibitory neurotransmission in the basolateral amygdala through a feedforward inhibitory circuit from the MD. Together, these results reveal the neural circuit that underlies an effective strategy for sustainably attenuating traumatic memories.


Subject(s)
Anxiety/psychology , Anxiety/therapy , Extinction, Psychological/physiology , Fear/physiology , Fear/psychology , Neural Pathways/physiology , Superior Colliculi/cytology , Superior Colliculi/physiology , Animals , Anxiety/physiopathology , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/physiology , Conditioning, Classical/physiology , Feedback, Physiological , Male , Mediodorsal Thalamic Nucleus/cytology , Mediodorsal Thalamic Nucleus/physiology , Mice , Neural Inhibition , Optogenetics , Photic Stimulation , Stress Disorders, Post-Traumatic , Time Factors
5.
Nat Biotechnol ; 33(10): 1092-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26368050

ABSTRACT

Calcium (Ca(2+)) signals that are precisely modulated in space and time mediate a myriad of cellular processes, including contraction, excitation, growth, differentiation and apoptosis. However, study of Ca(2+) responses has been hampered by technological limitations of existing Ca(2+)-modulating tools. Here we present OptoSTIM1, an optogenetic tool for manipulating intracellular Ca(2+) levels through activation of Ca(2+)-selective endogenous Ca(2+) release-activated Ca(2+) (CRAC) channels. Using OptoSTIM1, which combines a plant photoreceptor and the CRAC channel regulator STIM1 (ref. 4), we quantitatively and qualitatively controlled intracellular Ca(2+) levels in various biological systems, including zebrafish embryos and human embryonic stem cells. We demonstrate that activating OptoSTIM1 in the CA1 hippocampal region of mice selectively reinforced contextual memory formation. The broad utility of OptoSTIM1 will expand our mechanistic understanding of numerous Ca(2+)-associated processes and facilitate screening for drug candidates that antagonize Ca(2+) signals.


Subject(s)
Calcium Channels/genetics , Calcium Signaling/genetics , Calcium/metabolism , Embryonic Stem Cells/physiology , Hippocampus/physiology , Optogenetics/methods , Animals , Embryonic Stem Cells/cytology , Genetic Engineering/methods , Hippocampus/cytology , Humans , Mice
6.
Mol Pain ; 7: 96, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22168443

ABSTRACT

Phosphoinositide 3-kinases (PI3Ks) are important for synaptic plasticity and various brain functions. The only class IB isoform of PI3K, PI3Kγ, has received the most attention due to its unique roles in synaptic plasticity and cognition. However, the potential role of PI3Kγ in sensory transmission, such as pain and itch has not been examined. In this study, we present the evidence for the first time, that genetic deletion of PI3Kγ enhanced scratching behaviours in histamine-dependent and protease-activated receptor 2 (PAR-2)-dependent itch. In contrast, PI3Kγ-deficient mice did not exhibit enhanced scratching in chloroquine-induced itch, suggesting that PI3Kγ selectively contributes to certain types of behavioal itch response. Furthermore, PI3Kγ-deficient mice exhibited normal acute nociceptive responses to thermal and mechanical noxious stimuli. Behavioral licking responses to intraplantar injections of formalin and mechanical allodynia in a chronic inflammatory pain model (CFA) were also not affected by PI3Kγ gene deletion. Our findings indicate that PI3Kγ selectively contributes to behavioral itching induced by histamine and PAR-2 agonist, but not chloroquine agonist.


Subject(s)
Behavior, Animal , Class Ib Phosphatidylinositol 3-Kinase/genetics , Genetic Enhancement/methods , Pruritus/genetics , Animals , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Histamine/adverse effects , Histamine/pharmacology , Mice , Mice, Knockout , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics
7.
Nat Neurosci ; 14(11): 1447-54, 2011 Oct 23.
Article in English | MEDLINE | ID: mdl-22019731

ABSTRACT

Phosphatidylinositol 3-kinase (PI3K) has been implicated in synaptic plasticity and other neural functions in the brain. However, the role of individual PI3K isoforms in the brain is unclear. We investigated the role of PI3Kγ in hippocampal-dependent synaptic plasticity and cognitive functions. We found that PI3Kγ has a crucial and specific role in NMDA receptor (NMDAR)-mediated synaptic plasticity at mouse Schaffer collateral-commissural synapses. Both genetic deletion and pharmacological inhibition of PI3Kγ disrupted NMDAR long-term depression (LTD) while leaving other forms of synaptic plasticity intact. Accompanying this physiological deficit, the impairment of NMDAR LTD by PI3Kγ blockade was specifically correlated with deficits in behavioral flexibility. These findings suggest that a specific PI3K isoform, PI3Kγ, is critical for NMDAR LTD and some forms of cognitive function. Thus, individual isoforms of PI3Ks may have distinct roles in different types of synaptic plasticity and may therefore influence various kinds of behavior.


Subject(s)
Behavior, Animal/physiology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Long-Term Synaptic Depression/genetics , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Analysis of Variance , Animals , Behavior, Animal/drug effects , Biophysics , Chromones/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Electric Stimulation/methods , Environment , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Extinction, Psychological/physiology , Fear/physiology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/cytology , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Male , Maze Learning/drug effects , Maze Learning/physiology , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Neurons/drug effects , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Phosphorylation/genetics , Quinoxalines/pharmacology , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Thiazolidinediones/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...