1
|
Cohen N, Rabinowitch I. Resolving transitions between distinct phases of memory consolidation at high resolution in Caenorhabditis elegans. iScience 2024; 27:111147. [PMID: 39524366 PMCID: PMC11547966 DOI: 10.1016/j.isci.2024.111147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/26/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Memory consolidation following learning is a dynamic and complex process comprising several transitions between distinct memory phases. Although memory consolidation has been studied extensively, it remains difficult to draw an integral description that can delimit the transition points between specific memory phases at the behavioral, neuronal, and genetic levels. To this end, we have developed a rapid and robust aversive conditioning protocol for the nematode worm Caenorhabditis elegans, tracing memory consolidation within the first hour post conditioning and then up to 18 h post conditioning. This made it possible to uncover time-dependent involvement of primary sensory neurons, transcription and translation processes, and diverse gene populations in memory consolidation. The change in neuronal valence was strong enough to induce second order conditioning, and was amenable to considerable modulation in specific mutant strains. Together, our work lends memory consolidation to detailed temporal and spatial analysis, advancing system-wide understanding of learning and memory.
Collapse
Affiliation(s)
- Netanel Cohen
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Ithai Rabinowitch
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| |
Collapse
|
2
|
Brusman LE, Sadino JM, Fultz AC, Kelberman MA, Dowell RD, Allen MA, Donaldson ZR. Single nucleus RNA-sequencing reveals transcriptional synchrony across different relationships. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587112. [PMID: 39605537 PMCID: PMC11601461 DOI: 10.1101/2024.03.27.587112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
As relationships mature, partners share common goals, improve their ability to work together, and experience coordinated emotions. However, the neural underpinnings responsible for this unique, pair-specific experience remain largely unexplored. Here, we used single nucleus RNA-sequencing to examine the transcriptional landscape of the nucleus accumbens (NAc) in socially monogamous prairie voles in peer or mating-based relationships. We show that, regardless of pairing type, prairie voles exhibit transcriptional synchrony with a partner. Further, we identify genes expressed in oligodendrocyte progenitor cells that are synchronized between partners, correlated with dyadic behavior, and sensitive to partner separation. Together, our data indicate that the pair-specific social environment profoundly shapes transcription in the NAc. This provides a potential biological mechanism by which shared social experience reinforces and strengthens relationships.
Collapse
Affiliation(s)
- Liza E. Brusman
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder; Boulder, CO 80309 USA
| | - Julie M. Sadino
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder; Boulder, CO 80309 USA
| | - Allison C. Fultz
- Department of Psychology and Neuroscience, University of Colorado Boulder; Boulder, CO, 80309 USA
| | - Michael A. Kelberman
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder; Boulder, CO 80309 USA
| | - Robin D. Dowell
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder; Boulder, CO 80309 USA
- Biofrontiers Institute, University of Colorado Boulder; Boulder, CO, 80309 USA
| | - Mary A. Allen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder; Boulder, CO 80309 USA
- Biofrontiers Institute, University of Colorado Boulder; Boulder, CO, 80309 USA
| | - Zoe R. Donaldson
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder; Boulder, CO 80309 USA
- Department of Psychology and Neuroscience, University of Colorado Boulder; Boulder, CO, 80309 USA
| |
Collapse
|
3
|
Seemiller LR, Goldberg LR, Sebastian A, Siegel SR, Praul C, Zeid D, Albert I, Beierle J, Bryant CD, Gould TJ. Alcohol and fear conditioning produce strain-specific changes in the dorsal hippocampal transcriptome of adolescent C57BL/6J and DBA/2J mice. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:2022-2034. [PMID: 39279663 DOI: 10.1111/acer.15440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/22/2024] [Accepted: 08/21/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Adolescent sensitivity to alcohol is influenced by genetic background. Data from our laboratory suggested that adolescent C57BL/6J and DBA/2J inbred mice differed in susceptibility to alcohol-induced deficits in dorsal hippocampus-dependent contextual fear learning. METHODS To investigate the biological underpinnings of this strain difference, we examined dorsal hippocampus gene expression using RNA-sequencing after alcohol or saline administration followed by Pavlovian fear conditioning across male and female C57BL/6J and DBA/2J adolescents. RESULTS Strains exhibited dramatic differences in dorsal hippocampus gene expression. Specifically, C57BL/6J and DBA/2J strains differed by 3526 transcripts in males and 2675 transcripts in females. We identified pathways likely to be involved in mediating alcohol's effects on learning, including networks associated with Chrna7, a gene encoding the nicotinic cholinergic receptor alpha 7 subunit, and Fmr1, a gene encoding the fragile X messenger ribonucleoprotein. CONCLUSIONS These findings provide insight into the mechanisms underlying strain differences in alcohol's effects on learning and suggest that different biological networks are recruited for learning based on genetics, sex, and alcohol exposure.
Collapse
Affiliation(s)
- Laurel R Seemiller
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Lisa R Goldberg
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Aswathy Sebastian
- Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania, USA
| | - Sue Rutherford Siegel
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Craig Praul
- Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania, USA
| | - Dana Zeid
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| | - Istvan Albert
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA
| | - Jacob Beierle
- Department of Pharmacology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Camron D Bryant
- Department of Pharmaceutical Sciences, Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, Pennsylvania, USA
| |
Collapse
|
4
|
Brito DVC, Kupke J, Sokolov R, Cambridge S, Both M, Bengtson CP, Rozov A, Oliveira AMM. Biphasic Npas4 expression promotes inhibitory plasticity and suppression of fear memory consolidation in mice. Mol Psychiatry 2024; 29:1929-1940. [PMID: 38347124 PMCID: PMC11408256 DOI: 10.1038/s41380-024-02454-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/19/2024]
Abstract
Long-term memories are believed to be encoded by unique transcriptional signatures in the brain. The expression of immediate early genes (IEG) promotes structural and molecular changes required for memory consolidation. Recent evidence has shown that the brain is equipped with mechanisms that not only promote, but actively constrict memory formation. However, it remains unknown whether IEG expression may play a role in memory suppression. Here we uncovered a novel function of the IEG neuronal PAS domain protein 4 (Npas4), as an inducible memory suppressor gene of highly salient aversive experiences. Using a contextual fear conditioning paradigm, we found that low stimulus salience leads to monophasic Npas4 expression, while highly salient learning induces a biphasic expression of Npas4 in the hippocampus. The later phase requires N-methyl-D-aspartate (NMDA) receptor activity and is independent of dopaminergic neurotransmission. Our in vivo pharmacological and genetic manipulation experiments suggested that the later phase of Npas4 expression restricts the consolidation of a fear memory and promote behavioral flexibility, by facilitating fear extinction and the contextual specificity of fear responses. Moreover, immunofluorescence and electrophysiological analysis revealed a concomitant increase in synaptic input from cholecystokinin (CCK)-expressing interneurons. Our results demonstrate how salient experiences evoke unique temporal patterns of IEG expression that fine-tune memory consolidation. Moreover, our study provides evidence for inducible gene expression associated with memory suppression as a possible mechanism to balance the consolidation of highly salient memories, and thereby to evade the formation of maladaptive behavior.
Collapse
Affiliation(s)
- David V C Brito
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139, Faro, Portugal
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve, 8005-139, Faro, Portugal
| | - Janina Kupke
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, the Netherlands
| | - Rostilav Sokolov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia
- Federal Center of Brain Research and Neurotechnology, 117513, Moscow, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny, Novgorod, Russia
| | - Sidney Cambridge
- Anatomy II, Dr. Senckenberg Anatomy, Goethe-University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Martin Both
- Institute of Physiology and Pathophysiology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
| | - Andrei Rozov
- Federal Center of Brain Research and Neurotechnology, 117513, Moscow, Russia
- Institute of Physiology and Pathophysiology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
- OpenLab of Neurobiology, Kazan Federal University, 420008, Kazan, Russia
| | - Ana M M Oliveira
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany.
- Department of Molecular and Cellular Cognition Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany.
| |
Collapse
|
5
|
Smies CW, Bellfy L, Wright DS, Bennetts SG, Urban MW, Brunswick CA, Shu G, Kwapis JL. Pharmacological HDAC3 inhibition alters memory updating in young and old male mice. Front Mol Neurosci 2024; 17:1429880. [PMID: 38989157 PMCID: PMC11234845 DOI: 10.3389/fnmol.2024.1429880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
Long-term memories are not stored in a stable state but must be flexible and dynamic to maintain relevance in response to new information. Existing memories are thought to be updated through the process of reconsolidation, in which memory retrieval initiates destabilization and updating to incorporate new information. Memory updating is impaired in old age, yet little is known about the mechanisms that go awry. One potential mechanism is the repressive histone deacetylase 3 (HDAC3), which is a powerful negative regulator of memory formation that contributes to age-related impairments in memory formation. Here, we tested whether HDAC3 also contributes to age-related impairments in memory updating using the Objects in Updated Locations (OUL) paradigm. We show that blocking HDAC3 immediately after updating with the pharmacological inhibitor RGFP966 ameliorated age-related impairments in memory updating in 18-m.o. male mice. Surprisingly, we found that post-update HDAC3 inhibition in young (3-m.o.) male mice had no effect on memory updating but instead impaired memory for the original information, suggesting that the original and updated information may compete for expression at test and HDAC3 helps regulate which information is expressed. To test this idea, we next assessed whether HDAC3 inhibition would improve memory updating in young male mice given a weak, subthreshold update. Consistent with our hypothesis, we found that HDAC3 blockade strengthened the subthreshold update without impairing memory for the original information, enabling balanced expression of the original and updated information. Together, this research suggests that HDAC3 may contribute to age-related impairments in memory updating and may regulate the strength of a memory update in young mice, shifting the balance between the original and updated information at test.
Collapse
Affiliation(s)
- Chad W. Smies
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Lauren Bellfy
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Destiny S. Wright
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Sofia G. Bennetts
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Mark W. Urban
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Chad A. Brunswick
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Guanhua Shu
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - Janine L. Kwapis
- Department of Biology, Pennsylvania State University, University Park, PA, United States
- Center for the Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
6
|
Duggins P, Eliasmith C. A scalable spiking amygdala model that explains fear conditioning, extinction, renewal and generalization. Eur J Neurosci 2024; 59:3093-3116. [PMID: 38616566 DOI: 10.1111/ejn.16338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/03/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024]
Abstract
The amygdala (AMY) is widely implicated in fear learning and fear behaviour, but it remains unclear how the many biological components present within AMY interact to achieve these abilities. Building on previous work, we hypothesize that individual AMY nuclei represent different quantities and that fear conditioning arises from error-driven learning on the synapses between AMY nuclei. We present a computational model of AMY that (a) recreates the divisions and connections between AMY nuclei and their constituent pyramidal and inhibitory neurons; (b) accommodates scalable high-dimensional representations of external stimuli; (c) learns to associate complex stimuli with the presence (or absence) of an aversive stimulus; (d) preserves feature information when mapping inputs to salience estimates, such that these estimates generalize to similar stimuli; and (e) induces a diverse profile of neural responses within each nucleus. Our model predicts (1) defensive responses and neural activities in several experimental conditions, (2) the consequence of artificially ablating particular nuclei and (3) the tendency to generalize defensive responses to novel stimuli. We test these predictions by comparing model outputs to neural and behavioural data from animals and humans. Despite the relative simplicity of our model, we find significant overlap between simulated and empirical data, which supports our claim that the model captures many of the neural mechanisms that support fear conditioning. We conclude by comparing our model to other computational models and by characterizing the theoretical relationship between pattern separation and fear generalization in healthy versus anxious individuals.
Collapse
Affiliation(s)
- Peter Duggins
- Centre for Theoretical Neuroscience, University of Waterloo, Waterloo, Ontario, Canada
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Chris Eliasmith
- Centre for Theoretical Neuroscience, University of Waterloo, Waterloo, Ontario, Canada
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Department of Philosophy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
7
|
Yang J, Lin L, Zou GJ, Wang LF, Li F, Li CQ, Cui YH, Huang FL. CK2 negatively regulates the extinction of remote fear memory. Behav Brain Res 2024; 465:114960. [PMID: 38494129 DOI: 10.1016/j.bbr.2024.114960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Cognitive behavioral therapy, rooted in exposure therapy, is currently the primary approach employed in the treatment of anxiety-related conditions, including post-traumatic stress disorder (PTSD). In laboratory settings, fear extinction in animals is a commonly employed technique to investigate exposure therapy; however, the precise mechanisms underlying fear extinction remain elusive. Casein kinase 2 (CK2), which regulates neuroplasticity via phosphorylation of its substrates, has a significant influence in various neurological disorders, such as Alzheimer's disease and Parkinson's disease, as well as in the process of learning and memory. In this study, we adopted a classical Pavlovian fear conditioning model to investigate the involvement of CK2 in remote fear memory extinction and its underlying mechanisms. The results indicated that the activity of CK2 in the medial prefrontal cortex (mPFC) of mice was significantly upregulated after extinction training of remote cued fear memory. Notably, administration of the CK2 inhibitor CX-4945 prior to extinction training facilitated the extinction of remote fear memory. In addition, CX-4945 significantly upregulated the expression of p-ERK1/2 and p-CREB in the mPFC. Our results suggest that CK2 negatively regulates remote fear memory extinction, at least in part, by inhibiting the ERK-CREB pathway. These findings contribute to our understanding of the underlying mechanisms of remote cued fear extinction, thereby offering a theoretical foundation and identifying potential targets for the intervention and treatment of PTSD.
Collapse
Affiliation(s)
- Jie Yang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China; School of Basic Medicine, Yiyang Medical College, Yiyang, Hunan 413000, China
| | - Lin Lin
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Guang-Jing Zou
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Lai-Fa Wang
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha, Hunan 410219, China
| | - Fang Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Chang-Qi Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Yan-Hui Cui
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China.
| | - Fu-Lian Huang
- School of Basic Medicine, Yiyang Medical College, Yiyang, Hunan 413000, China.
| |
Collapse
|
8
|
Smies CW, Bellfy L, Wright DS, Bennetts SS, Urban MW, Brunswick CA, Shu G, Kwapis JL. Pharmacological HDAC3 inhibition alters memory updating in young and old mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593015. [PMID: 38766057 PMCID: PMC11100699 DOI: 10.1101/2024.05.08.593015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Long-term memories are not stored in a stable state but must be flexible and dynamic to maintain relevance in response to new information. Existing memories are thought to be updated through the process of reconsolidation, in which memory retrieval initiates destabilization and updating to incorporate new information. Memory updating is impaired in old age, yet little is known about the mechanisms that go awry. One potential mechanism is the repressive histone deacetylase 3 (HDAC3), which is a powerful negative regulator of memory formation that contributes to age-related impairments in memory formation. Here, we tested whether HDAC3 also contributes to age-related impairments in memory updating using the Objects in Updated Locations (OUL) paradigm. We show that blocking HDAC3 immediately after updating with the pharmacological inhibitor RGFP966 ameliorated age-related impairments in memory updating in 18-m.o. mice. Surprisingly, we found that post-update HDAC3 inhibition in young (3-m.o.) mice had no effect on memory updating but instead impaired memory for the original information, suggesting that the original and updated information may compete for expression at test and HDAC3 helps regulate which information is expressed. To test this idea, we next assessed whether HDAC3 inhibition would improve memory updating in young mice given a weak, subthreshold update. Consistent with our hypothesis, we found that HDAC3 blockade strengthened the subthreshold update without impairing memory for the original information, enabling balanced expression of the original and updated information. Together, this research suggests that HDAC3 may contribute to age-related impairments in memory updating and may regulate the strength of a memory update in young mice, shifting the balance between the original and updated information at test.
Collapse
|
9
|
Gebicke-Haerter PJ. The computational power of the human brain. Front Cell Neurosci 2023; 17:1220030. [PMID: 37608987 PMCID: PMC10441807 DOI: 10.3389/fncel.2023.1220030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/05/2023] [Indexed: 08/24/2023] Open
Abstract
At the end of the 20th century, analog systems in computer science have been widely replaced by digital systems due to their higher computing power. Nevertheless, the question keeps being intriguing until now: is the brain analog or digital? Initially, the latter has been favored, considering it as a Turing machine that works like a digital computer. However, more recently, digital and analog processes have been combined to implant human behavior in robots, endowing them with artificial intelligence (AI). Therefore, we think it is timely to compare mathematical models with the biology of computation in the brain. To this end, digital and analog processes clearly identified in cellular and molecular interactions in the Central Nervous System are highlighted. But above that, we try to pinpoint reasons distinguishing in silico computation from salient features of biological computation. First, genuinely analog information processing has been observed in electrical synapses and through gap junctions, the latter both in neurons and astrocytes. Apparently opposed to that, neuronal action potentials (APs) or spikes represent clearly digital events, like the yes/no or 1/0 of a Turing machine. However, spikes are rarely uniform, but can vary in amplitude and widths, which has significant, differential effects on transmitter release at the presynaptic terminal, where notwithstanding the quantal (vesicular) release itself is digital. Conversely, at the dendritic site of the postsynaptic neuron, there are numerous analog events of computation. Moreover, synaptic transmission of information is not only neuronal, but heavily influenced by astrocytes tightly ensheathing the majority of synapses in brain (tripartite synapse). At least at this point, LTP and LTD modifying synaptic plasticity and believed to induce short and long-term memory processes including consolidation (equivalent to RAM and ROM in electronic devices) have to be discussed. The present knowledge of how the brain stores and retrieves memories includes a variety of options (e.g., neuronal network oscillations, engram cells, astrocytic syncytium). Also epigenetic features play crucial roles in memory formation and its consolidation, which necessarily guides to molecular events like gene transcription and translation. In conclusion, brain computation is not only digital or analog, or a combination of both, but encompasses features in parallel, and of higher orders of complexity.
Collapse
Affiliation(s)
- Peter J. Gebicke-Haerter
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
10
|
Nachtigall EG, D R de Freitas J, de C Myskiw J, R G Furini C. Role of hippocampal Wnt signaling pathways on contextual fear memory reconsolidation. Neuroscience 2023:S0306-4522(23)00248-8. [PMID: 37286160 DOI: 10.1016/j.neuroscience.2023.05.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 05/15/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023]
Abstract
Memories already consolidated when reactivated return to a labile state and can be modified, this process is known as reconsolidation. It is known the Wnt signaling pathways can modulate hippocampal synaptic plasticity as well as learning and memory. Yet, Wnt signaling pathways interact with NMDA (N-methyl-D-aspartate) receptors. However, whether canonical Wnt/β-catenin and non-canonical Wnt/Ca2+ signaling pathways are required in the CA1 region of hippocampus for contextual fear memory reconsolidation remains unclear. So, here we verified that the inhibition of canonical Wnt/β-catenin pathway with DKK1 (Dickkopf-1) into CA1 impaired the reconsolidation of contextual fear conditioning (CFC) memory when administered immediately and 2h after reactivation session but not 6h later, while the inhibition of non-canonical Wnt/Ca2+ signaling pathway with SFRP1 (Secreted frizzled-related protein-1) into CA1 immediately after reactivation session had no effect. Moreover, the impairment induced by DKK1 was blocked by the administration of the agonist of the NMDA receptors glycine site, D-Serine, immediately and 2h after reactivation session. We found that hippocampal canonical Wnt/β-catenin is necessary to the reconsolidation of CFC memory at least two hours after reactivation, while non-canonical Wnt/Ca2+ signaling pathway is not involved in this process and, that there is a link between Wnt/β-catenin signaling pathway and NMDA receptors. In view of this, this study provides new evidence regarding the neural mechanisms underlying contextual fear memory reconsolidation and contributes to provide a new possible target for the treatment of fear related disorders.
Collapse
Affiliation(s)
- Eduarda G Nachtigall
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - Bldg. 63, 3(rd) floor, 90610-000, Porto Alegre, RS, Brazil
| | - Júlia D R de Freitas
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - Bldg. 63, 3(rd) floor, 90610-000, Porto Alegre, RS, Brazil
| | - Jociane de C Myskiw
- Psychobiology and Neurocomputation Laboratory (LPBNC), Department of Biophysics, Institute of Biosciences, Federal University of Rio Grande do Sul (UFRGS). Av. Bento Gonçalves, 9500, Bldg. 43422, room 208A, 91501-970, Porto Alegre, RS, Brazil
| | - Cristiane R G Furini
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - Bldg. 63, 3(rd) floor, 90610-000, Porto Alegre, RS, Brazil; Institute of Geriatrics and Gerontology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681 - Bldg. 40, 8(th) floor, 90610-000, Porto Alegre, RS, Brazil.
| |
Collapse
|
11
|
Inactivation of the dorsal CA1 hippocampus impairs the consolidation of discriminative avoidance memory by modulating the intrinsic and extrinsic hippocampal circuitry. J Chem Neuroanat 2023; 128:102209. [PMID: 36496001 DOI: 10.1016/j.jchemneu.2022.102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Despite progress in understanding the role of the dorsal hippocampus in the acquisition, consolidation and retrieval of episodic-like memory, plastic changes within the intra- and extrahippocampal circuits for aversive memory formation and anxiety-like behaviours must still be identified since both processes contribute to multiple aspects of flexible decision-making. Here, we investigated the effect of reversible inactivation induced by a muscimol microinfusion into the dorsal CA1 subfield (dCA1) either prior to acquisition or to retrieval testing of a discriminative avoidance task performed in a plus-maze apparatus (PM-DAT). Differential cAMP-response-element-binding protein 1 (CREB-1) expression in the dorsal and ventral CA1 and CA3 of the hippocampus (dCA1, dCA3, vCA1, and vCA3), dorsal dentate gyrus (dDG), and infralimbic (IL) and prelimbic (PrL) regions of the medial prefrontal cortex was also assessed to investigate the molecular changes associated with the consolidation or retrieval of episodic-like memory and anxiety. Adult male Wistar rats were assigned to two control groups, learning (no surgery/no microinfusion, n = 7) and sham-operated (sham surgery/no microinfusion, n = 6) groups, or four experimental groups, in which the vehicle (0.5 µl per side, n = 8/per group) or a GABAA receptor agonist (0.5 µg/0.5 µl muscimol/per side) was bilaterally microinfused in the dCA1 30 min prior to training (n = 9) or prior to testing sessions (n = 6) with a 24 h intertrial interval. Memory was evaluated using the percentage of time spent in the nonaversive enclosed arms, whereas anxiety was measured by calculating the percentages of time spent and entries into open arms and the percentage of time spent self-grooming. Our findings corroborated previous data showing that the dCA1 is required for discriminative avoidance consolidation. Furthermore, additional information indicated that impaired long-term memory was associated with downregulated CREB-1 expression in the dDG and vCA3. Moreover, memory retrieval was not impaired by dCA1 inactivation prior to the testing session, which was associated with the upregulation of CREB-1 in the dCA3 and vCA1 and downregulation in the dCA1 and vCA3. Differential expression of CREB was not identified in the IL or PrL areas. These results improve our understanding of how the hippocampal circuitry mediates the acquisition and retrieval of aversive memory and anxiety.
Collapse
|
12
|
Varela V, Evdokimidis I, Potagas C. Binding objects to their spatiotemporal context: Age gradient and neuropsychological correlates of What-Where-When task performance. APPLIED NEUROPSYCHOLOGY. ADULT 2023; 30:214-226. [PMID: 34053387 DOI: 10.1080/23279095.2021.1924719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A number of recent studies have shown that the ability to accurately recall bound object and spatiotemporal aspects of an experienced event develops gradually in children and is greatly impaired in the elderly, reflecting developmental discontinuities in the integrity of the underlying medial temporal lobe network. Using a novel What-Where-When (WWW) visuospatial reconstruction task, the experiential memory performance of a group of healthy older adults (aged 60-80) was compared to that of a group of younger adults (aged 20-40). Both groups were equated on their general cognitive ability, their executive functioning, and on the presence of depression, anxiety, and stress symptomatology. As hypothesized, the performance of the older adults in the binding task was significantly lower, with younger participants recalling three times the amount of bound object and spatiotemporal triads than their older counterparts. Psychomotor speed was found to be lower in older adults and was the only neuropsychological index to significantly affect success on the WWW binding task. Based on this and other relevant studies, the selective associative memory impairment obtained using a non-verbal What-Where-When paradigm emerges as a marker for the detection of early pre-clinical signs of experiential memory pathology.
Collapse
Affiliation(s)
- Vasiliki Varela
- Eginition Hospital, Department of Neurology, School of Medicine, University of Athens, Athens, Greece
| | - Ioannis Evdokimidis
- Eginition Hospital, Department of Neurology, School of Medicine, University of Athens, Athens, Greece
| | - Constantin Potagas
- Eginition Hospital, Department of Neurology, School of Medicine, University of Athens, Athens, Greece
| |
Collapse
|
13
|
Correa-Netto NF, Masukawa MY, Silva-Gomes AM, Linardi A, Santos-Junior JG. Memory reactivation mediates emotional valence updating of contextual memory in mice with protracted morphine withdrawal. Behav Brain Res 2023; 438:114212. [PMID: 36370948 DOI: 10.1016/j.bbr.2022.114212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/29/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Mice subjected to morphine locomotor sensitization develop increased anxiety-behavior expression during protracted morphine withdrawal. This behavioral change is dependent on reexposure to the context of locomotor sensitization and reflects a state of conditioned anxiety. In this study, the effect of memory reconsolidation on the expression of conditioned anxiety in mice with protracted morphine withdrawal was examined. Five experimental protocols involving male C57BL/6 mice were used in which the animals were subjected to locomotor sensitization induced by morphine and reexposed to the context associated with the drug effect 28 days after locomotor sensitization and immediately after subjected to elevated plus maze. In experiment 1, mice were subjected or not to memory reactivation session and was observed that memory reactivation 27 days after sensitization reduced conditioned anxiety. In experiment 2, mice were subjected to memory reactivation, 24 h, 6 h or 1 h before contextual reexposure, and the effect of memory reactivation coincided with the temporal requirement for reconsolidation. In experiment 3, which involved exposure to a situation of acute stress immediately before memory reactivation, the mice demonstrated a return to increased conditioned anxiety. To confirm the influence of reconsolidation, in experiments 4 and 5, mice subjected to memory reactivation were treated with Nimodipine, diazepam or cyclohexamine, substances commonly used as pharmacological controls in reconsolidation experiments. Treatment with each substance separately inhibited the effect of reactivation in experiment 5 (presence of acute stressor) but not in experiment 4 (absence of acute stressor). These results suggest that, in our experimental model, reconsolidation is mediated through updating of the emotional valence of contextual memory associated with the administration of morphine.
Collapse
Affiliation(s)
- Nelson Francisco Correa-Netto
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, São Paulo 01221-020, SP, Brazil.
| | - Márcia Yuriko Masukawa
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, São Paulo 01221-020, SP, Brazil
| | - Alessandro Marcos Silva-Gomes
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, São Paulo 01221-020, SP, Brazil
| | - Alessandra Linardi
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, São Paulo 01221-020, SP, Brazil
| | - Jair Guilherme Santos-Junior
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, São Paulo 01221-020, SP, Brazil
| |
Collapse
|
14
|
Rosa J, de Carvalho Myskiw J, Fiorenza NG, Furini CRG, Sapiras GG, Izquierdo I. Hippocampal cholinergic receptors and the mTOR participation in fear-motivated inhibitory avoidance extinction memory. Behav Brain Res 2023; 437:114129. [PMID: 36179804 DOI: 10.1016/j.bbr.2022.114129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 09/07/2022] [Accepted: 09/24/2022] [Indexed: 10/14/2022]
Abstract
Evidence has demonstrated the hippocampal cholinergic system and the mammalian target of rapamycin (mTOR) participation during the memory formation of aversive events. This study assessed the role of these systems in the hippocampus for the extinction memory process by submitting male Wistar rats to fear-motivated step-down inhibitory avoidance (IA). The post-extinction session administration of the nicotinic and muscarinic cholinergic receptor antagonists, mecamylamine and scopolamine, respectively, both at doses of 2 µg/µl/side, and rapamycin, an mTOR inhibitor (0.02 µg/µl/side), into the CA1 region of the dorsal hippocampus, impaired the IA extinction memory. Furthermore, the nicotinic and muscarinic cholinergic receptor agonists, nicotine and muscarine, respectively, had a dose-dependent effect on the IA extinction memory when administered intra-CA1, immediately after the extinction session. Nicotine (0.6 µg/µl/side) and muscarine (0.02 µg/µl/side), respectively, had no effect, while the higher doses (6 and 2 µg/µl/side, respectively) impaired the IA extinction memory. Interestingly, the co-administration of muscarine at the lower dose blocked the impairment that was induced by rapamycin. This effect was not observed when nicotine at the lower dose was co-administered. These results have demonstrated the participation of the cholinergic receptors and mTOR in the hippocampus for IA extinction, and that the cholinergic agonists had a dose-dependent effect on the IA extinction memory. This study provides insights related to the behavioural aspects and the neurobiological properties underlying the early stage of fear-motivated IA extinction memory consolidation and suggests that there is hippocampal muscarinic receptor participation independent of mTOR in this memory process.
Collapse
Affiliation(s)
- Jessica Rosa
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Ipiranga 6690, Floor 2, 90610-600 Porto Alegre, RS, Brazil; Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo (USP), Bandeirantes 3900, 14049-900 Ribeirao Preto, SP, Brazil.
| | - Jociane de Carvalho Myskiw
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Ipiranga 6690, Floor 2, 90610-600 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil; Department of Biophysics, Institute of Biosciences, Federal University of Rio Grande do Sul (UFRGS), Bento Gonçalves 9500, Building 43422, Room 208 A, 91501-970 Porto Alegre, RS, Brazil
| | - Natalia Gindri Fiorenza
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Ipiranga 6690, Floor 2, 90610-600 Porto Alegre, RS, Brazil; Oswaldo Cruz Foundation (FIOCRUZ), Branch Ceara, 60760-000 Eusebio, CE, Brazil
| | - Cristiane Regina Guerino Furini
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Ipiranga 6690, Floor 2, 90610-600 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil; Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Ipiranga 6690, 3rd Floor, 90610-000 Porto Alegre, RS, Brazil
| | - Gerson Guilherme Sapiras
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Ipiranga 6690, Floor 2, 90610-600 Porto Alegre, RS, Brazil; Clinical Hospital of Passo Fundo (HCPF), Tiradentes 295, 99010-260 Passo Fundo, RS, Brazil
| | - Ivan Izquierdo
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Ipiranga 6690, Floor 2, 90610-600 Porto Alegre, RS, Brazil; National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
15
|
Inhibition of late mRNA synthesis in the hippocampus impairs consolidation and reconsolidation of spatial memory in male rats. Neurobiol Learn Mem 2022; 195:107687. [DOI: 10.1016/j.nlm.2022.107687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/19/2022]
|
16
|
Gold PE. Revisiting and revising memory consolidation: Personal reflections on the research legacy of Ivan Izquierdo. Neuroscience 2022; 497:4-13. [PMID: 35667494 DOI: 10.1016/j.neuroscience.2022.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 10/18/2022]
Abstract
Two important themes in Ivan Izquierdo's research each offered both answers and questions about the topic of memory formation and maintenance. The first theme provided evidence supporting the view that short- and long-term memory were distinct processes and could be selectively modulated by several treatments, with some affecting only short-term, others only affecting long-term memory, and still others affecting both. Over many years, Izquierdo's laboratory documented molecular responses across time after training obtaining results that showed differences as well as similarities in the biochemical changes during the first 1-2 hours and the next 4-6 hours after training, i.e., during the transition from short- to long-term memory. This work clarified the biological underpinnings of the memory processes. The second theme described waves of susceptibility of memory to enhancing and impairing treatments after time, a biphasic profile that contrasted with earlier monotonic decreases in the efficacy of memory modulating treatments as a function of time between training and treatment. Remarkably, these waves of susceptibility to modification were accompanied by biphasic changes in molecular measures at similar times after training. Remarkably, some of the molecular players exhibited persistent changes after training, with increases in levels lasting days following the training experience. These persistent molecular changes may reveal a biological basis for the dynamic nature of memories seen long after the initial memory is consolidated.
Collapse
Affiliation(s)
- Paul E Gold
- Department of Biology, Syracuse, NY, 13224, United States.
| |
Collapse
|
17
|
Zhang Y, Smolen P, Alberini CM, Baxter DA, Byrne JH. Computational analysis of memory consolidation following inhibitory avoidance (IA) training in adult and infant rats: Critical roles of CaMKIIα and MeCP2. PLoS Comput Biol 2022; 18:e1010239. [PMID: 35759520 PMCID: PMC9269953 DOI: 10.1371/journal.pcbi.1010239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/08/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Key features of long-term memory (LTM), such as its stability and persistence, are acquired during processes collectively referred to as consolidation. The dynamics of biological changes during consolidation are complex. In adult rodents, consolidation exhibits distinct periods during which the engram is more or less resistant to disruption. Moreover, the ability to consolidate memories differs during developmental periods. Although the molecular mechanisms underlying consolidation are poorly understood, the initial stages rely on interacting signaling pathways that regulate gene expression, including brain-derived neurotrophic factor (BDNF) and Ca2+/calmodulin-dependent protein kinase II α (CaMKIIα) dependent feedback loops. We investigated the ways in which these pathways may contribute to developmental and dynamical features of consolidation. A computational model of molecular processes underlying consolidation following inhibitory avoidance (IA) training in rats was developed. Differential equations described the actions of CaMKIIα, multiple feedback loops regulating BDNF expression, and several transcription factors including methyl-CpG binding protein 2 (MeCP2), histone deacetylase 2 (HDAC2), and SIN3 transcription regulator family member A (Sin3a). This model provides novel explanations for the (apparent) rapid forgetting of infantile memory and the temporal progression of memory consolidation in adults. Simulations predict that dual effects of MeCP2 on the expression of bdnf, and interaction between MeCP2 and CaMKIIα, play critical roles in the rapid forgetting of infantile memory and the progress of memory resistance to disruptions. These insights suggest new potential targets of therapy for memory impairment.
Collapse
Affiliation(s)
- Yili Zhang
- Department of Neurobiology and Anatomy; W.M. Keck Center for the Neurobiology of Learning and Memory; The University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Paul Smolen
- Department of Neurobiology and Anatomy; W.M. Keck Center for the Neurobiology of Learning and Memory; The University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Cristina M. Alberini
- Center for Neural Science, New York University, New York City, New York, United States of America
| | - Douglas A. Baxter
- Department of Neurobiology and Anatomy; W.M. Keck Center for the Neurobiology of Learning and Memory; The University of Texas Medical School at Houston, Houston, Texas, United States of America
- Department of Neurobiology and Experimental Therapeutics, College of Medicine, Texas A&M University, Houston, Texas, United States of America
| | - John H. Byrne
- Department of Neurobiology and Anatomy; W.M. Keck Center for the Neurobiology of Learning and Memory; The University of Texas Medical School at Houston, Houston, Texas, United States of America
| |
Collapse
|
18
|
Shrestha P, Klann E. Spatiotemporally resolved protein synthesis as a molecular framework for memory consolidation. Trends Neurosci 2022; 45:297-311. [PMID: 35184897 PMCID: PMC8930706 DOI: 10.1016/j.tins.2022.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 01/25/2023]
Abstract
De novo protein synthesis is required for long-term memory consolidation. Dynamic regulation of protein synthesis occurs via a complex interplay of translation factors and modulators. Many components of the protein synthesis machinery have been targeted either pharmacologically or genetically to establish its requirement for memory. The combination of ligand/light-gating and genetic strategies, that is, chemogenetics and optogenetics, has begun to reveal the spatiotemporal resolution of protein synthesis in specific cell types during memory consolidation. This review summarizes current knowledge of the macroscopic and microscopic neural substrates for protein synthesis in memory consolidation. In addition, we highlight future directions for determining the localization and timing of de novo protein synthesis for memory consolidation with tools that permit unprecedented spatiotemporal precision.
Collapse
Affiliation(s)
- Prerana Shrestha
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA.
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10012, USA; NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
19
|
Hippocampal neurons' cytosolic and membrane-bound ribosomal transcript profiles are differentially regulated by learning and subsequent sleep. Proc Natl Acad Sci U S A 2021; 118:2108534118. [PMID: 34819370 PMCID: PMC8640746 DOI: 10.1073/pnas.2108534118] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/25/2022] Open
Abstract
Sleep loss disrupts consolidation of hippocampus-dependent memory. To understand the cellular basis for this effect, we quantified RNAs associated with translating ribosomes in cytosol and on cellular membranes of different hippocampal neuron populations. Our analysis suggests that while sleep loss (but not learning) alters numerous ribosomal transcripts in cytosol, learning has dramatic effects on transcript profiles for less–well-characterized membrane-bound ribosomes. We demonstrate that postlearning sleep deprivation occludes already minimal learning-driven changes on cytosolic ribosomes. It simultaneously alters transcripts associated with metabolic and biosynthetic processes in membrane-bound ribosomes in excitatory hippocampal neurons and highly active, putative “engram” neurons, respectively. Together, these findings provide insights into the cellular mechanisms altered by learning and their disruption by subsequent sleep loss. The hippocampus is essential for consolidating transient experiences into long-lasting memories. Memory consolidation is facilitated by postlearning sleep, although the underlying cellular mechanisms are largely unknown. We took an unbiased approach to this question by using a mouse model of hippocampally mediated, sleep-dependent memory consolidation (contextual fear memory). Because synaptic plasticity is associated with changes to both neuronal cell membranes (e.g., receptors) and cytosol (e.g., cytoskeletal elements), we characterized how these cell compartments are affected by learning and subsequent sleep or sleep deprivation (SD). Translating ribosome affinity purification was used to profile ribosome-associated RNAs in different subcellular compartments (cytosol and membrane) and in different cell populations (whole hippocampus, Camk2a+ neurons, or highly active neurons with phosphorylated ribosomal subunit S6 [pS6+]). We examined how transcript profiles change as a function of sleep versus SD and prior learning (contextual fear conditioning; CFC). While sleep loss altered many cytosolic ribosomal transcripts, CFC altered almost none, and CFC-driven changes were occluded by subsequent SD. In striking contrast, SD altered few transcripts on membrane-bound (MB) ribosomes, while learning altered many more (including long non-coding RNAs [lncRNAs]). The cellular pathways most affected by CFC were involved in structural remodeling. Comparisons of post-CFC MB transcript profiles between sleeping and SD mice implicated changes in cellular metabolism in Camk2a+ neurons and protein synthesis in highly active pS6+ (putative “engram”) neurons as biological processes disrupted by SD. These findings provide insights into how learning affects hippocampal neurons and suggest that the effects of SD on memory consolidation are cell type and subcellular compartment specific.
Collapse
|
20
|
Hernández-Matias A, Bermúdez-Rattoni F, Osorio-Gómez D. Maintenance of conditioned place avoidance induced by gastric malaise requires NMDA activity within the ventral hippocampus. ACTA ACUST UNITED AC 2021; 28:270-276. [PMID: 34400528 PMCID: PMC8372560 DOI: 10.1101/lm.052720.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/28/2021] [Indexed: 11/24/2022]
Abstract
It has been reported that during chemotherapy treatment, some patients can experience nausea before pharmacological administration, suggesting that contextual stimuli are associated with the nauseating effects. There are attempts to reproduce with animal models the conditions under which this phenomenon is observed to provide a useful paradigm for studying contextual aversion learning and the brain structures involved. This manuscript assessed the hippocampus involvement in acquiring and maintaining long-term conditioned place avoidance (CPA) induced by a gastric malaise-inducing agent, LiCl. Our results demonstrate that a reliable induction of CPA is possible after one acquisition trial. However, CPA establishment requires a 20-min confinement in the compartment associated with LiCl administration. Interestingly, both hippocampal regions seem to be necessary for CPA establishment; nonetheless, inactivation of the ventral hippocampus results in a reversion of avoidance and turns it into preference. Moreover, we demonstrate that activation of dorsal/ventral hippocampal NMDA receptors after CS–US association is required for long-term CPA memory maintenance.
Collapse
Affiliation(s)
- Arturo Hernández-Matias
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Daniel Osorio-Gómez
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| |
Collapse
|
21
|
Garay PM, Chen A, Tsukahara T, Rodríguez Díaz JC, Kohen R, Althaus JC, Wallner MA, Giger RJ, Jones KS, Sutton MA, Iwase S. RAI1 Regulates Activity-Dependent Nascent Transcription and Synaptic Scaling. Cell Rep 2021; 32:108002. [PMID: 32783930 PMCID: PMC7418709 DOI: 10.1016/j.celrep.2020.108002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/17/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Long-lasting forms of synaptic plasticity such as synaptic scaling are critically dependent on transcription. Activity-dependent transcriptional dynamics in neurons, however, remain incompletely characterized because most previous efforts relied on measurement of steady-state mRNAs. Here, we use nascent RNA sequencing to profile transcriptional dynamics of primary neuron cultures undergoing network activity shifts. We find pervasive transcriptional changes, in which ∼45% of expressed genes respond to network activity shifts. We further link retinoic acid-induced 1 (RAI1), the Smith-Magenis syndrome gene, to the transcriptional program driven by reduced network activity. Remarkable agreement among nascent transcriptomes, dynamic chromatin occupancy of RAI1, and electrophysiological properties of Rai1-deficient neurons demonstrates the essential roles of RAI1 in suppressing synaptic upscaling in the naive network, while promoting upscaling triggered by activity silencing. These results highlight the utility of bona fide transcription profiling to discover mechanisms of activity-dependent chromatin remodeling that underlie normal and pathological synaptic plasticity. BrU-seq reveals nascent transcription during synaptic up- or downscaling Smith-Magenis syndrome protein RAI1 regulates upscaling-associated transcription RAI1 departs chromatin in response to network activity shifts RAI1 blocks upscaling in naive networks and promotes inactivity-induced upscaling
Collapse
Affiliation(s)
- Patricia M Garay
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alex Chen
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Takao Tsukahara
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Rafi Kohen
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J Christian Althaus
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margarete A Wallner
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Roman J Giger
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Kevin S Jones
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Michael A Sutton
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Shigeki Iwase
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Mizuno I, Matsuda S. The role of endocannabinoids in consolidation, retrieval, reconsolidation, and extinction of fear memory. Pharmacol Rep 2021; 73:984-1003. [PMID: 33954935 DOI: 10.1007/s43440-021-00246-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/07/2021] [Indexed: 12/19/2022]
Abstract
Endocannabinoids are involved in various physiological functions, including synaptic plasticity and memory, and some psychiatric disorders, such as posttraumatic stress disorder (PTSD), through the activation of cannabinoid (CB) receptors. Patients with PTSD often show excessive fear memory and impairment of fear extinction (FE). It has been reported that the stability of acquired fear memory is altered through multiple memory stages, such as consolidation and reconsolidation. FE also affects the stability of fear memory. Each stage of fear memory formation and FE are regulated by different molecular mechanisms, including the CB system. However, to the best of our knowledge, no review summarizes the role of the CB system during each stage of fear memory formation and FE. In this review, we summarize the roles of endocannabinoids in fear memory formation and FE. Moreover, based on the summary, we propose a new hypothesis for the role of endocannabinoids in fear regulation, and discuss treatment for PTSD using CB system-related drugs.
Collapse
Affiliation(s)
- Ikumi Mizuno
- Department of Pharmacotherapeutics, Showa Pharmaceutical University, 3-3165, Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Shingo Matsuda
- Department of Pharmacotherapeutics, Showa Pharmaceutical University, 3-3165, Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan. .,Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba, Chiba, 260-8670, Japan. .,Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
23
|
Buurstede JC, van Weert LTCM, Colucci P, Gentenaar M, Viho EMG, Koorneef LL, Schoonderwoerd RA, Lanooij SD, Moustakas I, Balog J, Mei H, Kielbasa SM, Campolongo P, Roozendaal B, Meijer OC. Hippocampal glucocorticoid target genes associated with enhancement of memory consolidation. Eur J Neurosci 2021; 55:2666-2683. [PMID: 33840130 PMCID: PMC9292385 DOI: 10.1111/ejn.15226] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/04/2021] [Indexed: 12/16/2022]
Abstract
Glucocorticoids enhance memory consolidation of emotionally arousing events via largely unknown molecular mechanisms. This glucocorticoid effect on the consolidation process also requires central noradrenergic neurotransmission. The intracellular pathways of these two stress mediators converge on two transcription factors: the glucocorticoid receptor (GR) and phosphorylated cAMP response element‐binding protein (pCREB). We therefore investigated, in male rats, whether glucocorticoid effects on memory are associated with genomic interactions between the GR and pCREB in the hippocampus. In a two‐by‐two design, object exploration training or no training was combined with post‐training administration of a memory‐enhancing dose of corticosterone or vehicle. Genomic effects were studied by chromatin immunoprecipitation followed by sequencing (ChIP‐seq) of GR and pCREB 45 min after training and transcriptome analysis after 3 hr. Corticosterone administration induced differential GR DNA‐binding and regulation of target genes within the hippocampus, largely independent of training. Training alone did not result in long‐term memory nor did it affect GR or pCREB DNA‐binding and gene expression. No strong evidence was found for an interaction between GR and pCREB. Combination of the GR DNA‐binding and transcriptome data identified a set of novel, likely direct, GR target genes that are candidate mediators of corticosterone effects on memory consolidation. Cell‐specific expression of the identified target genes using single‐cell expression data suggests that the effects of corticosterone reflect in part non‐neuronal cells. Together, our data identified new GR targets associated with memory consolidation that reflect effects in both neuronal and non‐neuronal cells.
Collapse
Affiliation(s)
- Jacobus C Buurstede
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa T C M van Weert
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Paola Colucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Max Gentenaar
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eva M G Viho
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa L Koorneef
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Robin A Schoonderwoerd
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne D Lanooij
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Ioannis Moustakas
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Judit Balog
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, Leiden, The Netherlands
| | - Szymon M Kielbasa
- Department of Medical Statistics and Bioinformatics, Bioinformatics Center of Expertise, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Benno Roozendaal
- Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Onno C Meijer
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Molecular and cellular mechanisms of engram allocation and maintenance. Brain Res Bull 2021; 170:274-282. [PMID: 33647419 DOI: 10.1016/j.brainresbull.2021.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/29/2021] [Accepted: 02/18/2021] [Indexed: 01/19/2023]
Abstract
Understanding how we learn and remember has been a long-standing question in neuroscience. Technological developments of the past 15 years have allowed for dramatically increased access to the neurons that hold the physical representation of memory, also known as a memory trace or engram. Such developments have tremendously facilitated advancement of the memory field, since they made possible interrogation of the cellular and molecular mechanisms underlying memory formation with unprecedented cellular specificity. Here, we discuss the studies that have investigated rules governing neuronal recruitment to a particular memory engram. Furthermore, we provide an overview of the evidence that functional and structural changes associated with memory consolidation occur in engram neurons. Moreover, we summarize the expanding literature showing that transcriptional regulatory factors such as transcription factors and epigenetic mechanisms play an important role in the maintained allocation of behaviorally-selected neurons to an engram. Together, these studies have begun elucidating how neuronal networks are selected and modified in order to support memory formation and storage.
Collapse
|
25
|
Kramar CP, Castillo-Díaz F, Gigante ED, Medina JH, Barbano MF. The late consolidation of an aversive memory is promoted by VTA dopamine release in the dorsal hippocampus. Eur J Neurosci 2021; 53:841-851. [PMID: 33617053 DOI: 10.1111/ejn.15076] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 11/30/2022]
Abstract
The hippocampus has been implicated in the processing and storage of aversive memories but the precise mechanisms by which these memories persist in time remain elusive. We have demonstrated that dopaminergic neurotransmission in the dorsal hippocampus regulates the long-term storage of both appetitive and aversive memories at a critical time point known as "late consolidation" (12 hr after the learning experience). This modulation appears to have opposite effects depending on the valence of the stimuli, with hippocampal dopamine release peaking immediately and 13-17 hr after a rewarding experience. Here, we determined the release pattern of hippocampal dopamine following an aversive experience, in order to better understand this opposite modulation process. We observed significant increases in dopamine levels at several times (6-8, 11-12, and 15 hr) after subjecting rats to a conditioned place aversion (CPA) task with the aversive agent lithium chloride (LiCl). Early pharmacological blockade of hippocampal DA receptors impaired CPA memory consolidation. In addition and consistent with previous findings showing that late post-training infusions of dopaminergic agents into the hippocampus modulate the long-term storage of aversive memories, we found that the photostimulation of dopaminergic VTA fibers in the dorsal hippocampus 11-12 hr after CPA training was enough to transform a short-lasting long-term memory into a long-lasting one. The fact that the persistence of an aversive memory can still be affected several hours after the learning experience opens new avenues to develop behavioral and pharmacological strategies for the treatment of a variety of mental disorders.
Collapse
Affiliation(s)
- Cecilia P Kramar
- Instituto de Biología Celular y Neurociencias (CONICET-UBA), Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - Fernando Castillo-Díaz
- Instituto de Biología Celular y Neurociencias (CONICET-UBA), Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - Eduardo D Gigante
- National Institute on Drug Abuse (NIDA/NIH), Neuronal Networks Section, Baltimore, MD, USA
| | - Jorge H Medina
- Instituto de Biología Celular y Neurociencias (CONICET-UBA), Facultad de Medicina, UBA, Buenos Aires, Argentina
| | - M Flavia Barbano
- Instituto de Biología Celular y Neurociencias (CONICET-UBA), Facultad de Medicina, UBA, Buenos Aires, Argentina.,National Institute on Drug Abuse (NIDA/NIH), Neuronal Networks Section, Baltimore, MD, USA
| |
Collapse
|
26
|
Reshetnikov VV, Kisaretova PE, Ershov NI, Shulyupova AS, Oshchepkov DY, Klimova NV, Ivanchihina AV, Merkulova TI, Bondar NP. Genes associated with cognitive performance in the Morris water maze: an RNA-seq study. Sci Rep 2020; 10:22078. [PMID: 33328525 PMCID: PMC7744575 DOI: 10.1038/s41598-020-78997-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Learning and memory are among higher-order cognitive functions that are based on numerous molecular processes including changes in the expression of genes. To identify genes associated with learning and memory formation, here, we used the RNA-seq (high-throughput mRNA sequencing) technology to compare hippocampal transcriptomes between mice with high and low Morris water maze (MWM) cognitive performance. We identified 88 differentially expressed genes (DEGs) and 24 differentially alternatively spliced transcripts between the high- and low-MWM-performance mice. Although the sets of DEGs and differentially alternatively spliced transcripts did not overlap, both were found to be enriched with genes related to the same type of biological processes: trans-synaptic signaling, cognition, and glutamatergic transmission. These findings were supported by the results of weighted-gene co-expression network analysis (WGCNA) revealing the enrichment of MWM-cognitive-performance-correlating gene modules with very similar Gene Ontology terms. High-MWM-performance mice manifested mostly higher expression of the genes associated with glutamatergic transmission and long-term potentiation implementation, which are processes necessary for memory acquisition and consolidation. In this set, there were genes participating in the regulation of trans-synaptic signaling, primarily AMPA receptor signaling (Nrn1, Nptx1, Homer3, Prkce, Napa, Camk2b, Syt7, and Nrgn) and calcium turnover (Hpca, Caln1, Orai2, Cpne4, and Cpne9). In high-MWM-performance mice, we also demonstrated significant upregulation of the “flip” splice variant of Gria1 and Gria2 transcripts encoding subunits of AMPA receptor. Altogether, our data helped to identify specific genes in the hippocampus that are associated with learning and long-term memory. We hypothesized that the differences in MWM cognitive performance between the mouse groups are linked with increased long-term potentiation, which is mainly mediated by increased glutamatergic transmission, primarily AMPA receptor signaling.
Collapse
Affiliation(s)
- Vasiliy V Reshetnikov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Polina E Kisaretova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Nikita I Ershov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Anastasia S Shulyupova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Dmitry Yu Oshchepkov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Natalia V Klimova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | | | - Tatiana I Merkulova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | - Natalia P Bondar
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, Russia. .,Novosibirsk State University, Novosibirsk, Russia.
| |
Collapse
|
27
|
Sleep Deprivation and Neurological Disorders. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5764017. [PMID: 33381558 PMCID: PMC7755475 DOI: 10.1155/2020/5764017] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
Sleep plays an important role in maintaining neuronal circuitry, signalling and helps maintain overall health and wellbeing. Sleep deprivation (SD) disturbs the circadian physiology and exerts a negative impact on brain and behavioural functions. SD impairs the cellular clearance of misfolded neurotoxin proteins like α-synuclein, amyloid-β, and tau which are involved in major neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. In addition, SD is also shown to affect the glymphatic system, a glial-dependent metabolic waste clearance pathway, causing accumulation of misfolded faulty proteins in synaptic compartments resulting in cognitive decline. Also, SD affects the immunological and redox system resulting in neuroinflammation and oxidative stress. Hence, it is important to understand the molecular and biochemical alterations that are the causative factors leading to these pathophysiological effects on the neuronal system. This review is an attempt in this direction. It provides up-to-date information on the alterations in the key processes, pathways, and proteins that are negatively affected by SD and become reasons for neurological disorders over a prolonged period of time, if left unattended.
Collapse
|
28
|
Manganiello-Terra FA, Correa-Netto NF, Masukawa MY, Ruzzi A, Linardi A, Santos-Junior JG. Inhaled Lavandula angustifolia essential oil enhances extinction learning and inhibits memory updating in mice submitted to the contextual fear conditioning. JOURNAL OF ETHNOPHARMACOLOGY 2020; 260:113048. [PMID: 32525067 DOI: 10.1016/j.jep.2020.113048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 05/04/2020] [Accepted: 05/29/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lavender (Lavandula angustifolia) essential oil (EO) has a long history of use in emotional illness, including anxiety disorders. Cognitive mechanisms of learning and memory play a pivotal role in the etiology and maintenance of anxiety since exposure to cues related to aversive situations induces high arousal and anticipatory anxiety. Memory become labile after its reactivation and can be modulated by reconsolidation or extinction. Inhibition of memory reconsolidation or facilitation of memory extinction may be effective in preventing or minimizing the effect of contextual cues on anticipatory anxiety. AIM OF THE STUDY We investigated the effect of Lavandula angustifolia EO in the memory updating of conditioned contextual fear. MATERIALS AND METHODS Adult male C57Bl6 mice were submitted to fear conditioning. Two days after conditioning the mice underwent a reactivation session in a hybrid context and were then immediately exposed to vaporized water or essential oil at concentrations of 1%, 2.5% or 5% for 3 h. Two days later, the mice were tested in the original or an altered context and their freezing behavior was measured. In addition, mice were subjected to a fear memory recovery protocol followed by a reinstatement session. RESULTS In the contextual fear test, 1% essential oil, but not 2.5% or 5%, reduced the freezing behavior response, whereas after a reinstatement session, exposure to 1% essential oil increased the freezing behavior response. CONCLUSIONS These results suggest that Lavandula angustifolia essential oil enhances memory extinction and, consequently, inhibits memory updating.
Collapse
Affiliation(s)
- Fabiana Aparecida Manganiello-Terra
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, 01221-020, São Paulo, SP, Brazil
| | - Nelson Francisco Correa-Netto
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, 01221-020, São Paulo, SP, Brazil
| | - Márcia Yuriko Masukawa
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, 01221-020, São Paulo, SP, Brazil
| | - André Ruzzi
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, 01221-020, São Paulo, SP, Brazil
| | - Alessandra Linardi
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, 01221-020, São Paulo, SP, Brazil
| | - Jair Guilherme Santos-Junior
- Department of Physiological Sciences, Santa Casa of São Paulo Medical School, Rua Cesário Mota Junior, 61, Vila Buarque, 01221-020, São Paulo, SP, Brazil.
| |
Collapse
|
29
|
Reikhardt BA, Shabanov PD. The Effect of Structural Analogues of Etimizole on Protein Kinase CK2, Protein Phosphorylation, and Transcription of Chromatin in Rat Cortical and Hippocampal Neurons. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2020. [DOI: 10.1134/s1990750820040101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Prado-Alcalá RA, González-Salinas S, Antaramián A, Quirarte GL, Bello-Medina PC, Medina AC. Imbalance in cerebral protein homeostasis: Effects on memory consolidation. Behav Brain Res 2020; 393:112767. [DOI: 10.1016/j.bbr.2020.112767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/15/2020] [Accepted: 06/07/2020] [Indexed: 01/29/2023]
|
31
|
Fabbrin SB, Girardi BA, de Lorena Wendel A, Coelho Ilha Valin C, Pillat MM, Viero FT, Mello CF, Rubin MA. Spermidine-induced improvement of memory consolidation involves PI3K/Akt signaling pathway. Brain Res Bull 2020; 164:208-213. [PMID: 32858125 DOI: 10.1016/j.brainresbull.2020.08.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 01/23/2023]
Abstract
Spermidine (SPD) is an endogenous polyamine that plays a facilitatory role in memory acquisition and consolidation. Memory consolidation occurs immediately after learning and again around 3-6 hours later. Current evidence indicates that the polyamine binding site at the NMDA receptor (NMDAr) mediates the effects of SPD on memory. While NMDAr activation increases brain-derived neurotrophic factor (BDNF) release, no study has investigated whether BDNF-activated signaling pathways, such as the phosphatidylinositol 3-kinase (PI3K)/Akt pathway play a role in SPD-induced improvement of memory consolidation. Therefore, the aim of the current study was to evaluate whether the TrkB receptor and the PI3K/Akt pathway are involved in the facilitatory effect of SPD on memory consolidation. Male Wistar rats were trained in the contextual conditioned fear task. SPD, ANA-12 (TrkB antagonist), and LY294002 (PI3K inhibitor) were administered immediately after training. The animals were tested 24 h after training. We found that SPD improved fear memory consolidation and that both ANA-12 and LY294002 prevented the facilitatory effect of SPD on memory. These results suggest that SPD-induced improvement of memory consolidation involves the activation of the TrkB receptor and PI3K/Akt pathway.
Collapse
Affiliation(s)
- Shaiana Beck Fabbrin
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center of Exact and Natural Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Bruna Amanda Girardi
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center of Exact and Natural Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Arithane de Lorena Wendel
- School of Pharmacy, Center of Health Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Carolina Coelho Ilha Valin
- School of Pharmacy, Center of Health Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Micheli Mainardi Pillat
- Graduate Program in Pharmacology, Center of Health Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Fernanda Tibolla Viero
- Graduate Program in Pharmacology, Center of Health Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Carlos Fernando Mello
- Graduate Program in Pharmacology, Center of Health Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil.
| | - Maribel Antonello Rubin
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center of Exact and Natural Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil; Graduate Program in Pharmacology, Center of Health Sciences, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
32
|
Kornhuber J, Zoicas I. Social Fear Memory Requires Two Stages of Protein Synthesis in Mice. Int J Mol Sci 2020; 21:ijms21155537. [PMID: 32748831 PMCID: PMC7432563 DOI: 10.3390/ijms21155537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
It is well known that long-term consolidation of newly acquired information, including information related to social fear, require de novo protein synthesis. However, the temporal dynamics of protein synthesis during the consolidation of social fear memories is unclear. To address this question, mice received a single systemic injection with the protein synthesis inhibitor, anisomycin, at different time-points before or after social fear conditioning (SFC), and memory was assessed 24 h later. We showed that anisomycin impaired the consolidation of social fear memories in a time-point-dependent manner. Mice that received anisomycin 20 min before, immediately after, 6 h, or 8 h after SFC showed reduced expression of social fear, indicating impaired social fear memory, whereas anisomycin caused no effects when administered 4 h after SFC. These results suggest that consolidation of social fear memories requires two stages of protein synthesis: (1) an initial stage starting during or immediately after SFC, and (2) a second stage starting around 6 h after SFC and lasting for at least 5 h.
Collapse
Affiliation(s)
- Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| | - Iulia Zoicas
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93040 Regensburg, Germany
- Correspondence: ; Tel.: +49-9131-85-46005
| |
Collapse
|
33
|
Koltun B, Ironi S, Gershoni-Emek N, Barrera I, Hleihil M, Nanguneri S, Sasmal R, Agasti SS, Nair D, Rosenblum K. Measuring mRNA translation in neuronal processes and somata by tRNA-FRET. Nucleic Acids Res 2020; 48:e32. [PMID: 31974573 PMCID: PMC7102941 DOI: 10.1093/nar/gkaa042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 12/04/2019] [Accepted: 01/15/2020] [Indexed: 01/12/2023] Open
Abstract
In neurons, the specific spatial and temporal localization of protein synthesis is of great importance for function and survival. Here, we visualized tRNA and protein synthesis events in fixed and live mouse primary cortical culture using fluorescently-labeled tRNAs. We were able to characterize the distribution and transport of tRNAs in different neuronal sub-compartments and to study their association with the ribosome. We found that tRNA mobility in neural processes is lower than in somata and corresponds to patterns of slow transport mechanisms, and that larger tRNA puncta co-localize with translational machinery components and are likely the functional fraction. Furthermore, chemical induction of long-term potentiation (LTP) in culture revealed up-regulation of mRNA translation with a similar effect in dendrites and somata, which appeared to be GluR-dependent 6 h post-activation. Importantly, measurement of protein synthesis in neurons with high resolutions offers new insights into neuronal function in health and disease states.
Collapse
Affiliation(s)
- Bella Koltun
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Sivan Ironi
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | | | - Iliana Barrera
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Mohammad Hleihil
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | | | - Ranjan Sasmal
- New Chemistry Unit and Chemistry & Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka, India
| | - Sarit S Agasti
- New Chemistry Unit and Chemistry & Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka, India
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Kobi Rosenblum
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel.,Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| |
Collapse
|
34
|
Reikhardt BA, Shabanov PD. [Effect of etimizole structural analogues on protein kinase CK2, protein phosphorylation and transcription of chromatin in rat brain cortex and hippocampus]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:130-137. [PMID: 32420893 DOI: 10.18097/pbmc20206602130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Protein kinase CK2 is an important enzyme in the nervous system. The nuclear forms of CK2 regulate chromatin structure and gene expression, the key processes for long-term memory formation. Memory modulators, the Structural Analogues of Etimizole (SAE), were able to increase or decrease the activity of chromatin-associated CK in the cortex and hippocampus of rat brain in vitro. In vivo memory enhancers from SAE-group (3 mg/kg) stimulated CK2 activity and the transcriptional ability of chromatin in the cortex and hippocampus, starting from 30 min with a peak for 60 min and a duration up to 180 min. At these periods the memory inhibitor from the SAE-group reduced CK2 activity and chromatin transcription. It is assumed that the modulating effect of SAE on CK2 activity and transcription underlies the effects of these compounds on long-term memory.
Collapse
Affiliation(s)
- B A Reikhardt
- Institute of Experimental Medicine, St. Petersburg, Russia
| | - P D Shabanov
- Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
35
|
Jeon SG, Yoo A, Chun DW, Hong SB, Chung H, Kim JI, Moon M. The Critical Role of Nurr1 as a Mediator and Therapeutic Target in Alzheimer's Disease-related Pathogenesis. Aging Dis 2020; 11:705-724. [PMID: 32489714 PMCID: PMC7220289 DOI: 10.14336/ad.2019.0718] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/18/2019] [Indexed: 01/16/2023] Open
Abstract
Several studies have revealed that the transcription factor nuclear receptor related 1 (Nurr1) plays several roles not only in the regulation of gene expression related to dopamine synthesis, but also in alternative splicing, and miRNA targeting. Moreover, it regulates cognitive functions and protects against inflammation-induced neuronal death. In particular, the role of Nurr1 in the pathogenesis of Parkinson's disease (PD) has been well investigated; for example, it has been shown that it restores behavioral and histological impairments in PD models. Although many studies have evaluated the connection between Nurr1 and PD pathogenesis, the role of Nurr1 in Alzheimer's disease (AD) remain to be studied. There have been several studies describing Nurr1 protein expression in the AD brain. However, only a few studies have examined the role of Nurr1 in the context of AD. Therefore, in this review, we highlight the overall effects of Nurr1 under the neuropathologic conditions related to AD. Furthermore, we suggest the possibility of using Nurr1 as a therapeutic target for AD or other neurodegenerative disorders.
Collapse
Affiliation(s)
- Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Anji Yoo
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Dong Wook Chun
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Hyunju Chung
- Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Jin-il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si 63243, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| |
Collapse
|
36
|
A time-dependent role for the transcription factor CREB in neuronal allocation to an engram underlying a fear memory revealed using a novel in vivo optogenetic tool to modulate CREB function. Neuropsychopharmacology 2020; 45:916-924. [PMID: 31837649 PMCID: PMC7162924 DOI: 10.1038/s41386-019-0588-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/08/2019] [Accepted: 12/04/2019] [Indexed: 12/21/2022]
Abstract
The internal representation of an experience is thought to be encoded by long-lasting physical changes to the brain ("engrams") . Previously, we and others showed within the lateral amygdala (LA), a region critical for auditory conditioned fear, eligible neurons compete against one other for allocation to an engram. Neurons with relatively higher function of the transcription factor CREB were more likely to be allocated to the engram. In these studies, though, CREB function was artificially increased for several days before training. Precisely when increased CREB function is important for allocation remains an unanswered question. Here, we took advantage of a novel optogenetic tool (opto-DN-CREB) to gain spatial and temporal control of CREB function in freely behaving mice. We found increasing CREB function in a small, random population of LA principal neurons in the minutes, but not 24 h, before training was sufficient to enhance memory, likely because these neurons were preferentially allocated to the underlying engram. However, similarly increasing CREB activity in a small population of random LA neurons immediately after training disrupted subsequent memory retrieval, likely by disrupting the precise spatial and temporal patterns of offline post-training neuronal activity and/or function required for consolidation. These findings reveal the importance of the timing of CREB activity in regulating allocation and subsequent memory retrieval, and further, highlight the potential of optogenetic approaches to control protein function with temporal specificity in behaving animals.
Collapse
|
37
|
Molecular Mechanisms in Hippocampus Involved on Object Recognition Memory Consolidation and Reconsolidation. Neuroscience 2020; 435:112-123. [PMID: 32272151 DOI: 10.1016/j.neuroscience.2020.03.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/01/2020] [Accepted: 03/31/2020] [Indexed: 11/20/2022]
Abstract
Acquired information is stabilized into long-term memory through a process known as consolidation. Though, after consolidation, when stored information is retrieved they can be again susceptible, allowing modification, updating and strengthening and to be re-stabilized they need a new process referred to as memory reconsolidation. However, the molecular mechanisms of recognition memory consolidation and reconsolidation are not fully understood. Also, considering that the study of the link between synaptic proteins is key to understanding of memory processes, we investigated, in male Wistar rats, molecular mechanisms in the hippocampus involved on object recognition memory (ORM) consolidation and reconsolidation. We verified that the blockade of AMPA receptors (AMPAr) and L-VDCCs calcium channels impaired ORM consolidation and reconsolidation when administered into CA1 immediately after sample phase or reactivation phase and that these impairments were blocked by the administration of AMPAr agonist and of neurotrophin BDNF. Also, the blockade of CaMKII impaired ORM consolidation when administered 3 h after sample phase but had no effect on ORM reconsolidation and its effect was blocked by the administration of BDNF, but not of AMPAr agonist. So, this study provides new evidence of the molecular mechanisms involved on the consolidation and reconsolidation of ORM, demonstrating that AMPAr and L-VDCCs are necessary for the consolidation and reconsolidation of ORM while CaMKII is necessary only for the consolidation and also that there is a link between BDNF and AMPAr, L-VDCCs and CaMKII as well as a link between AMPAr and L-VDCCs on ORM consolidation and reconsolidation.
Collapse
|
38
|
Bridi M, Schoch H, Florian C, Poplawski SG, Banerjee A, Hawk JD, Porcari GS, Lejards C, Hahn CG, Giese KP, Havekes R, Spruston N, Abel T. Transcriptional corepressor SIN3A regulates hippocampal synaptic plasticity via Homer1/mGluR5 signaling. JCI Insight 2020; 5:92385. [PMID: 32069266 DOI: 10.1172/jci.insight.92385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Long-term memory depends on the control of activity-dependent neuronal gene expression, which is regulated by epigenetic modifications. The epigenetic modification of histones is orchestrated by the opposing activities of 2 classes of regulatory complexes: permissive coactivators and silencing corepressors. Much work has focused on coactivator complexes, but little is known about the corepressor complexes that suppress the expression of plasticity-related genes. Here, we define a critical role for the corepressor SIN3A in memory and synaptic plasticity, showing that postnatal neuronal deletion of Sin3a enhances hippocampal long-term potentiation and long-term contextual fear memory. SIN3A regulates the expression of genes encoding proteins in the postsynaptic density. Loss of SIN3A increases expression of the synaptic scaffold Homer1, alters the metabotropic glutamate receptor 1α (mGluR1α) and mGluR5 dependence of long-term potentiation, and increases activation of ERK in the hippocampus after learning. Our studies define a critical role for corepressors in modulating neural plasticity and memory consolidation and reveal that Homer1/mGluR signaling pathways may be central molecular mechanisms for memory enhancement.
Collapse
Affiliation(s)
| | | | | | | | - Anamika Banerjee
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | - Chang-Gyu Hahn
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Nelson Spruston
- Howard Hughes Medical Institute (HHMI) Janelia Research Campus, Ashburn, Virginia, USA
| | | |
Collapse
|
39
|
Raymundi AM, da Silva TR, Zampronio AR, Guimarães FS, Bertoglio LJ, Stern CAJ. A time-dependent contribution of hippocampal CB 1 , CB 2 and PPARγ receptors to cannabidiol-induced disruption of fear memory consolidation. Br J Pharmacol 2020; 177:945-957. [PMID: 31648363 DOI: 10.1111/bph.14895] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 09/03/2019] [Accepted: 09/30/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE In preclinical studies, cannabidiol (CBD) mitigates fear memories by facilitating their extinction or interfering with their generalization and reconsolidation. The brain regions and mechanisms underlying these effects, and their temporal window, are still poorly understood. Here, we have investigated related questions in the dorsal hippocampus (DH) during contextual fear consolidation. EXPERIMENTAL APPROACH Adult male Wistar rats received CBD (10-30 pmol) intra-DH immediately, 1 or 3 hr after fear conditioning. Effects of CBD on consolidation were inferred behaviourally and by analysing expression of the activity-regulated, cytoskeleton-associated (Arc) protein. The contribution of anandamide, CB1 , CB2 , 5-HT1A , A2A , and PPARγ receptors was also assessed. KEY RESULTS CBD impaired memory consolidation when given immediately or 1 hr after fear conditioning, but not after 3 hr. Expression of Arc protein in DH was reduced by systemic CBD treatment in both cases. Immediately after fear conditioning, CBD effects were abolished by CB1 or CB2 receptor blockade, partly reduced by 5-HT1A or A2A antagonism, and remained unchanged after antagonism of PPARγ receptors. One hour after fear conditioning, CBD effects were prevented only by PPARγ receptor antagonism. Also, inhibition of fatty acid amide hydrolase by URB597, impaired memory consolidation when infused immediately, but not 1 hr after fear conditioning. CONCLUSIONS AND IMPLICATIONS CBD disrupts memory consolidation up to 1 hr after fear conditioning, allowing an extended window of opportunity to mitigate aversive memories after their acquisition. Our results suggest time-dependent participation of anandamide, CB1 , CB2 and PPARγ receptors in the DH, during this process.
Collapse
Affiliation(s)
- Ana Maria Raymundi
- Department of Pharmacology, Federal University of Parana, Curitiba, Brazil
| | - Thiago R da Silva
- Department of Pharmacology, Federal University of Parana, Curitiba, Brazil
| | | | | | - Leandro J Bertoglio
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Cristina A J Stern
- Department of Pharmacology, Federal University of Parana, Curitiba, Brazil
| |
Collapse
|
40
|
Cell-type-specific drug-inducible protein synthesis inhibition demonstrates that memory consolidation requires rapid neuronal translation. Nat Neurosci 2020; 23:281-292. [PMID: 31959934 PMCID: PMC7147976 DOI: 10.1038/s41593-019-0568-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 12/05/2019] [Indexed: 12/04/2022]
Abstract
New protein synthesis is known to be required for the consolidation of memories, yet existing methods to block translation lack spatiotemporal precision and cell-type specificity, preventing investigation of cell-specific contributions of protein synthesis. Here, we developed a combined knock-in mouse and chemogenetic approach for cell type-specific and drug-inducible protein synthesis inhibition (ciPSI) that enables rapid and reversible phosphorylation of eIF2α, leading to inhibition of general translation by 50% in vivo. We use ciPSI to show that targeted protein synthesis inhibition pan-neuronally and in excitatory neurons in lateral amygdala (LA) impaired long-term memory. This could be recovered with artificial chemogenetic activation of LA neurons, though at the cost of stimulus generalization. Conversely, genetically reducing phosphorylation of eIF2α in excitatory neurons in LA enhanced memory strength, but reduced memory fidelity and behavioral flexibility. Our findings provide evidence for a cell-specific translation program during consolidation of threat memories.
Collapse
|
41
|
Osorio-Gómez D, Bermúdez-Rattoni F, Guzmán-Ramos K. Artificial taste avoidance memory induced by coactivation of NMDA and β-adrenergic receptors in the amygdala. Behav Brain Res 2019; 376:112193. [PMID: 31473281 DOI: 10.1016/j.bbr.2019.112193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/07/2019] [Accepted: 08/28/2019] [Indexed: 11/18/2022]
Abstract
The association between a taste and gastric malaise allows animals to avoid the ingestion of potentially toxic food. This association has been termed conditioned taste aversion (CTA) and relies on the activity of key brain structures such as the amygdala and the insular cortex. The establishment of this gustatory-avoidance memory is related to glutamatergic and noradrenergic activity within the amygdala during two crucial events: gastric malaise (unconditioned stimulus, US) and the post-acquisition spontaneous activity related to the association of both stimuli. To understand the functional implications of these neurochemical changes on avoidance memory formation, we assessed the effects of pharmacological stimulation of β-adrenergic and glutamatergic NMDA receptors through the administration of a mixture of L-homocysteic acid and isoproterenol into the amygdala after saccharin exposure on specific times to emulate the US and post-acquisition local signals that would be occurring naturally under CTA training. Our results show that activation of NMDA and β-adrenergic receptors generated a long-term avoidance response to saccharin, like a naturally induced rejection with LiCl. Moreover, the behavioral outcome was accompanied by changes in glutamate, norepinephrine and dopamine levels within the insular cortex, analogous to those displayed during memory retrieval of taste aversion memory. Therefore, we suggest that taste avoidance memory can be induced artificially through the emulation of specific amygdalar neurochemical signals, promoting changes in the amygdala-insular cortex circuit enabling memory establishment.
Collapse
Affiliation(s)
- Daniel Osorio-Gómez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Mexico City, Mexico
| | - Kioko Guzmán-Ramos
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud Universidad Autónoma Metropolitana, Unidad Lerma Av. de las Garzas No. 10, Col. El Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico.
| |
Collapse
|
42
|
Tohyama S, Matsuda S, Mizutani A. Sex-dependent opposite effects of a tropomyosin-related kinase B receptor (TrkB) agonist 7,8-dihydroxyflavone on cued fear extinction in mice. Neurosci Lett 2019; 715:134670. [PMID: 31805374 DOI: 10.1016/j.neulet.2019.134670] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/16/2019] [Accepted: 11/30/2019] [Indexed: 12/22/2022]
Abstract
Tropomyosin-related kinase B receptor (TrkB) is one of the new candidate receptors for drugs targeting psychiatric and neurodegenerative disorders. Recently, 7,8-dihydroxyflavone (7,8-DHF) has been identified as a selective TrkB agonist that crosses the blood-brain barrier after oral or intraperitoneal administration, and it enhances cued fear extinction in male rodents. However, its effects on females remain unclear. Preclinical research including both sexes is important for the development of treatment, particularly, for stress-related disorders such as post-traumatic stress disorder because such disorders are more prevalent in women. Therefore, we investigated the effects of 7,8-DHF on cued and contextual fear extinction in both male and female mice. Here we demonstrated that the administration of 7,8-DHF before each extinction session attenuated cued fear extinction in females; conversely, it enhanced cued fear extinction in males. However, administration of 7,8-DHF immediately after each extinction session did not affect cued fear extinction in either sex. Moreover, in contextual fear extinction, administration of 7,8-DHF before each extinction session did not affect fear extinction in either sex. Thus, 7,8-DHF showed sex-dependent opposite effects on cued fear extinction in mice when administered before but not immediately after each extinction session. Our results could contribute to the development of pharmacotherapy involving 7,8-DHF, particularly for stress-related disorders.
Collapse
Affiliation(s)
- Suguru Tohyama
- Department of Pharmacotherapeutics, Showa Pharmaceutical University, 3-3165, Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| | - Shingo Matsuda
- Department of Pharmacotherapeutics, Showa Pharmaceutical University, 3-3165, Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan; Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba, Chiba 260-8670, Japan; Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo 187-8502, Japan.
| | - Akihiro Mizutani
- Department of Pharmacotherapeutics, Showa Pharmaceutical University, 3-3165, Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan
| |
Collapse
|
43
|
Inhibition of transcription and translation in dorsal hippocampus does not interfere with consolidation of memory of intense training. Neurobiol Learn Mem 2019; 166:107092. [DOI: 10.1016/j.nlm.2019.107092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/23/2019] [Accepted: 09/14/2019] [Indexed: 01/01/2023]
|
44
|
Robinson HA, Pozzo-Miller L. The role of MeCP2 in learning and memory. ACTA ACUST UNITED AC 2019; 26:343-350. [PMID: 31416907 PMCID: PMC6699413 DOI: 10.1101/lm.048876.118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/21/2019] [Indexed: 01/31/2023]
Abstract
Gene transcription is a crucial step in the sequence of molecular, synaptic, cellular, and systems mechanisms underlying learning and memory. Here, we review the experimental evidence demonstrating that alterations in the levels and functionality of the methylated DNA-binding transcriptional regulator MeCP2 are implicated in the learning and memory deficits present in mouse models of Rett syndrome and MECP2 duplication syndrome. The significant impact that MeCP2 has on gene transcription through a variety of mechanisms, combined with well-defined models of learning and memory, make MeCP2 an excellent candidate to exemplify the role of gene transcription in learning and memory. Together, these studies have strengthened the concept that precise control of activity-dependent gene transcription is a fundamental mechanism that ensures long-term adaptive behaviors necessary for the survival of individuals interacting with their congeners in an ever-changing environment.
Collapse
Affiliation(s)
- Holly A Robinson
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| |
Collapse
|
45
|
Histone acetylation determines transcription of atypical protein kinases in rat neurons. Sci Rep 2019; 9:4332. [PMID: 30867503 PMCID: PMC6416243 DOI: 10.1038/s41598-019-40823-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 02/11/2019] [Indexed: 01/19/2023] Open
Abstract
It is widely accepted that memory consolidation requires de-novo transcription of memory-related genes. Epigenetic modifications, particularly histone acetylation, may facilitate gene transcription, but their potential molecular targets are poorly characterized. In the current study, we addressed the question of epigenetic control of atypical protein kinases (aPKC) that are critically involved in memory consolidation and maintenance. We examined the patterns of expression of two aPKC genes (Prkci and Prkcz) in rat cultured cortical neurons treated with histone deacetylase inhibitors. Histone hyperacetylation in the promoter region of Prkci gene elicited direct activation of transcriptional machinery, resulting in increased production of PKCλ mRNA. In parallel, histone hyperacetylation in the upstream promoter of Prkcz gene led to appearance of the corresponding PKCζ transcripts that are almost absent in the brain in resting conditions. In contrast, histone hyperacetylation in the downstream promoter of Prkcz gene was accompanied by a decreased expression of the brain-specific PKMζ products. We showed that epigenetically-triggered differential expression of PKMζ and PKCζ mRNA depended on protein synthesis. Summarizing, our results suggest that genes, encoding memory-related aPKC, may represent the molecular targets for epigenetic regulation through posttranslational histone modifications.
Collapse
|
46
|
Eimerbrink M, Pendry R, Hodges S, Wiles J, Peterman J, White J, Hayes H, Chumley M, Boehm G. The α5-GABAAR inverse agonist MRK-016 upregulates hippocampal BDNF expression and prevents cognitive deficits in LPS-treated mice, despite elevations in hippocampal Aβ. Behav Brain Res 2019; 359:871-877. [DOI: 10.1016/j.bbr.2018.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/03/2018] [Accepted: 07/19/2018] [Indexed: 12/18/2022]
|
47
|
Estradiol modulation of the renin-angiotensin system and the regulation of fear extinction. Transl Psychiatry 2019; 9:36. [PMID: 30696810 PMCID: PMC6351608 DOI: 10.1038/s41398-019-0374-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/20/2018] [Accepted: 01/01/2019] [Indexed: 01/31/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is more prevalent in women than men, yet much remains to be determined regarding the mechanism underlying this sex difference. Clinical and preclinical studies have shown that low estradiol levels during extinction of fear conditioning in rodents (i.e., cue exposure therapy in humans) leads to poor extinction consolidation and increased fear during extinction recall. The renin-angiotensin system (RAS) is also associated with stress-related pathologies, and RAS antagonists can enhance extinction consolidation in males. However, less is known about how estradiol and the RAS converge to alter fear extinction consolidation in females. Since estradiol downregulates the RAS, we determined the role of surgically (via ovariectomy [OVX]) and pharmacologically (via the hormonal contraceptive [HC], levonorgestrel) clamping estradiol at low levels in female rats on fear-related behavior, serum estradiol and angiotensin II (Ang II) levels, and angiotensin II type I receptor (AT1R) binding in the brain. We then tested whether the AT1R antagonist losartan would alter fear-related behavior in an estradiol-dependent manner. We found that both OVX and HC treatment produced extinction consolidation deficits relative to intact female rats in proestrus (when estradiol levels are high), and that losartan treatment mitigated these deficits and reduced freezing. OVX, but not HC, altered AT1R ligand binding, though HC reduced estradiol and increased Ang II levels in plasma. These findings have significant clinical implications, indicating that administration of an AT1R antagonist, especially if estradiol levels are low, prior to an exposure therapy session may improve treatment outcomes in females.
Collapse
|
48
|
Penha Farias C, Guerino Furini CR, Godfried Nachtigall E, Kielbovicz Behling JA, Silva de Assis Brasil E, Bühler L, Izquierdo I, de Carvalho Myskiw J. Extinction learning with social support depends on protein synthesis in prefrontal cortex but not hippocampus. Proc Natl Acad Sci U S A 2019; 116:1765-1769. [PMID: 30635411 PMCID: PMC6358673 DOI: 10.1073/pnas.1815893116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extinction of contextual fear conditioning (CFC) in the presence of a familiar nonfearful conspecific (social support), such as that of others tasks, can occur regardless of whether the original memory is retrieved during the extinction training. Extinction with social support is blocked by the protein synthesis inhibitors anisomycin and rapamycin and by the inhibitor of gene expression 5,6-dichloro-1-β-d-ribofuranosylbenzimidazole infused immediately after extinction training into the ventromedial prefrontal cortex (vmPFC) but unlike regular CFC extinction not in the CA1 region of the dorsal hippocampus. So social support generates a form of learning that differs from extinction acquired without social support in terms of the brain structures involved. This finding may lead to a better understanding of the brain mechanisms involved in the social support of memories and in therapies for disorders related to dysfunctional fear memories. Thus, here we show that the consolidation of extinction memory with social support relies on vmPFC rather than hippocampus gene expression and ribosomal- and mammalian target of rapamycin-dependent protein synthesis. These results provide additional knowledge about the cellular mechanisms and brain structures involved on the effect of social support in changing behavior and fear extinction memory.
Collapse
Affiliation(s)
- Clarissa Penha Farias
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Cristiane Regina Guerino Furini
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Eduarda Godfried Nachtigall
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Jonny Anderson Kielbovicz Behling
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Eduardo Silva de Assis Brasil
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Letícia Bühler
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Ivan Izquierdo
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil;
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Jociane de Carvalho Myskiw
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil;
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
49
|
Affiliation(s)
- Andre Fischer
- Department for Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.
- Department for Systems Medicine and Brain Diseases, German Center for Neurodegenerative Diseases (DZNE) site Göttingen, Göttingen, Germany.
| |
Collapse
|
50
|
Rodríguez-Blanco LA, Rivera-Olvera A, Escobar ML. Consolidation of an aversive taste memory requires two rounds of transcriptional and epigenetic regulation in the insular cortex. Behav Brain Res 2019; 356:371-374. [PMID: 30219263 DOI: 10.1016/j.bbr.2018.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022]
Abstract
The current view of the neurobiology of learning and memory suggests that long-term memory (LTM) depends not only on the de novo protein synthesis but also on the synthesis of mRNA even hours after the acquisition of memory, as well as that the regulation of transcription through the histone acetylation is essential for the memory establishment. Our previous studies showed that protein synthesis inhibition around the time of training and 5-7 hours after acquisition in the insular cortex (IC) prevents the consolidation of conditioned taste aversion (CTA), a well-established learning and memory paradigm in which an animal learns to associate a novel taste with nausea. However, the participation of mRNA synthesis and the epigenetic regulation through histone acetylation in this process remains unexplored. In the present study we evaluated the effect of the inhibition of transcription as well as deacetylation of histones at two temporal windows on the consolidation of CTA. Thus, immediately or seven hours after CTA acquisition animals received a microinfusion of 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) or MS-275 in the IC, respectively. The present results show that transcription inhibition immediately and 7 h after acquisition impairs the CTA memory consolidation, whereas the inhibition of histone deacetylation strengths this memory at those temporal windows. These findings reveal that CTA memory requires recurrent rounds of transcriptional modulation events in the IC in order to consolidate this memory trace, demonstrating that transcriptional and epigenetic modulation substantially contribute to memory-consolidation-related functions performed by a neocortical area even several hours after memory acquisition.
Collapse
Affiliation(s)
- Luis Alfredo Rodríguez-Blanco
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, Mexico D.F., Mexico
| | - Alejandro Rivera-Olvera
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, Mexico D.F., Mexico
| | - Martha L Escobar
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, Mexico D.F., Mexico.
| |
Collapse
|