Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Psychiatry ; 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38409596

ABSTRACT

Posttraumatic stress disorder (PTSD) is a psychiatric disorder associated with traumatic memory, yet its etiology remains unclear. Reexperiencing symptoms are specific to PTSD compared to other anxiety-related disorders. Importantly, reexperiencing can be mimicked by retrieval-related events of fear memory in animal models of traumatic memory. Recent studies revealed candidate PTSD-associated genes that were related to the cyclic adenosine monophosphate (cAMP) signaling pathway. Here, we demonstrate the tight linkage between facilitated cAMP signaling and PTSD by analyzing loss- and gain-of-cAMP signaling effects on fear memory in mice and the transcriptomes of fear memory-activated mice and female PTSD patients with reexperiencing symptoms. Pharmacological and optogenetic upregulation or downregulation of cAMP signaling transduction enhanced or impaired, respectively, the retrieval and subsequent maintenance of fear memory in mice. In line with these observations, integrative mouse and human transcriptome analysis revealed the reduced mRNA expression of phosphodiesterase 4B (PDE4B), an enzyme that degrades cAMP, in the peripheral blood of PTSD patients showing more severe reexperiencing symptoms and the mouse hippocampus after fear memory retrieval. Importantly, more severe reexperiencing symptoms and lower PDE4B mRNA levels were correlated with decreased DNA methylation of a locus within PDE4B, suggesting the involvement of methylation in the mechanism of PTSD. These findings raise the possibility that the facilitation of cAMP signaling mediating the downregulation of PDE4B expression enhances traumatic memory, thereby playing a key role in the reexperiencing symptoms of PTSD patients as a functional index of these symptoms.

2.
Data Brief ; 46: 108862, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36624765

ABSTRACT

The transcription profile of microglia related to fear conditioning remains unclear. Here, we used Illumina MouseWG-6v2 microarrays to investigate the gene transcription changes in microglia and peripheral monocytes after contextual fear conditioning of C57BL/6 J mice. Mice were trained with or without a single minimized footshock stimulation (0-s or 2-s, 0.4 mA) and re-exposed to the training context without footshock for three different durations 24 h later: 0 min (FS0), 3 min (FS3), or 30 min (FS30). Whole brain microglia and peripheral monocytes were prepared 24 h after re-exposure using a neural tissue dissociation kit, including non-footshock controls for two re-exposure durations (Con3 and Con30). The data can be valuable for researchers interested in glial cells and neurotransmission studies and are related to the research article "Contextual fear conditioning regulates synapse-related gene transcription in mouse microglia".

3.
Brain Res Bull ; 189: 57-68, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35987296

ABSTRACT

Microglia have been suggested to be involved in the underlying mechanism of conditional fear memory formation by regulating inflammatory cytokines. However, the mechanism linking microglia and neuronal activity related to fear conditioning remains unclear. This study characterized the transcription profile of microglia in a fear memory conditional mouse model. Compared with those in control mice microglia, the most significantly induced genes were synapse-related, whereas immune-related genes were reduced due to fear memory consolidation. Whilst the increased expression of synapse-related genes was reversed after fear memory extinction, that of immunological genes was not, strongly suggesting a connection between microglia, neurons, and a dysregulated immune response following contextual fear conditioning. Furthermore, in the hippocampal microglia, we found that the expression of neurotransmitter release regulators, γ-aminobutyric acid (GABA) receptor GABRB3 and synapsin 1/2, increased under fear memory consolidation and restored (decreased) after extinction. In addition, compared with the transcription profile in peripheral monocytes, few overlapping genes were not enriched in biological processes. Taken together, the identified conditional fear stress-induced changes in mouse microglial transcription profiles suggest that microglia-neuron communication mediates contextual fear conditioning.


Subject(s)
Microglia , Synapsins , Animals , Cytokines/metabolism , Fear/physiology , Hippocampus/metabolism , Mice , Microglia/metabolism , Neurotransmitter Agents/metabolism , Synapses/metabolism , Synapsins/metabolism , Transcription, Genetic , gamma-Aminobutyric Acid/metabolism
4.
Biochem Biophys Res Commun ; 605: 45-50, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35313230

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by altered social communication, restricted interests, and stereotypic behaviors. Although the molecular and cellular pathogeneses of ASD remain elusive, impaired neural stem cell differentiation and neuronal migration during cortical development are suggested to be critically involved in ASD. ANK2, which encodes for a cytoskeletal scaffolding protein involved in recruiting membrane proteins into specialized membrane domains, has been identified as a high-confidence ASD risk gene. However, the role of ANK2 in early neural development remains unclear. In this study, we analyzed the role of ANK2 in the cerebral cortex of developing mouse using in utero electroporation. We provide evidence suggesting that ANK2 regulates neural stem cell differentiation and neuronal migration in the embryonic cerebral cortex, where Ank2 is highly expressed. We also demonstrated that Ank2 knockdown alters the expression of genes involved in neural development. Taken together, these results support the view that ANK2 haploinsufficiency in patients may impair neural development, resulting in an increased risk of ASD. Our study findings provide new insights into the molecular and cellular pathogenesis of ASD, given that among high-confidence ASD genes, ANK2 is rare in that it encodes for a scaffolding protein for the membrane protein complex required for neuronal functions.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Neural Stem Cells , Animals , Ankyrins/genetics , Ankyrins/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Humans , Mice , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism
5.
Mol Brain ; 14(1): 44, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33653368

ABSTRACT

Memory reconsolidation is thought to maintain or enhance an original memory or add new information to the memory. Retrieved inhibitory avoidance (IA) memory is enhanced through memory reconsolidation by activating gene expression in the amygdala, medial prefrontal cortex (mPFC), and hippocampus. However, it remains unclear how these regions interact to reconsolidate/enhance IA memory. Here, we found the interactions between the amygdala and mPFC as upstream regulators of the hippocampus for IA memory reconsolidation. Pharmacological inactivation of the amygdala, mPFC, or hippocampus immediately after IA memory retrieval blocked IA memory enhancement. More importantly, inactivation of the amygdala or mPFC blocked the induction of c-Fos in the amygdala, mPFC, and hippocampus, whereas hippocampal blockade inhibited it only in the hippocampus. These observations suggest interactions between the amygdala and mPFC and they both function as upstream regulators of the hippocampus to reconsolidate IA memory. Our findings suggest circuitry mechanisms underlying IA memory enhancement through reconsolidation between the amygdala, mPFC, and hippocampus.


Subject(s)
Amygdala/physiology , Avoidance Learning/physiology , Hippocampus/physiology , Memory/physiology , Mental Recall , Prefrontal Cortex/physiology , Amygdala/drug effects , Animals , Avoidance Learning/drug effects , Hippocampus/drug effects , Lidocaine/pharmacology , Memory/drug effects , Mental Recall/drug effects , Mice , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/metabolism
6.
Brain Behav Immun ; 94: 79-88, 2021 05.
Article in English | MEDLINE | ID: mdl-33677026

ABSTRACT

Neural inflammation is associated with cognitive decline, especially learning and memory. Tumor necrosis factor α (TNFα) is a major cytokine generated during neuroinflammation. Previous studies indicated that TNFα impairs hippocampus-dependent memory including contextual fear and spatial memories. However, it is unknown which memory processes are impaired by TNFα. Here, we show that TNFα blocked the retrieval and reconsolidation of contextual fear and spatial memories. Micro-infusion of TNFα into the dorsal hippocampus at 6-18 h before retrieval impaired the retrieval of contextual fear memory, although micro-infusion before contextual fear conditioning had no effect on memory formation. Interestingly, hippocampal TNFα micro-infusion before memory retrieval decreased freezing responses, even at 24 h after retrieval, suggesting that TNFα impairs the reconsolidation of contextual fear memory. Similarly, hippocampal TNFα micro-infusion impaired the retrieval and reconsolidation of spatial memory in the Morris water maze. Consistent with these observations, hippocampal TNFα micro-infusion before retrieval blocked the induction of c-fos expression in the hippocampus, which is a marker of neural activation, in response to the retrieval of contextual fear memory. Collectively, our findings indicate that TNFα negatively regulates the retrieval and reconsolidation of hippocampus-dependent memory.


Subject(s)
Fear , Hippocampus , Spatial Memory , Tumor Necrosis Factor-alpha , Animals , Hippocampus/metabolism , Proto-Oncogene Proteins c-fos/metabolism
7.
J Neurosci ; 41(6): 1288-1300, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33293359

ABSTRACT

The retrieval of fear memory induces two opposite memory process, i.e., reconsolidation and extinction. Brief retrieval induces reconsolidation to maintain or enhance fear memory, while prolonged retrieval extinguishes this memory. Although the mechanisms of reconsolidation and extinction have been investigated, it remains unknown how fear memory phases are switched from reconsolidation to extinction during memory retrieval. Here, we show that an extracellular signal-regulated kinase (ERK)-dependent memory transition process after retrieval regulates the switch of memory phases from reconsolidation to extinction by preventing induction of reconsolidation in an inhibitory avoidance (IA) task in male mice. First, the transition memory phase, which cancels the induction of reconsolidation, but is insufficient for the acquisition of extinction, was identified after reconsolidation, but before extinction phases. Second, the reconsolidation, transition, and extinction phases after memory retrieval showed distinct molecular and cellular signatures through cAMP responsive element binding protein (CREB) and ERK phosphorylation in the amygdala, hippocampus, and medial prefrontal cortex (mPFC). The reconsolidation phase showed increased CREB phosphorylation, while the extinction phase displayed several neural populations with various combinations of CREB and/or ERK phosphorylation, in these brain regions. Interestingly, the three memory phases, including the transition phase, showed transient ERK activation immediately after retrieval. Most importantly, the blockade of ERK in the amygdala, hippocampus, or mPFC at the transition memory phase disinhibited reconsolidation-induced enhancement of IA memory. These observations suggest that the ERK-signaling pathway actively regulates the transition of memory phase from reconsolidation to extinction and this process functions as a switch that cancels reconsolidation of fear memory.SIGNIFICANCE STATEMENT Retrieval of fear memory induces two opposite memory process; reconsolidation and extinction. Reconsolidation maintains/enhances fear memory, while extinction weakens fear memory. It remains unknown how memory phases are switched from reconsolidation to extinction during retrieval. Here, we identified an active memory transition process functioning as a switch that inhibits reconsolidation. This memory transition phase showed a transient increase of extracellular signal-regulated kinase (ERK) phosphorylation in the amygdala, hippocampus and medial prefrontal cortex (mPFC). Interestingly, inhibition of ERK in these regions at the transition phase disinhibited the reconsolidation-mediated enhancement of inhibitory avoidance (IA) memory. These findings suggest that the transition memory process actively regulates the switch of fear memory phases of fear memory by preventing induction of reconsolidation through the activation of the ERK-signaling pathway.


Subject(s)
Amygdala/enzymology , Extinction, Psychological/physiology , Hippocampus/enzymology , MAP Kinase Signaling System/physiology , Memory Consolidation/physiology , Prefrontal Cortex/enzymology , Animals , Fear , Male , Memory/physiology , Mice , Mice, Inbred C57BL
8.
Nat Commun ; 10(1): 5766, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852900

ABSTRACT

Cognitive performance in people varies according to time-of-day, with memory retrieval declining in the late afternoon-early evening. However, functional roles of local brain circadian clocks in memory performance remains unclear. Here, we show that hippocampal clock controlled by the circadian-dependent transcription factor BMAL1 regulates time-of-day retrieval profile. Inducible transgenic dominant negative BMAL1 (dnBMAL1) expression in mouse forebrain or hippocampus disrupted retrieval of hippocampal memories at Zeitgeber Time 8-12, independently of retention delay, encoding time and Zeitgeber entrainment cue. This altered retrieval profile was associated with downregulation of hippocampal Dopamine-cAMP signaling in dnBMAL1 mice. These changes included decreases in Dopamine Receptors (D1-R and D5-R) and GluA1-S845 phosphorylation by PKA. Consistently, pharmacological activation of cAMP-signals or D1/5Rs rescued impaired retrieval in dnBMAL1 mice. Importantly, GluA1 S845A knock-in mice showed similar retrieval deficits with dnBMAL1 mice. Our findings suggest mechanisms underlying regulation of retrieval by hippocampal clock through D1/5R-cAMP-PKA-mediated GluA1 phosphorylation.


Subject(s)
Circadian Clocks/physiology , Hippocampus/metabolism , Mental Recall/physiology , Receptors, AMPA/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/metabolism , Female , Gene Knock-In Techniques , Male , Maze Learning , Mice , Mice, Transgenic , Models, Animal , Phosphorylation/physiology
9.
Neuropsychopharmacol Rep ; 39(2): 140-145, 2019 06.
Article in English | MEDLINE | ID: mdl-30977307

ABSTRACT

AIMS: Lactobacillus species are used widely as various food and supplements to improve health. Previous studies have shown that heat-killed Lactobacillus brevis SBC8803 induces serotonin release from intestinal cells and affects sleep rhythm and the autonomic nervous system. However, the effect of SBC8803 on cognitive function remains unknown. Here, we examined the effects of dietary heat-killed SBC8803 on hippocampus-dependent memory and adult hippocampal neurogenesis. METHODS: Hippocampus-dependent memory performance was assessed in mice fed heat-killed SBC8803 using social recognition and contextual fear conditioning tasks. Adult hippocampal neurogenesis was evaluated before, during, and after feeding heat-killed SBC8803 by measuring the number of 5-bromo-2-deoxyuridine (BrdU)-positive cells following systemic injections of BrdU using immunohistochemistry. RESULTS: Mice fed a heat-killed SBC8803 diet showed an improvement of hippocampus-dependent social recognition and contextual fear memories and enhanced adult hippocampal neurogenesis by increasing the survival, but not proliferation, of newborn neurons. CONCLUSION: Dietary heat-killed SBC8803 functions as memory and neurogenesis enhancers.


Subject(s)
Hippocampus/drug effects , Levilactobacillus brevis/chemistry , Memory , Neurogenesis , Neuroprotective Agents/pharmacology , Animals , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL
10.
Mol Brain ; 12(1): 13, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30736823

ABSTRACT

The cerebellum regulates complex animal behaviors, such as motor control and spatial recognition, through communication with many other brain regions. The major targets of the cerebellar projections are the thalamic regions including the ventroanterior nucleus (VA) and ventrolateral nucleus (VL). Another thalamic target is the central lateral nucleus (CL), which receives the innervations mainly from the dentate nucleus (DN) in the cerebellum. Although previous electrophysiological studies suggest the role of the CL as the relay of cerebellar functions, the kinds of behavioral functions mediated by cerebellothalamic tracts projecting to the CL remain unknown. Here, we used immunotoxin (IT) targeting technology combined with a neuron-specific retrograde labeling technique, and selectively eliminated the cerebellothalamic tracts of mice. We confirmed that the number of neurons in the DN was selectively decreased by the IT treatment. These IT-treated mice showed normal overground locomotion with no ataxic behavior. However, elimination of these neurons impaired motor coordination in the rotarod test and forelimb movement in the reaching test. These mice showed intact acquisition and flexible change of spatial information processing in the place discrimination, Morris water maze, and T-maze tests. Although the tract labeling indicated the existence of axonal collaterals of the DN-CL pathway to the rostral part of the VA/VL complex, excitatory lesion of the rostral VA/VL did not show any significant alterations in motor coordination or forelimb reaching, suggesting no requirement of axonal branches connecting to the VL/VA complex for motor skill function. Taken together, our data highlight that the cerebellothalamic tracts projecting to the CL play a key role in the control of motor skills, including motor coordination and forelimb reaching, but not spatial recognition and its flexibility.


Subject(s)
Cerebellum/physiology , Intralaminar Thalamic Nuclei/physiology , Motor Skills/physiology , Neural Pathways/physiology , Animals , Axons/physiology , Behavior, Animal , Discrimination, Psychological , Gene Expression Regulation , HEK293 Cells , Humans , Learning , Male , Mice, Inbred C57BL
11.
Sci Rep ; 7: 42528, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195219

ABSTRACT

Transcription factor CREB is believed to play essential roles in the formation of long-term memory (LTM), but not in learning and short-term memory (STM). Surprisingly, we previously showed that transgenic mice expressing a dominant active mutant of CREB (DIEDML) in the forebrain (DIEDML mice) demonstrated enhanced STM and LTM in hippocampal-dependent, rapid, one-trial learning tasks. Here we show that constitutive activation of CREB enhances hippocampal-dependent learning of temporal association in trace fear conditioning and delayed matching-to-place tasks. We then show that in DIEDML mice the apical tuft dendrites of hippocampal CA1 pyramidal neurons, required for temporal association learning, display increased spine density, especially of thin spines and of Homer1-negative spines. In contrast, the basal and apical oblique dendrites of CA1 neurons, required for rapid one-trial learning, show increased density of thin, stubby, and mushroom spines and of Homer1-positive spines. Furthermore, DIEDML mice showed increased dendritic complexity in the proximal portion of apical CA1 dendrites to the soma. In contrast, forebrain overexpression of CaMKIV, leading to enhanced LTM but not STM, show normal learning and CA1 neuron morphology. These findings suggest that dendritic region-specific morphological changes in CA1 neurons by constitutive activation of CREB may contribute to improved learning and STM.


Subject(s)
Association Learning , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Cyclic AMP Response Element-Binding Protein/genetics , Dendritic Spines , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Transcriptional Activation , Animals , Conditioning, Classical , Cyclic AMP Response Element-Binding Protein/metabolism , Fear , Female , Male , Maze Learning , Memory, Short-Term , Mice
12.
Brain Behav Immun ; 59: 313-321, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27562421

ABSTRACT

The proinflammatory cytokine productions in the brain are altered in a process of fear memory formation, indicating a possibility that altered microglial function may contribute to fear memory formation. We aimed to investigate whether and how microglial function contributes to fear memory formation. Expression levels of M1- and M2-type microglial marker molecules in microglia isolated from each conditioned mice group were assessed by real-time PCR and immunohistochemistry. Levels of tumor necrosis factor (TNF)-α, but not of other proinflammatory cytokines produced by M1-type microglia, increased in microglia from mice representing retention of fear memory, and returned to basal levels in microglia from mice representing extinction of fear memory. Administration of inhibitors of TNF-α production facilitated extinction of fear memory. On the other hand, expression levels of M2-type microglia-specific cell adhesion molecules, CD206 and CD209, were decreased in microglia from mice representing retention of fear memory, and returned to basal levels in microglia from mice representing extinction of fear memory. Our findings indicate that microglial TNF-α is a key element of sustained fear memory and suggest that TNF-α inhibitors can be candidate molecules for mitigating posttraumatic reactions caused by persistent fear memory.


Subject(s)
Fear , Memory , Microglia/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Extinction, Psychological , Hippocampus/metabolism , Lectins, C-Type/metabolism , Macrophage Activation/drug effects , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , Minocycline/pharmacology , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/blood
13.
Elife ; 52016 Sep 26.
Article in English | MEDLINE | ID: mdl-27669409

ABSTRACT

Forgetting of recent fear memory is promoted by treatment with memantine (MEM), which increases hippocampal neurogenesis. The approaches for treatment of post-traumatic stress disorder (PTSD) using rodent models have focused on the extinction and reconsolidation of recent, but not remote, memories. Here we show that, following prolonged re-exposure to the conditioning context, enhancers of hippocampal neurogenesis, including MEM, promote forgetting of remote contextual fear memory. However, these interventions are ineffective following shorter re-exposures. Importantly, we find that long, but not short re-exposures activate gene expression in the hippocampus and induce hippocampus-dependent reconsolidation of remote contextual fear memory. Furthermore, remote memory retrieval becomes hippocampus-dependent after the long-time recall, suggesting that remote fear memory returns to a hippocampus dependent state after the long-time recall, thereby allowing enhanced forgetting by increased hippocampal neurogenesis. Forgetting of traumatic memory may contribute to the development of PTSD treatment.

14.
Neuron ; 84(1): 92-106, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25277455

ABSTRACT

CREB is a pivotal mediator of activity-regulated gene transcription that underlies memory formation and allocation. The contribution of a key CREB cofactor, CREB-regulated transcription coactivator 1 (CRTC1), has, however, remained elusive. Here we show that several constitutive kinase pathways and an activity-regulated phosphatase, calcineurin, converge to determine the nucleocytoplasmic shuttling of CRTC1. This, in turn, triggered an activity-dependent association of CRTC1 with CREB-dependent regulatory elements found on IEG promoters. Forced expression of nuclear CRTC1 in hippocampal neurons activated CREB-dependent transcription, and was sufficient to enhance contextual fear memory. Surprisingly, during contextual fear conditioning, we found evidence of nuclear recruitment of endogenous CRTC1 only in the basolateral amygdala, and not in the hippocampus. Consistently, CRTC1 knockdown in the amygdala, but not in the hippocampus, significantly attenuated fear memory. Thus, CRTC1 has a wide impact on CREB-dependent memory processes, but fine-tunes CREB output in a region-specific manner.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Fear/physiology , Hippocampus/metabolism , Memory/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Animals, Newborn , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/metabolism , Rats
15.
Elife ; 3: e02736, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24963141

ABSTRACT

Memory retrieval is considered to have roles in memory enhancement. Recently, memory reconsolidation was suggested to reinforce or integrate new information into reactivated memory. Here, we show that reactivated inhibitory avoidance (IA) memory is enhanced through reconsolidation under conditions in which memory extinction is not induced. This memory enhancement is mediated by neurons in the amygdala, hippocampus, and medial prefrontal cortex (mPFC) through the simultaneous activation of calcineurin-induced proteasome-dependent protein degradation and cAMP responsive element binding protein-mediated gene expression. Interestingly, the amygdala is required for memory reconsolidation and enhancement, whereas the hippocampus and mPFC are required for only memory enhancement. Furthermore, memory enhancement triggered by retrieval utilizes distinct mechanisms to strengthen IA memory by additional learning that depends only on the amygdala. Our findings indicate that reconsolidation functions to strengthen the original memory and show the dynamic nature of reactivated memory through protein degradation and gene expression in multiple brain regions.DOI: http://dx.doi.org/10.7554/eLife.02736.001.


Subject(s)
Fear/physiology , Memory/physiology , Amygdala/metabolism , Animals , Calcineurin/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Phosphorylation , Prefrontal Cortex/metabolism , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism
16.
Mol Pain ; 8: 90, 2012 Dec 31.
Article in English | MEDLINE | ID: mdl-23272977

ABSTRACT

CREB has been reported to be activated by injury and is commonly used as marker for pain-related plasticity changes in somatosensory pathways, including spinal dorsal horn neurons and the anterior cingulate cortex (ACC). However no evidence has been reported to support the direct role of activated CREB in injury-related behavioral sensitization (or allodynia). Here we report that genetic enhancement of CREB-mediated transcription selectively in forebrain areas enhanced behavioral responses to non-noxious stimuli after chronic inflammation (CFA model) or nerve injury. In contrast, behavioral acute responses to peripheral subcutaneous injection of formalin did not show any significant difference. Furthermore, acute pain responses to noxious thermal stimuli were also not affected. Our results thus provide direct evidence that cortical CREB-mediated transcription contributes to behavioral allodynia in animal models of chronic inflammatory or neuropathic pain.


Subject(s)
CREB-Binding Protein/genetics , Inflammation/metabolism , Neuralgia/metabolism , Pain/metabolism , Prosencephalon/metabolism , Animals , Genetic Enhancement , Hyperalgesia/genetics , Hyperalgesia/metabolism , Inflammation/genetics , Mice , Mice, Mutant Strains , Neuralgia/genetics , Pain/genetics
17.
J Neurosci ; 31(24): 8786-802, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21677163

ABSTRACT

Unraveling the mechanisms by which the molecular manipulation of genes of interest enhances cognitive function is important to establish genetic therapies for cognitive disorders. Although CREB is thought to positively regulate formation of long-term memory (LTM), gain-of-function effects of CREB remain poorly understood, especially at the behavioral level. To address this, we generated four lines of transgenic mice expressing dominant active CREB mutants (CREB-Y134F or CREB-DIEDML) in the forebrain that exhibited moderate upregulation of CREB activity. These transgenic lines improved not only LTM but also long-lasting long-term potentiation in the CA1 area in the hippocampus. However, we also observed enhanced short-term memory (STM) in contextual fear-conditioning and social recognition tasks. Enhanced LTM and STM could be dissociated behaviorally in these four lines of transgenic mice, suggesting that the underlying mechanism for enhanced STM and LTM are distinct. LTM enhancement seems to be attributable to the improvement of memory consolidation by the upregulation of CREB transcriptional activity, whereas higher basal levels of BDNF, a CREB target gene, predicted enhanced shorter-term memory. The importance of BDNF in STM was verified by microinfusing BDNF or BDNF inhibitors into the hippocampus of wild-type or transgenic mice. Additionally, increasing BDNF further enhanced LTM in one of the lines of transgenic mice that displayed a normal BDNF level but enhanced LTM, suggesting that upregulation of BDNF and CREB activity cooperatively enhances LTM formation. Our findings suggest that CREB positively regulates memory consolidation and affects memory performance by regulating BDNF expression.


Subject(s)
CREB-Binding Protein/metabolism , Memory, Long-Term/physiology , Memory, Short-Term/physiology , Up-Regulation/physiology , Analysis of Variance , Animals , Bacterial Proteins/genetics , Behavior, Animal , Brain-Derived Neurotrophic Factor/pharmacology , CREB-Binding Protein/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Carbazoles/pharmacology , Cell Line, Transformed , Chlorocebus aethiops , Conditioning, Classical/physiology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Discrimination, Psychological , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fear , Fluorescence Resonance Energy Transfer , Hippocampus/drug effects , Hippocampus/physiology , Indole Alkaloids/pharmacology , Long-Term Potentiation/genetics , Luminescent Proteins/genetics , Maze Learning , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Patch-Clamp Techniques , Phenylalanine/genetics , RNA, Messenger/metabolism , Rats , Social Behavior , Transfection/methods , Tyrosine/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
18.
Mol Brain ; 4: 4, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21244716

ABSTRACT

BACKGROUND: Memory consolidation is a process to stabilize short-term memory, generating long-term memory. A critical biochemical feature of memory consolidation is a requirement for gene expression. Previous studies have shown that fear memories are consolidated through the activation of gene expression in the amygdala and hippocampus, indicating essential roles of these brain regions in memory formation. However, it is still poorly understood whether gene expression in brain regions other than the amygdala/hippocampus is required for the consolidation of fear memory; however, several brain regions are known to play modulatory roles in fear memory formation. RESULTS: To further understand the mechanisms underlying the formation of fear memory, we first identified brain regions where gene expression is activated after learning inhibitory avoidance (IA) by analyzing the expression of the immediately early genes c-fos and Arc as markers. Similarly with previous findings, the induction of c-fos and Arc expression was observed in the amygdala and hippocampus. Interestingly, we also observed the induction of c-fos and Arc expression in the medial prefrontal cortex (mPFC: prelimbic (PL) and infralimbic (IL) regions) and Arc expression in the anterior cingulate cortex (ACC). We next examined the roles of these brain regions in the consolidation of IA memory. Consistent with previous findings, inhibiting protein synthesis in the hippocampus blocked the consolidation of IA memory. More importantly, inhibition in the mPFC or ACC also blocked the formation of IA memory. CONCLUSION: Our observations indicated that the formation of IA memory requires gene expression in the ACC and mPFC as well as in the amygdala and hippocampus, suggesting essential roles of the ACC and mPFC in IA memory formation.


Subject(s)
Avoidance Learning/physiology , Gene Expression , Gyrus Cinguli/physiology , Memory, Long-Term/physiology , Memory, Short-Term/physiology , Prefrontal Cortex/physiology , Transcriptional Activation , Animals , Biomarkers/metabolism , Conditioning, Operant , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Fear/physiology , Gyrus Cinguli/anatomy & histology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/anatomy & histology , Protein Biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism
19.
Mol Brain ; 3: 16, 2010 May 24.
Article in English | MEDLINE | ID: mdl-20497541

ABSTRACT

Mounting evidence suggests that neural oscillations are related to the learning and consolidation of newly formed memory in the mammalian brain. Four to seven Hertz (4-7 Hz) oscillations in the prefrontal cortex are also postulated to be involved in learning and attention processes. Additionally, slow delta oscillations (1-4 Hz) have been proposed to be involved in memory consolidation or even synaptic down scaling during sleep. The molecular mechanisms which link learning-related oscillations during wakefulness to sleep-related oscillations remain unknown. We show that increasing the expression of calcium/calmodulin dependent protein kinase IV (CaMKIV), a key nucleic protein kinase, selectively enhances 4-7.5 Hz oscillation power during trace fear learning and slow delta oscillations during subsequent sleep. These oscillations were found to be boosted in response to the trace fear paradigm and are likely to be localized to regions of the prefrontal cortex. Correlation analyses demonstrate that a proportion of the variance in 4-7.5 Hz oscillations, during fear conditioning, could account for some degree of learning and subsequent memory formation, while changes in slow delta power did not share this predictive strength. Our data emphasize the role of CaMKIV in controlling learning and sleep-related oscillations and suggest that oscillatory activity during wakefulness may be a relevant predictor of subsequent memory consolidation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/physiology , Memory/physiology , Sleep/physiology , Animals , Behavior, Animal/physiology , Biological Clocks , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Conditioning, Operant , Electroencephalography , Fear , Humans , Learning/physiology , Mice , Mice, Transgenic
20.
Article in Japanese | MEDLINE | ID: mdl-19663263

ABSTRACT

Reconsolidation and extinction of fear memories are induced by re-exposure to the conditioned stimulus (CS) but they appear to be opposite processes; as the fear memory is maintained or inhibited through reconsolidation and extinction, respectively. More importantly, reconsolidation and extinction are thought to be potential targets for the treatment of Post Traumatic Stress Disorder (PTSD). From this view, it is important to understand mechanisms by which reactivated fear memories are reconsolidated or extinguished. Here, we review processes for fear memory regulation including consolidation, reconsolidation and extinction, and discuss implications of fear memory regulation with PTSD.


Subject(s)
Extinction, Psychological , Fear/psychology , Memory , Humans , Stress Disorders, Post-Traumatic/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...