1
|
Kolibius LD, Josselyn SA, Hanslmayr S. On the origin of memory neurons in the human hippocampus. Trends Cogn Sci 2025; 29:421-433. [PMID: 40037964 DOI: 10.1016/j.tics.2025.01.013] [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: 09/03/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 03/06/2025]
Abstract
The hippocampus is essential for episodic memory, yet its coding mechanism remains debated. In humans, two main theories have been proposed: one suggests that concept neurons represent specific elements of an episode, while another posits a conjunctive code, where index neurons code the entire episode. Here, we integrate new findings of index neurons in humans and other animals with the concept-specific memory framework, proposing that concept neurons evolve from index neurons through overlapping memories. This process is supported by engram literature, which posits that neurons are allocated to a memory trace based on excitability and that reactivation induces excitability. By integrating these insights, we connect two historically disparate fields of neuroscience: engram research and human single neuron episodic memory research.
Collapse
Affiliation(s)
- Luca D Kolibius
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA.
| | - Sheena A Josselyn
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Simon Hanslmayr
- School of Psychology and Neuroscience and Centre for Neurotechnology, University of Glasgow, Glasgow, UK; Centre for Neurotechnology, University of Glasgow, Glasgow, UK.
| |
Collapse
|
2
|
Ghandour K, Haga T, Ohkawa N, Fung CCA, Nomoto M, Fayed MR, Asai H, Sato M, Fukai T, Inokuchi K. Parallel processing of past and future memories through reactivation and synaptic plasticity mechanisms during sleep. Nat Commun 2025; 16:3618. [PMID: 40295514 PMCID: PMC12037800 DOI: 10.1038/s41467-025-58860-w] [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: 06/17/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Every day, we experience new episodes and store new memories. Although memories are stored in corresponding engram cells, how different sets of engram cells are selected for current and next episodes, and how they create their memories, remains unclear. Here we show that in male mice, hippocampal CA1 neurons show an organized synchronous activity in prelearning home cage sleep that correlates with the learning ensembles only in engram cells, termed preconfigured ensembles. Moreover, after learning, a subset of nonengram cells develops population activity, which is constructed during postlearning offline periods, and then emerges to represent engram cells for new learning. Our model suggests a potential role of synaptic depression and scaling in the reorganization of the activity of nonengram cells. Together, our findings indicate that during offline periods there are two parallel processes occurring: conserving of past memories through reactivation, and preparation for upcoming ones through offline synaptic plasticity mechanisms.
Collapse
Affiliation(s)
- Khaled Ghandour
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Center initiative for training international researchers (CITIR), University of Toyama, Toyama, Japan
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Tatsuya Haga
- Neural Coding and Brain Computing unit, OIST, Okinawa, Japan
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology, Osaka, Japan
| | - Noriaki Ohkawa
- Research Center for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
| | - Chi Chung Alan Fung
- Neural Coding and Brain Computing unit, OIST, Okinawa, Japan
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, Hong Kong
| | - Masanori Nomoto
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Mostafa R Fayed
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Department of Pharmacology and Toxicology, Kafrelsheikh University, Kafr El Sheikh, Egypt
| | - Hirotaka Asai
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaaki Sato
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tomoki Fukai
- Neural Coding and Brain Computing unit, OIST, Okinawa, Japan
| | - Kaoru Inokuchi
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.
| |
Collapse
|
3
|
Dovek L, Nguyen AT, Green E, Santhakumar V. Differential Glutamatergic Inputs to Semilunar Granule Cells and Granule Cells Underscore Dentate Gyrus Projection Neuron Diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643192. [PMID: 40161709 PMCID: PMC11952520 DOI: 10.1101/2025.03.14.643192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Semilunar Granule Cells (SGCs) are sparse dentate gyrus projection neurons whose role in the dentate circuit, including pathway specific inputs, remains unknown. We report that SGCs receive more frequent spontaneous excitatory synaptic inputs than granule cells (GCs). Dual GC-SGC recordings identified that SGCs receive stronger medial entorhinal cortex and associational synaptic drive but lack short-term facilitation of lateral entorhinal cortex inputs observed in GCs. SGCs dendritic spine density in proximal and middle dendrites was greater than in GCs. However, the strength of commissural inputs and dendritic input integration, examined in passive morphometric simulations, were not different between cell types. Activity dependent labeling identified an overrepresentation of SGCs among neuronal ensembles in both mice trained in a spatial memory task and task naïve controls. The divergence of modality specific inputs to SGCs and GCs can enable parallel processing of information streams and expand the computational capacity of the dentate gyrus.
Collapse
Affiliation(s)
- Laura Dovek
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Anh-Tho Nguyen
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Emmanuel Green
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Vijayalakshmi Santhakumar
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| |
Collapse
|
4
|
Willems TS, Xiong H, Kessels HW, Lesuis SL. GluA1-containing AMPA receptors are necessary for sparse memory engram formation. Neurobiol Learn Mem 2025; 218:108031. [PMID: 39922481 DOI: 10.1016/j.nlm.2025.108031] [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: 11/20/2024] [Revised: 01/17/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Memory formation depends on the selective recruitment of neuronal ensembles into circuits known as engrams, which represent the physical substrate of memory. Sparse encoding of these ensembles is essential for memory specificity and efficiency. AMPA receptor (AMPAR) subunits, particularly GluA1, play a central role in synaptic plasticity, which underpins memory encoding. This study investigates how GluA1 expression influences the recruitment of neurons into memory engrams. Using global GluA1 knockout (GluA1KO) mice, localized knockout models, and contextual fear-conditioning paradigms, we evaluated the role of GluA1 in memory formation and engram sparsity. GluA1KO mice exhibited impaired short-term memory retention but preserved 24-hour contextual memory. Despite this, these mice displayed increased expression of the immediate early gene Arc in hippocampal neurons, indicative of a denser engram network. Electrophysiological analyses revealed reduced synaptic strength in GluA1-deficient neurons, irrespective of Arc expression. Localized GluA1 knockout in the hippocampus confirmed that GluA1 deficiency increases neuronal recruitment into engrams, disrupting the sparse encoding typically observed in wild-type mice. These findings demonstrate that GluA1-containing AMPARs constrain engram size, ensuring selective recruitment of neurons for efficient memory encoding. By regulating synaptic plasticity, GluA1 facilitates both the encoding and size of memory circuits. This study highlights the critical role of GluA1 in maintaining sparse engram formation and provides insight into mechanisms underlying memory deficits in conditions where synaptic composition is altered.
Collapse
Affiliation(s)
- Thije S Willems
- Department of Cell and Circuit Neuroscience, Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Hui Xiong
- Department of Cell and Circuit Neuroscience, Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Helmut W Kessels
- Department of Cell and Circuit Neuroscience, Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Sylvie L Lesuis
- Department of Cell and Circuit Neuroscience, Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Mueller-Buehl C, Pakusch J, Bader V, Winklhofer KF, Mark MD, Faissner A. Combined loss of brevican, neurocan, tenascin-C and tenascin-R leads to impaired fear retrieval due to perineuronal net loss. Sci Rep 2025; 15:5528. [PMID: 39953103 PMCID: PMC11828866 DOI: 10.1038/s41598-025-89580-2] [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: 10/22/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
In conditions such as neurodegenerative diseases, posttraumatic stress disorder (PTSD), addiction and spinal cord injuries, restricted synaptic plasticity hinders the formation of new neuronal connections, preventing the compensation and treatment of adverse behaviors. Perineuronal nets (PNNs) significantly restrict synaptic plasticity by inhibiting synapse formation. The digestion of PNNs has been associated with short-term cognitive improvements and reduced long-term memory, offering potential therapeutic benefits in PTSD. This study investigates the correlation between PNNs and fear memory processes in extracellular matrix (ECM) mutant mice, particularly focusing on the amygdala-medial prefrontal cortex (mPFC) circuit, which is crucial for fear memory generation and maintenance. Fear conditioning was conducted on mice lacking four key ECM-molecules: brevican, neurocan, tenascin-C and tenascin-R (4x KO). These mice exhibited severe impairments in memory consolidation, as evident by their inability to retrieve previously learned fear memories, coupled with reduced PNN density and disturbed synaptic integrity along their PNNs. Additionally, changes in neural activity in the basolateral amygdala (BL) and reductions in VGAT+ synaptic puncta in the amygdala-mPFC circuit were observed. In contrast, tenascin single KOs showed intact fear behavior and memory compared to their control groups. Impaired fear memory consolidation can be advantageous in certain conditions, such as PTSD, making the 4x KO mice an intriguing model for future fear conditioning studies and highlighting brevican, neurocan, Tnc, and Tnr as compelling targets for further investigation. This study underscores the significance of ECM regulation for synaptic organization and the potential of PNN modulation as a therapeutic target for fear memory-related conditions.
Collapse
Affiliation(s)
- Cornelius Mueller-Buehl
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Johanna Pakusch
- Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, D- 44780, Bochum, Germany
| | - Verian Bader
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, D-44780, Bochum, Germany
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Konstanze F Winklhofer
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, D-44780, Bochum, Germany
- Cluster of Excellence RESOLV, D-44780, Bochum, Germany
| | - Melanie D Mark
- Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, D- 44780, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, D-44780, Bochum, Germany.
| |
Collapse
|
6
|
Lesuis SL, Park S, Hoorn A, Rashid AJ, Mocle AJ, Salter EW, Vislavski S, Gray MT, Torelli AM, DeCristofaro A, Driever WPF, van der Stelt M, Zweifel LS, Collingridge GL, Lefebvre JL, Walters BJ, Frankland PW, Hill MN, Josselyn SA. Stress disrupts engram ensembles in lateral amygdala to generalize threat memory in mice. Cell 2025; 188:121-140.e20. [PMID: 39549697 PMCID: PMC11726195 DOI: 10.1016/j.cell.2024.10.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 08/25/2024] [Accepted: 10/17/2024] [Indexed: 11/18/2024]
Abstract
Stress induces aversive memory overgeneralization, a hallmark of many psychiatric disorders. Memories are encoded by a sparse ensemble of neurons active during an event (an engram ensemble). We examined the molecular and circuit processes mediating stress-induced threat memory overgeneralization in mice. Stress, acting via corticosterone, increased the density of engram ensembles supporting a threat memory in lateral amygdala, and this engram ensemble was reactivated by both specific and non-specific retrieval cues (generalized threat memory). Furthermore, we identified a critical role for endocannabinoids, acting retrogradely on parvalbumin-positive (PV+) lateral amygdala interneurons in the formation of a less-sparse engram and memory generalization induced by stress. Glucocorticoid receptor antagonists, endocannabinoid synthesis inhibitors, increasing PV+ neuronal activity, and knocking down cannabinoid receptors in lateral amygdala PV+ neurons restored threat memory specificity and a sparse engram in stressed mice. These findings offer insights into stress-induced memory alterations, providing potential therapeutic avenues for stress-related disorders.
Collapse
Affiliation(s)
- Sylvie L Lesuis
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Cellular and Computational Neuroscience, Swammerdam Institute for Life Science, Amsterdam Neuroscience, University of Amsterdam, 1090 GE Amsterdam, the Netherlands
| | - Sungmo Park
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Annelies Hoorn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Asim J Rashid
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Andrew J Mocle
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Eric W Salter
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, and TANZ Centre for Research in Neurodegenerative Diseases, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Stefan Vislavski
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Madison T Gray
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Angelica M Torelli
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Antonietta DeCristofaro
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Wouter P F Driever
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University and Oncode Institute, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, 2815 Eastlake Ave E Suite 200, Seattle, WA 98102, USA
| | - Graham L Collingridge
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, and TANZ Centre for Research in Neurodegenerative Diseases, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Julie L Lefebvre
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Brandon J Walters
- Department of Cell and Systems Biology, University of Toronto Mississauga, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Matthew N Hill
- Hotchkiss Brain Institute, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
7
|
Martz J, Shelton MA, Langen TJ, Srinivasan S, Seney ML, Kentner AC. Peripubertal antagonism of corticotropin-releasing factor receptor 1 results in sustained changes in behavioral plasticity and the transcriptomic profile of the amygdala. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.14.607957. [PMID: 39185241 PMCID: PMC11343213 DOI: 10.1101/2024.08.14.607957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Peripuberty is a significant period of neurodevelopment with long-lasting effects on the brain and behavior. Blocking type 1 corticotropin-releasing factor receptors (CRFR1) in neonatal and peripubertal rats attenuates detrimental effects of early-life stress on neural plasticity, behavior, and stress hormone action, long after exposure to the drug has ended. CRFR1 antagonism can also impact neural and behavioral development in the absence of stressful stimuli, suggesting sustained alterations under baseline conditions. To investigate this further, we administered the CRFR1 antagonist (CRFR1a) R121919 to young adolescent male and female rats across 4 days. Following each treatment, rats were tested for locomotion, social behavior, mechanical allodynia, or prepulse inhibition (PPI). Acute CRFR1 blockade immediately reduced PPI in peripubertal males, but not females. In adulthood, each assay was repeated without CRFR1a exposure to test for persistent effects of the adolescent treatment. Males continued to experience deficits in PPI while females displayed altered locomotion, PPI, and social behavior. The amygdala was collected to measure long-term effects on gene expression. In the adult amygdala, peripubertal CRFR1a induced alterations in pathways related to neural plasticity and stress in males. In females, pathways related to central nervous system myelination, cell junction organization, and glutamatergic regulation of synaptic transmission were affected. Understanding how acute exposure to neuropharmacological agents can have sustained impacts on brain and behavior, in the absence of further exposures, has important clinical implications for developing adolescents.
Collapse
Affiliation(s)
- Julia Martz
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| | - Micah A. Shelton
- Department of Psychiatry, University of Pittsburgh, 450
Technology Drive Pittsburgh, PA, 15219
| | - Tristen J. Langen
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| | - Sakhi Srinivasan
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| | - Marianne L. Seney
- Department of Psychiatry, University of Pittsburgh, 450
Technology Drive Pittsburgh, PA, 15219
| | - Amanda C. Kentner
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| |
Collapse
|
8
|
Vingan I, Phatarpekar S, Tung VSK, Hernández AI, Evgrafov OV, Alarcon JM. Spatially resolved transcriptomic signatures of hippocampal subregions and Arc-expressing ensembles in active place avoidance memory. Front Mol Neurosci 2024; 17:1386239. [PMID: 39544521 PMCID: PMC11560897 DOI: 10.3389/fnmol.2024.1386239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc + and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc + and Arc- spatial transcriptomic spots.
Collapse
Affiliation(s)
- Isaac Vingan
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Shwetha Phatarpekar
- Institute for Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Victoria Sook Keng Tung
- School of Graduate Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Alejandro Iván Hernández
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Oleg V. Evgrafov
- Institute for Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- School of Graduate Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States
| | - Juan Marcos Alarcon
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| |
Collapse
|
9
|
Petrany A, Chen R, Zhang S, Chen Y. Theoretical framework for the difference of two negative binomial distributions and its application in comparative analysis of sequencing data. Genome Res 2024; 34:1636-1650. [PMID: 39406498 PMCID: PMC11529838 DOI: 10.1101/gr.278843.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 09/10/2024] [Indexed: 11/01/2024]
Abstract
High-throughput sequencing (HTS) technologies have been instrumental in investigating biological questions at the bulk and single-cell levels. Comparative analysis of two HTS data sets often relies on testing the statistical significance for the difference of two negative binomial distributions (DOTNB). Although negative binomial distributions are well studied, the theoretical results for DOTNB remain largely unexplored. Here, we derive basic analytical results for DOTNB and examine its asymptotic properties. As a state-of-the-art application of DOTNB, we introduce DEGage, a computational method for detecting differentially expressed genes (DEGs) in scRNA-seq data. DEGage calculates the mean of the sample-wise differences of gene expression levels as the test statistic and determines significant differential expression by computing the P-value with DOTNB. Extensive validation using simulated and real scRNA-seq data sets demonstrates that DEGage outperforms five popular DEG analysis tools: DEGseq2, DEsingle, edgeR, Monocle3, and scDD. DEGage is robust against high dropout levels and exhibits superior sensitivity when applied to balanced and imbalanced data sets, even with small sample sizes. We utilize DEGage to analyze prostate cancer scRNA-seq data sets and identify marker genes for 17 cell types. Furthermore, we apply DEGage to scRNA-seq data sets of mouse neurons with and without fear memory and reveal eight potential memory-related genes overlooked in previous analyses. The theoretical results and supporting software for DOTNB can be widely applied to comparative analyses of dispersed count data in HTS and broad research questions.
Collapse
Affiliation(s)
- Alicia Petrany
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, New Jersey 08028, USA
| | - Ruoyu Chen
- Moorestown High School, Moorestown, New Jersey 08057, USA
| | - Shaoqiang Zhang
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China
| | - Yong Chen
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, New Jersey 08028, USA;
| |
Collapse
|
10
|
Sanguino-Gómez J, Huijgens S, den Hartog M, Schenk IJM, Kluck W, Versluis TD, Krugers HJ. Neural correlates of learning and memory are altered by early-life stress. Neurobiol Learn Mem 2024; 213:107952. [PMID: 38906243 DOI: 10.1016/j.nlm.2024.107952] [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: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024]
Abstract
The ability to learn and remember, which is fundamental for behavioral adaptation, is susceptible to stressful experiences during the early postnatal period, such as abnormal levels of maternal care. The exact mechanisms underlying these effects still remain elusive. This study examined whether early life stress (ELS) alters memory and brain activation patterns in male mice. Therefore, we examined the expression of the immediate early genes (IEGs) c-Fos and Arc in the dentate gyrus (DG) and basolateral amygdala (BLA) after training and memory retrieval in a fear conditioning task. Furthermore, we examined the potential of RU38486 (RU486), a glucocorticoid receptor antagonist, to mitigate ELS-induced memory deficits by blocking stress signalling during adolescence. Arc::dVenus reporter mice, which allow investigating experience-dependent expression of the immediate early gene Arc also at more remote time points, were exposed to ELS by housing dams and offspring with limited bedding and nesting material (LBN) between postnatal days (PND) 2-9 and trained in a fear conditioning task at adult age. We found that ELS reduced both fear acquisition and contextual memory retrieval. RU486 did not prevent these effects. ELS reduced the number of Arc::dVenus+ cells in DG and BLA after training, while the number of c-Fos+ cells were left unaffected. After memory retrieval, ELS decreased c-Fos+ cells in the ventral DG and BLA. ELS also altered the colocalization of c-Fos+ cells with Arc::dVenus+ cells in the ventral DG, possibly indicating impaired engram allocation in the ventral DG after memory retrieval. In conclusion, this study shows that ELS alters neuronal activation patterns after fear acquisition and retrieval, which may provide mechanistic insights into enduring impact of ELS on the processing of fear memories, possibly via changes in cell (co-) activation and engram cell allocation.
Collapse
Affiliation(s)
| | - Stefan Huijgens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Maxine den Hartog
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Inim J M Schenk
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Wenya Kluck
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamara D Versluis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Dovek L, Marrero K, Zagha E, Santhakumar V. Cellular and circuit features distinguish dentate gyrus semilunar granule cells and granule cells activated during contextual memory formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608983. [PMID: 39229181 PMCID: PMC11370351 DOI: 10.1101/2024.08.21.608983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The dentate gyrus is critical for spatial memory formation and shows task related activation of cellular ensembles considered as memory engrams. Semilunar granule cells (SGCs), a sparse dentate projection neuron subtype distinct from granule cells (GCs), were recently reported to be enriched among behaviorally activated neurons. However, the mechanisms governing SGC recruitment during memory formation and their role in engram refinement remains unresolved. By examining neurons labeled during contextual memory formation in TRAP2 mice, we empirically tested competing hypotheses for GC and SGC recruitment into memory ensembles. In support of the proposal that more excitable neurons are preferentially recruited into memory ensembles, SGCs showed greater sustained firing than GCs. Additionally, SGCs labeled during memory formation showed less adapting firing than unlabeled SGCs. Our recordings did not reveal glutamatergic connections between behaviorally labeled SGCs and GCs, providing evidence against SGCs driving local circuit feedforward excitation in ensemble recruitment. Contrary to a leading hypothesis, there was little evidence for individual SGCs or labeled neuronal ensembles supporting lateral inhibition of unlabeled neurons. Instead, pairs of GCs and SGCs within labeled neuronal cohorts received more temporally correlated spontaneous excitatory synaptic inputs than labeled-unlabeled neuronal pairs, validating a role for correlated afferent inputs in neuronal ensemble selection. These findings challenge the proposal that SGCs drive dentate GC ensemble refinement, while supporting a role for intrinsic active properties and correlated inputs in preferential SGC recruitment to contextual memory engrams. Impact Statement Evaluation of semilunar granule cell involvement in dentate gyrus contextual memory processing supports recruitment based on intrinsic and input characteristics while revealing limited contribution to ensemble refinement.
Collapse
Affiliation(s)
- Laura Dovek
- Biomedical Sciences Graduate Program, University of California Riverside, Riverside, California 92521
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Krista Marrero
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Edward Zagha
- Biomedical Sciences Graduate Program, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
- Department of Psychology, University of California Riverside, Riverside, California 92521
| | - Vijayalakshmi Santhakumar
- Biomedical Sciences Graduate Program, University of California Riverside, Riverside, California 92521
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| |
Collapse
|
12
|
Wu X, Zhu X, Pan Y, Gu X, Liu X, Chen S, Zhang Y, Xu T, Xu N, Sun S. Amygdala neuronal dyshomeostasis via 5-HT receptors mediates mood and cognitive defects in Alzheimer's disease. Aging Cell 2024; 23:e14187. [PMID: 38716507 PMCID: PMC11320345 DOI: 10.1111/acel.14187] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 08/15/2024] Open
Abstract
Behavioral changes or neuropsychiatric symptoms (NPSs) are common features in dementia and are associated with accelerated cognitive impairment and earlier deaths. However, how NPSs are intertwined with cognitive decline remains elusive. In this study, we identify that the basolateral amygdala (BLA) is a key brain region that is associated with mood disorders and memory decline in the AD course. During the process from pre- to post-onset in AD, the dysfunction of parvalbumin (PV) interneurons and pyramidal neurons in the amygdala leads to hyperactivity of pyramidal neurons in the basal state and insensitivity to external stimuli. We further demonstrate that serotonin (5-HT) receptors in distinct neurons synergistically regulate the BLA microcircuit of AD rather than 5-HT levels, in which both restrained inhibitory inputs by excessive 5-HT1AR signaling in PV interneurons and depolarized pyramidal neurons via upregulated 5-HT2AR contribute to aberrant neuronal hyperactivity. Downregulation of these two 5-HT receptors simultaneously enables neurons to resist β-amyloid peptides (Aβ) neurotoxicity and ameliorates the mood and cognitive defects. Therefore, our study reveals a crucial role of 5-HT receptors for regulating neuronal homeostasis in AD pathogenesis, and this would provide early intervention and potential targets for AD cognitive decline.
Collapse
Affiliation(s)
- Xin‐Rong Wu
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao‐Na Zhu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuan‐Bo Pan
- Department of Neurosurgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue Gu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xian‐Dong Liu
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Si Chen
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yu Zhang
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tian‐Le Xu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Nan‐Jie Xu
- Department of Anatomy and Physiology, Collaborative Innovation Center for Brain ScienceShanghai Jiao Tong University School of MedicineShanghaiChina
- Songjiang Hospital and Songjiang Research InstituteShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Suya Sun
- Department of NeurologyInstitute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
13
|
Santoni G, Astori S, Leleu M, Glauser L, Zamora SA, Schioppa M, Tarulli I, Sandi C, Gräff J. Chromatin plasticity predetermines neuronal eligibility for memory trace formation. Science 2024; 385:eadg9982. [PMID: 39052786 DOI: 10.1126/science.adg9982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/01/2024] [Accepted: 05/24/2024] [Indexed: 07/27/2024]
Abstract
Memories are encoded by sparse populations of neurons but how such sparsity arises remains largely unknown. We found that a neuron's eligibility to be recruited into the memory trace depends on its epigenetic state prior to encoding. Principal neurons in the mouse lateral amygdala display intrinsic chromatin plasticity, which when experimentally elevated favors neuronal allocation into the encoding ensemble. Such chromatin plasticity occurred at genomic regions underlying synaptic plasticity and was accompanied by increased neuronal excitability in single neurons in real time. Lastly, optogenetic silencing of the epigenetically altered neurons prevented memory expression, revealing a cell-autonomous relationship between chromatin plasticity and memory trace formation. These results identify the epigenetic state of a neuron as a key factor enabling information encoding.
Collapse
Affiliation(s)
- Giulia Santoni
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Simone Astori
- Laboratory of Behavioural Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Marion Leleu
- Bioinformatics Competence Center, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Liliane Glauser
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Simon A Zamora
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Myriam Schioppa
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
- The institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Isabella Tarulli
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioural Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
14
|
Park H, Kaang BK. Memory allocation at the neuronal and synaptic levels. BMB Rep 2024; 57:176-181. [PMID: 37964638 PMCID: PMC11058361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/05/2023] [Accepted: 11/10/2023] [Indexed: 11/16/2023] Open
Abstract
Memory allocation, which determines where memories are stored in specific neurons or synapses, has consistently been demonstrated to occur via specific mechanisms. Neuronal allocation studies have focused on the activated population of neurons and have shown that increased excitability via cAMP response element-binding protein (CREB) induces a bias toward memoryencoding neurons. Synaptic allocation suggests that synaptic tagging enables memory to be mediated through different synaptic strengthening mechanisms, even within a single neuron. In this review, we summarize the fundamental concepts of memory allocation at the neuronal and synaptic levels and discuss their potential interrelationships. [BMB Reports 2024; 57(4): 176-181].
Collapse
Affiliation(s)
- HyoJin Park
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Department of Biological Science, Seoul National University, Seoul 08826, Korea
| | - Bong-Kiun Kaang
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
| |
Collapse
|
15
|
Vingan I, Phatarpekar S, Tung VSK, Hernández AI, Evgrafov OV, Alarcon JM. Spatially Resolved Transcriptomic Signatures of Hippocampal Subregions and Arc-Expressing Ensembles in Active Place Avoidance Memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573225. [PMID: 38260257 PMCID: PMC10802250 DOI: 10.1101/2023.12.30.573225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc+ and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc+ and Arc- spatial transcriptomic spots.
Collapse
Affiliation(s)
- Isaac Vingan
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Shwetha Phatarpekar
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Victoria Sook Keng Tung
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - A. Iván Hernández
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Oleg V. Evgrafov
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Juan Marcos Alarcon
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
16
|
Jung JH, Wang Y, Rashid AJ, Zhang T, Frankland PW, Josselyn SA. Examining memory linking and generalization using scFLARE2, a temporally precise neuronal activity tagging system. Cell Rep 2023; 42:113592. [PMID: 38103203 PMCID: PMC10842737 DOI: 10.1016/j.celrep.2023.113592] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/26/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
How memories are organized in the brain influences whether they are remembered discretely versus linked with other experiences or whether generalized information is applied to entirely novel situations. Here, we used scFLARE2 (single-chain fast light- and activity-regulated expression 2), a temporally precise tagging system, to manipulate mouse lateral amygdala neurons active during one of two 3 min threat experiences occurring close (3 h) or further apart (27 h) in time. Silencing scFLARE2-tagged neurons showed that two threat experiences occurring at distal times are dis-allocated to orthogonal engram ensembles and remembered discretely, whereas the same two threat experiences occurring in close temporal proximity are linked via co-allocation to overlapping engram ensembles. Moreover, we found that co-allocation mediates memory generalization applied to a completely novel stimulus. These results indicate that endogenous temporal evolution of engram ensemble neuronal excitability determines how memories are organized and remembered and that this would not be possible using conventional immediate-early gene-based tagging methods.
Collapse
Affiliation(s)
- Jung Hoon Jung
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Ying Wang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Asim J Rashid
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Tao Zhang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada; Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada.
| |
Collapse
|
17
|
Hochgerner H, Singh S, Tibi M, Lin Z, Skarbianskis N, Admati I, Ophir O, Reinhardt N, Netser S, Wagner S, Zeisel A. Neuronal types in the mouse amygdala and their transcriptional response to fear conditioning. Nat Neurosci 2023; 26:2237-2249. [PMID: 37884748 PMCID: PMC10689239 DOI: 10.1038/s41593-023-01469-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
The amygdala is a brain region primarily associated with emotional response. The use of genetic markers and single-cell transcriptomics can provide insights into behavior-associated cell state changes. Here we present a detailed cell-type taxonomy of the adult mouse amygdala during fear learning and memory consolidation. We perform single-cell RNA sequencing on naïve and fear-conditioned mice, identify 130 neuronal cell types and validate their spatial distributions. A subset of all neuronal types is transcriptionally responsive to fear learning and memory retrieval. The activated engram cells upregulate activity-response genes and coordinate the expression of genes associated with neurite outgrowth, synaptic signaling, plasticity and development. We identify known and previously undescribed candidate genes responsive to fear learning. Our molecular atlas may be used to generate hypotheses to unveil the neuron types and neural circuits regulating the emotional component of learning and memory.
Collapse
Affiliation(s)
- Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shelly Singh
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Muhammad Tibi
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Zhige Lin
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Niv Skarbianskis
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Osnat Ophir
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nuphar Reinhardt
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shai Netser
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Shlomo Wagner
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Amit Zeisel
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
18
|
Lee H, Kaang BK. How engram mediates learning, extinction, and relapse. Curr Opin Neurobiol 2023; 81:102723. [PMID: 37030026 DOI: 10.1016/j.conb.2023.102723] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 04/08/2023]
Abstract
Fear learning ensures survival through an expression of certain behavior as a conditioned fear response. Fear memory is processed and stored in a fear memory circuit, including the amygdala, hippocampus, and prefrontal cortex. A gradual decrease in conditioned fear response can be induced by fear extinction, which is mediated through the weakening of the original fear memory traces and the newly formed inhibition of those traces. Fear memory can also recover after extinction, which shows flexible control of the fear memory state. Here, we demonstrate how fear engram, which is a physical substrate of fear memory, changes during fear extinction and relapse by reviewing recent studies regarding engram.
Collapse
Affiliation(s)
- Hoonwon Lee
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea.
| |
Collapse
|
19
|
Guskjolen A, Cembrowski MS. Engram neurons: Encoding, consolidation, retrieval, and forgetting of memory. Mol Psychiatry 2023; 28:3207-3219. [PMID: 37369721 PMCID: PMC10618102 DOI: 10.1038/s41380-023-02137-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
Tremendous strides have been made in our understanding of the neurobiological substrates of memory - the so-called memory "engram". Here, we integrate recent progress in the engram field to illustrate how engram neurons transform across the "lifespan" of a memory - from initial memory encoding, to consolidation and retrieval, and ultimately to forgetting. To do so, we first describe how cell-intrinsic properties shape the initial emergence of the engram at memory encoding. Second, we highlight how these encoding neurons preferentially participate in synaptic- and systems-level consolidation of memory. Third, we describe how these changes during encoding and consolidation guide neural reactivation during retrieval, and facilitate memory recall. Fourth, we describe neurobiological mechanisms of forgetting, and how these mechanisms can counteract engram properties established during memory encoding, consolidation, and retrieval. Motivated by recent experimental results across these four sections, we conclude by proposing some conceptual extensions to the traditional view of the engram, including broadening the view of cell-type participation within engrams and across memory stages. In collection, our review synthesizes general principles of the engram across memory stages, and describes future avenues to further understand the dynamic engram.
Collapse
Affiliation(s)
- Axel Guskjolen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Mark S Cembrowski
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Brosens N, Lesuis SL, Bassie I, Reyes L, Gajadien P, Lucassen PJ, Krugers HJ. Elevated corticosterone after fear learning impairs remote auditory memory retrieval and alters brain network connectivity. Learn Mem 2023; 30:125-132. [PMID: 37487708 PMCID: PMC10519398 DOI: 10.1101/lm.053836.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023]
Abstract
Glucocorticoids are potent memory modulators that can modify behavior in an adaptive or maladaptive manner. Elevated glucocorticoid levels after learning promote memory consolidation at recent time points, but their effects on remote time points are not well established. Here we set out to assess whether corticosterone (CORT) given after learning modifies remote fear memory. To that end, mice were exposed to a mild auditory fear conditioning paradigm followed by a single 2 mg/kg CORT injection, and after 28 d, auditory memory was assessed. Neuronal activation was investigated using immunohistochemistry for the immediate early gene c-Fos, and coactivation of brain regions was determined using a correlation matrix analysis. CORT-treated mice displayed significantly less remote auditory memory retrieval. While the net activity of studied brain regions was similar compared with the control condition, CORT-induced remote memory impairment was associated with altered correlated activity between brain regions. Specifically, connectivity of the lateral amygdala with the basal amygdala and the dorsal dentate gyrus was significantly reduced in CORT-treated mice, suggesting disrupted network connectivity that may underlie diminished remote memory retrieval. Elucidating the pathways underlying these effects could help provide mechanistic insight into the effects of stress on memory and possibly provide therapeutic targets for psychopathology.
Collapse
Affiliation(s)
- Niek Brosens
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Sylvie L Lesuis
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Ilse Bassie
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Lara Reyes
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Priya Gajadien
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| |
Collapse
|
21
|
Jung JH, Wang Y, Mocle AJ, Zhang T, Köhler S, Frankland PW, Josselyn SA. Examining the engram encoding specificity hypothesis in mice. Neuron 2023; 111:1830-1845.e5. [PMID: 36990091 DOI: 10.1016/j.neuron.2023.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023]
Abstract
According to the encoding specificity hypothesis, memory is best recalled by retrieval cues that overlap with training cues. Human studies generally support this hypothesis. However, memories are thought to be stored in neuronal ensembles (engrams), and retrieval cues are thought to reactivate neurons in an engram to induce memory recall. Here, we visualized engrams in mice to test whether retrieval cues that overlap with training cues produce maximal memory recall via high engram reactivation (engram encoding specificity hypothesis). Using variations of cued threat conditioning (pairing conditioned stimulus [CS] with footshock), we manipulated encoding and retrieval conditions along multiple domains, including pharmacological state, external sensory cue, and internal optogenetic cue. Maximal engram reactivation and memory recall occurred when retrieval conditions closely matched training conditions. These findings provide a biological basis for the encoding specificity hypothesis and highlight the important interaction between stored information (engram) and cues available at memory retrieval (ecphory).
Collapse
Affiliation(s)
- Jung Hoon Jung
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Ying Wang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Andrew J Mocle
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tao Zhang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Stefan Köhler
- Department of Psychology, University of Western Ontario, London, ON N6A 5C2, Canada; The Brain and Mind Institute, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada; Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada.
| |
Collapse
|
22
|
Scott DN, Frank MJ. Adaptive control of synaptic plasticity integrates micro- and macroscopic network function. Neuropsychopharmacology 2023; 48:121-144. [PMID: 36038780 PMCID: PMC9700774 DOI: 10.1038/s41386-022-01374-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022]
Abstract
Synaptic plasticity configures interactions between neurons and is therefore likely to be a primary driver of behavioral learning and development. How this microscopic-macroscopic interaction occurs is poorly understood, as researchers frequently examine models within particular ranges of abstraction and scale. Computational neuroscience and machine learning models offer theoretically powerful analyses of plasticity in neural networks, but results are often siloed and only coarsely linked to biology. In this review, we examine connections between these areas, asking how network computations change as a function of diverse features of plasticity and vice versa. We review how plasticity can be controlled at synapses by calcium dynamics and neuromodulatory signals, the manifestation of these changes in networks, and their impacts in specialized circuits. We conclude that metaplasticity-defined broadly as the adaptive control of plasticity-forges connections across scales by governing what groups of synapses can and can't learn about, when, and to what ends. The metaplasticity we discuss acts by co-opting Hebbian mechanisms, shifting network properties, and routing activity within and across brain systems. Asking how these operations can go awry should also be useful for understanding pathology, which we address in the context of autism, schizophrenia and Parkinson's disease.
Collapse
Affiliation(s)
- Daniel N Scott
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| | - Michael J Frank
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
23
|
Eriksen MS, Bramham CR. Molecular physiology of Arc/Arg3.1: The oligomeric state hypothesis of synaptic plasticity. Acta Physiol (Oxf) 2022; 236:e13886. [PMID: 36073248 PMCID: PMC9787330 DOI: 10.1111/apha.13886] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 01/29/2023]
Abstract
The immediate early gene, Arc, is a pivotal regulator of synaptic plasticity, memory, and cognitive flexibility. But what is Arc protein? How does it work? Inside the neuron, Arc is a protein interaction hub and dynamic regulator of intra-cellular signaling in synaptic plasticity. In remarkable contrast, Arc can also self-assemble into retrovirus-like capsids that are released in extracellular vesicles and capable of intercellular transfer of RNA. Elucidation of the molecular basis of Arc hub and capsid functions, and the relationship between them, is vital for progress. Here, we discuss recent findings on Arc structure-function and regulation of oligomerization that are giving insight into the molecular physiology of Arc. The unique features of mammalian Arc are emphasized, while drawing comparisons with Drosophila Arc and retroviral Gag. The Arc N-terminal domain, found only in mammals, is proposed to play a key role in regulating Arc hub signaling, oligomerization, and formation of capsids. Bringing together several lines of evidence, we hypothesize that Arc function in synaptic plasticity-long-term potentiation (LTP) and long-term depression (LTD)-are dictated by different oligomeric forms of Arc. Specifically, monomer/dimer function in LTP, tetramer function in basic LTD, and 32-unit oligomer function in enhanced LTD. The role of mammalian Arc capsids is unclear but likely depends on the cross-section of captured neuronal activity-induced RNAs. As the functional states of Arc are revealed, it may be possible to selectively manipulate specific forms of Arc-dependent plasticity and intercellular communication involved in brain function and dysfunction.
Collapse
Affiliation(s)
| | - Clive R. Bramham
- Department of BiomedicineUniversity of BergenBergenNorway,Mohn Research Center for the BrainUniversity of BergenBergenNorway
| |
Collapse
|
24
|
Awasthi R, Chandra N, Barkai E. Olfactory rule learning-induced enhancement in intrinsic neuronal excitability is maintained by shutdown of the cholinergic M-current. Front Cell Neurosci 2022; 16:934838. [PMID: 36246520 PMCID: PMC9556983 DOI: 10.3389/fncel.2022.934838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Training rats in a particularly difficult olfactory discrimination task initiates a period of accelerated learning, manifested as a dramatic increase in the rats' capacity to discriminate between pairs of odors once they have learned the discrimination task, implying that rule learning has taken place. At the cellular biophysical level, rule learning is maintained by reduction in the conductance of the slow current (sIAHP) simultaneously in most piriform cortex layer II pyramidal neurons. Such sIAHP reduction is expressed in attenuation of the post-burst afterhyperpolarization (AHP) potential and thus in enhanced repetitive action potential firing. Previous studies have shown that a causal relationship exists between long-lasting post-burst AHP reduction and rule learning. A specific channel through which the sIAHP flows has not been identified. The sIAHP in pyramidal cells is critically dependent on membrane phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)]. PtdIns(4,5)P(2) regulates the calcium sensitivity of the sIAHP by acting downstream from the rise in intracellular calcium. These findings led to the interesting hypothesis that PtdIns(4,5)P(2) activates a variety of potassium channels. Thus, the sIAHP would not represent a unitary ionic current but the embodiment of a generalized potassium channel gating mechanism. We thus hypothesized that the learning-induced increase in intrinsic excitability is mediated by reduced conductance of one or more of the currents that contribute to the sIAHP. Here we first show, using current-clamp recordings, that the post-burst AHP in piriform cortex pyramidal neurons is also mediated by the Ih, and the contribution of this current to the post-burst AHP is also affected by learning. We also show, using whole-cell patch-clamp recordings, that the sIAHP in neurons from trained rats is not sensitive to blocking membrane phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)], and to the blocking of the current mediated by the cholinergic muscarinic acetylcholine receptor (M-current). Further current-clamp recordings also show that blocking PtdIns(4,5)P(2) synthesis and application of a specific IKCa blocker have no effect on the post-burst AHP in neurons from trained as well as control rats. Taken together with results from our previous studies, these data suggest that rule learning-induced long-lasting enhancement in intrinsic neuronal excitability results from reduced conductance of the M-current and thus the slow potassium currents, which control repetitive spike firing.
Collapse
Affiliation(s)
| | | | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
25
|
Hwang FJ, Roth RH, Wu YW, Sun Y, Kwon DK, Liu Y, Ding JB. Motor learning selectively strengthens cortical and striatal synapses of motor engram neurons. Neuron 2022; 110:2790-2801.e5. [PMID: 35809573 PMCID: PMC9464700 DOI: 10.1016/j.neuron.2022.06.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/21/2022] [Accepted: 06/07/2022] [Indexed: 11/28/2022]
Abstract
Learning and consolidation of new motor skills require plasticity in the motor cortex and striatum, two key motor regions of the brain. However, how neurons undergo synaptic changes and become recruited during motor learning to form a memory engram remains unknown. Here, we train mice on a motor learning task and use a genetic approach to identify and manipulate behavior-relevant neurons selectively in the primary motor cortex (M1). We find that the degree of M1 engram neuron reactivation correlates with motor performance. We further demonstrate that learning-induced dendritic spine reorganization specifically occurs in these M1 engram neurons. In addition, we find that motor learning leads to an increase in the strength of M1 engram neuron outputs onto striatal spiny projection neurons (SPNs) and that these synapses form clusters along SPN dendrites. These results identify a highly specific synaptic plasticity during the formation of long-lasting motor memory traces in the corticostriatal circuit.
Collapse
Affiliation(s)
- Fuu-Jiun Hwang
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Yu-Wei Wu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Yue Sun
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Destany K Kwon
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Yu Liu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
26
|
Cox WR, Faliagkas L, Besseling A, van der Loo RJ, Spijker S, Kindt M, Rao-Ruiz P. Interfering With Contextual Fear Memories by Post-reactivation Administration of Propranolol in Mice: A Series of Null Findings. Front Behav Neurosci 2022; 16:893572. [PMID: 35832291 PMCID: PMC9272000 DOI: 10.3389/fnbeh.2022.893572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Post-reactivation amnesia of contextual fear memories by blockade of noradrenergic signaling has been shown to have limited replicability in rodents. This is usually attributed to several boundary conditions that gate the destabilization of memory during its retrieval. How these boundary conditions can be overcome, and what neural mechanisms underlie post-reactivation changes in contextual fear memories remain largely unknown. Here, we report a series of experiments in a contextual fear-conditioning paradigm in mice, that were aimed at solving these issues. We first attempted to obtain a training paradigm that would consistently result in contextual fear memory that could be destabilized upon reactivation, enabling post-retrieval amnesia by the administration of propranolol. Unexpectedly, our attempts were unsuccessful to this end. Specifically, over a series of experiments in which we varied different parameters of the fear acquisition procedure, at best small and inconsistent effects were observed. Additionally, we found that propranolol did not alter retrieval-induced neural activity, as measured by the number of c-Fos+ cells in the hippocampal dentate gyrus. To determine whether propranolol was perhaps ineffective in interfering with reactivated contextual fear memories, we also included anisomycin (i.e., a potent and well-known amnesic drug) in several experiments, and measures of synaptic glutamate receptor subunit GluA2 (i.e., a marker of memory destabilization). No post-retrieval amnesia by anisomycin and no altered GluA2 expression by reactivation was observed, suggesting that the memories did not undergo destabilization. The null findings are surprising, given that the training paradigms we implemented were previously shown to result in memories that could be modified upon reactivation. Together, our observations illustrate the elusive nature of reactivation-dependent changes in non-human fear memory.
Collapse
Affiliation(s)
- Wouter R. Cox
- Department of Psychology, Clinical Psychology, University of Amsterdam, Amsterdam, Netherlands
| | - Leonidas Faliagkas
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Amber Besseling
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Rolinka J. van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sabine Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Merel Kindt
- Department of Psychology, Clinical Psychology, University of Amsterdam, Amsterdam, Netherlands
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Priyanka Rao-Ruiz
| |
Collapse
|
27
|
Robertson EM. Memory leaks: information shared across memory systems. Trends Cogn Sci 2022; 26:544-554. [DOI: 10.1016/j.tics.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
|
28
|
The mouse brain after foot shock in four dimensions: Temporal dynamics at a single-cell resolution. Proc Natl Acad Sci U S A 2022; 119:2114002119. [PMID: 35181604 PMCID: PMC8872757 DOI: 10.1073/pnas.2114002119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2021] [Indexed: 02/08/2023] Open
Abstract
Acute stress involves the majority of brain areas, which can be sequentially organized in functional brain networks as shown by our study with foot shock in mice. We used whole-brain microscopy to investigate different spatial resolutions over time. From mesoscale region–based analyses, we identified the order of activation of brain areas. With single-cell analyses, we analyzed shifts in activation over time within small nuclei—a result impossible to achieve with functional MRI’s resolution. These findings required the development of a four-dimensional (4D) analytical pipeline, which is made available as an R package. This “atlas” of foot shock can be visualized in 4D in our interactive web portal. Acute stress leads to sequential activation of functional brain networks. A biologically relevant question is exactly which (single) cells belonging to brain networks are changed in activity over time after acute stress across the entire brain. We developed a preprocessing and analytical pipeline to chart whole-brain immediate early genes’ expression—as proxy for cellular activity—after a single stressful foot shock in four dimensions: that is, from functional networks up to three-dimensional (3D) single-cell resolution and over time. The pipeline is available as an R package. Most brain areas (96%) showed increased numbers of c-fos+ cells after foot shock, yet hypothalamic areas stood out as being most active and prompt in their activation, followed by amygdalar, prefrontal, hippocampal, and finally, thalamic areas. At the cellular level, c-fos+ density clearly shifted over time across subareas, as illustrated for the basolateral amygdala. Moreover, some brain areas showed increased numbers of c-fos+ cells, while others—like the dentate gyrus—dramatically increased c-fos intensity in just a subset of cells, reminiscent of engrams; importantly, this “strategy” changed after foot shock in half of the brain areas. One of the strengths of our approach is that single-cell data were simultaneously examined across all of the 90 brain areas and can be visualized in 3D in our interactive web portal.
Collapse
|
29
|
Poort J, Wilmes KA, Blot A, Chadwick A, Sahani M, Clopath C, Mrsic-Flogel TD, Hofer SB, Khan AG. Learning and attention increase visual response selectivity through distinct mechanisms. Neuron 2022; 110:686-697.e6. [PMID: 34906356 PMCID: PMC8860382 DOI: 10.1016/j.neuron.2021.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/31/2021] [Accepted: 11/15/2021] [Indexed: 11/28/2022]
Abstract
Selectivity of cortical neurons for sensory stimuli can increase across days as animals learn their behavioral relevance and across seconds when animals switch attention. While both phenomena occur in the same circuit, it is unknown whether they rely on similar mechanisms. We imaged primary visual cortex as mice learned a visual discrimination task and subsequently performed an attention switching task. Selectivity changes due to learning and attention were uncorrelated in individual neurons. Selectivity increases after learning mainly arose from selective suppression of responses to one of the stimuli but from selective enhancement and suppression during attention. Learning and attention differentially affected interactions between excitatory and PV, SOM, and VIP inhibitory cells. Circuit modeling revealed that cell class-specific top-down inputs best explained attentional modulation, while reorganization of local functional connectivity accounted for learning-related changes. Thus, distinct mechanisms underlie increased discriminability of relevant sensory stimuli across longer and shorter timescales.
Collapse
Affiliation(s)
- Jasper Poort
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Department of Psychology, University of Cambridge, Cambridge, UK.
| | | | - Antonin Blot
- Biozentrum, University of Basel, Basel, Switzerland; Sainsbury Wellcome Centre for Neural Circuits and Behavior, University College London, London, UK
| | - Angus Chadwick
- Gatsby Computational Neuroscience Unit, University College London, London, UK
| | - Maneesh Sahani
- Gatsby Computational Neuroscience Unit, University College London, London, UK
| | | | - Thomas D Mrsic-Flogel
- Biozentrum, University of Basel, Basel, Switzerland; Sainsbury Wellcome Centre for Neural Circuits and Behavior, University College London, London, UK
| | - Sonja B Hofer
- Biozentrum, University of Basel, Basel, Switzerland; Sainsbury Wellcome Centre for Neural Circuits and Behavior, University College London, London, UK
| | - Adil G Khan
- Biozentrum, University of Basel, Basel, Switzerland; Centre for Developmental Neurobiology, King's College London, London, UK.
| |
Collapse
|
30
|
Yan Y, Aierken A, Wang C, Jin W, Quan Z, Wang Z, Qing H, Ni J, Zhao J. Neuronal Circuits Associated with Fear Memory: Potential Therapeutic Targets for Posttraumatic Stress Disorder. Neuroscientist 2022; 29:332-351. [PMID: 35057666 DOI: 10.1177/10738584211069977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a psychiatric disorder that is associated with long-lasting memories of traumatic experiences. Extinction and discrimination of fear memory have become therapeutic targets for PTSD. Newly developed optogenetics and advanced in vivo imaging techniques have provided unprecedented spatiotemporal tools to characterize the activity, connectivity, and functionality of specific cell types in complicated neuronal circuits. The use of such tools has offered mechanistic insights into the exquisite organization of the circuitry underlying the extinction and discrimination of fear memory. This review focuses on the acquisition of more detailed, comprehensive, and integrated neural circuits to understand how the brain regulates the extinction and discrimination of fear memory. A future challenge is to translate these researches into effective therapeutic treatment for PTSD from the perspective of precise regulation of the neural circuits associated with the extinction and discrimination of fear memories.
Collapse
Affiliation(s)
- Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Ailikemu Aierken
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Chunjian Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Wei Jin
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhe Wang
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Juan Zhao
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
- Aerospace Medical Center, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
31
|
Cho HY, Shin W, Lee HS, Lee Y, Kim M, Oh JP, Han J, Jeong Y, Suh B, Kim E, Han JH. Turnover of fear engram cells by repeated experience. Curr Biol 2021; 31:5450-5461.e4. [PMID: 34687608 DOI: 10.1016/j.cub.2021.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/07/2021] [Accepted: 10/01/2021] [Indexed: 10/20/2022]
Abstract
A sparse population of neurons active during a learning event has been identified as memory engram cells. However, cells that are recruited to support memory when experience is repeated have been scarcely explored. Evidence from previous studies provides contradictory views. To address these questions, we employed learning-dependent cell labeling in the lateral amygdala (LA) and applied electrophysiological recording, spine imaging, and optogenetic tools to the labeled neurons with or without retraining. We found that engram cells established from original fear learning became dispensable for memory retrieval specifically with relearning, and this correlated with a reduction of synaptic transmission and loss of dendritic spines in these neurons. Despite such decreased connectivity, direct activation of these neurons resulted in fear-memory recall. We further identified that repeated memory was encoded in neurons active during relearning. These results suggest a shift in neuronal ensembles encoding fear memory in the LA by relearning through disconnection of the existing engram neurons established from original experience.
Collapse
Affiliation(s)
- Hye-Yeon Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Wangyong Shin
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Han-Sol Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Yeji Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Mujun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Jung-Pyo Oh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Junho Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Yire Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Boin Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Jin-Hee Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea; KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, Korea.
| |
Collapse
|
32
|
Lesuis SL, Brosens N, Immerzeel N, van der Loo RJ, Mitrić M, Bielefeld P, Fitzsimons CP, Lucassen PJ, Kushner SA, van den Oever MC, Krugers HJ. Glucocorticoids Promote Fear Generalization by Increasing the Size of a Dentate Gyrus Engram Cell Population. Biol Psychiatry 2021; 90:494-504. [PMID: 34503674 DOI: 10.1016/j.biopsych.2021.04.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Traumatic experiences, such as conditioned threat, are coded as enduring memories that are frequently subject to generalization, which is characterized by (re-) expression of fear in safe environments. However, the neurobiological mechanisms underlying threat generalization after a traumatic experience and the role of stress hormones in this process remain poorly understood. METHODS We examined the influence of glucocorticoid hormones on the strength and specificity of conditioned fear memory at the level of sparsely distributed dentate gyrus (DG) engram cells in male mice. RESULTS We found that elevating glucocorticoid hormones after fear conditioning induces a generalized contextual fear response. This was accompanied by a selective and persistent increase in the excitability and number of activated DG granule cells. Selective chemogenetic suppression of these sparse cells in the DG prevented glucocorticoid-induced fear generalization and restored contextual memory specificity, while leaving expression of auditory fear memory unaffected. CONCLUSIONS These results implicate the sparse ensemble of DG engram cells as a critical cellular substrate underlying fear generalization induced by glucocorticoid stress hormones.
Collapse
Affiliation(s)
- Sylvie L Lesuis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands; Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Niek Brosens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Nathalie Immerzeel
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Miodrag Mitrić
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pascal Bielefeld
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlos P Fitzsimons
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Giorgi C, Marinelli S. Roles and Transcriptional Responses of Inhibitory Neurons in Learning and Memory. Front Mol Neurosci 2021; 14:689952. [PMID: 34211369 PMCID: PMC8239217 DOI: 10.3389/fnmol.2021.689952] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence supports a model whereby memories are encoded by sparse ensembles of neurons called engrams, activated during memory encoding and reactivated upon recall. An engram consists of a network of cells that undergo long-lasting modifications of their transcriptional programs and connectivity. Ground-breaking advancements in this field have been made possible by the creative exploitation of the characteristic transcriptional responses of neurons to activity, allowing both engram labeling and manipulation. Nevertheless, numerous aspects of engram cell-type composition and function remain to be addressed. As recent transcriptomic studies have revealed, memory encoding induces persistent transcriptional and functional changes in a plethora of neuronal subtypes and non-neuronal cells, including glutamatergic excitatory neurons, GABAergic inhibitory neurons, and glia cells. Dissecting the contribution of these different cellular classes to memory engram formation and activity is quite a challenging yet essential endeavor. In this review, we focus on the role played by the GABAergic inhibitory component of the engram through two complementary lenses. On one hand, we report on available physiological evidence addressing the involvement of inhibitory neurons to different stages of memory formation, consolidation, storage and recall. On the other, we capitalize on a growing number of transcriptomic studies that profile the transcriptional response of inhibitory neurons to activity, revealing important clues on their potential involvement in learning and memory processes. The picture that emerges suggests that inhibitory neurons are an essential component of the engram, likely involved in engram allocation, in tuning engram excitation and in storing the memory trace.
Collapse
Affiliation(s)
- Corinna Giorgi
- CNR, Institute of Molecular Biology and Pathology, Rome, Italy.,European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Silvia Marinelli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| |
Collapse
|
34
|
Han DH, Park P, Choi DI, Bliss TVP, Kaang BK. The essence of the engram: Cellular or synaptic? Semin Cell Dev Biol 2021; 125:122-135. [PMID: 34103208 DOI: 10.1016/j.semcdb.2021.05.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
Memory is composed of various phases including cellular consolidation, systems consolidation, reconsolidation, and extinction. In the last few years it has been shown that simple association memories can be encoded by a subset of the neuronal population called engram cells. Activity of these cells is necessary and sufficient for the recall of association memory. However, it is unclear which molecular mechanisms allow cellular engrams to encode the diverse phases of memory. Further research is needed to examine the possibility that it is the synapses between engram cells (the synaptic engram) that constitute the memory. In this review we summarize recent findings on cellular engrams with a focus on different phases of memory, and discuss the distinct molecular mechanism required for cellular and synaptic engrams.
Collapse
Affiliation(s)
- Dae Hee Han
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Pojeong Park
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong Il Choi
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Tim V P Bliss
- Group leader emeritus, The Francis Crick Institute, 1 Midland Rd, Somers Town, London NW1 1AT, UK
| | - Bong-Kiun Kaang
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
35
|
Rao-Ruiz P, Visser E, Mitrić M, Smit AB, van den Oever MC. A Synaptic Framework for the Persistence of Memory Engrams. Front Synaptic Neurosci 2021; 13:661476. [PMID: 33841124 PMCID: PMC8024575 DOI: 10.3389/fnsyn.2021.661476] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/26/2021] [Indexed: 12/31/2022] Open
Abstract
The ability to store and retrieve learned information over prolonged periods of time is an essential and intriguing property of the brain. Insight into the neurobiological mechanisms that underlie memory consolidation is of utmost importance for our understanding of memory persistence and how this is affected in memory disorders. Recent evidence indicates that a given memory is encoded by sparsely distributed neurons that become highly activated during learning, so-called engram cells. Research by us and others confirms the persistent nature of cortical engram cells by showing that these neurons are required for memory expression up to at least 1 month after they were activated during learning. Strengthened synaptic connectivity between engram cells is thought to ensure reactivation of the engram cell network during retrieval. However, given the continuous integration of new information into existing neuronal circuits and the relatively rapid turnover rate of synaptic proteins, it is unclear whether a lasting learning-induced increase in synaptic connectivity is mediated by stable synapses or by continuous dynamic turnover of synapses of the engram cell network. Here, we first discuss evidence for the persistence of engram cells and memory-relevant adaptations in synaptic plasticity, and then propose models of synaptic adaptations and molecular mechanisms that may support memory persistence through the maintenance of enhanced synaptic connectivity within an engram cell network.
Collapse
Affiliation(s)
- Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther Visser
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Miodrag Mitrić
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
36
|
Joy MT, Carmichael ST. Encouraging an excitable brain state: mechanisms of brain repair in stroke. Nat Rev Neurosci 2021; 22:38-53. [PMID: 33184469 PMCID: PMC10625167 DOI: 10.1038/s41583-020-00396-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2020] [Indexed: 02/02/2023]
Abstract
Stroke induces a plastic state in the brain. This period of enhanced plasticity leads to the sprouting of new axons, the formation of new synapses and the remapping of sensory-motor functions, and is associated with motor recovery. This is a remarkable process in the adult brain, which is normally constrained in its levels of neuronal plasticity and connectional change. Recent evidence indicates that these changes are driven by molecular systems that underlie learning and memory, such as changes in cellular excitability during memory formation. This Review examines circuit changes after stroke, the shared mechanisms between memory formation and brain repair, the changes in neuronal excitability that underlie stroke recovery, and the molecular and pharmacological interventions that follow from these findings to promote motor recovery in animal models. From these findings, a framework emerges for understanding recovery after stroke, central to which is the concept of neuronal allocation to damaged circuits. The translation of the concepts discussed here to recovery in humans is underway in clinical trials for stroke recovery drugs.
Collapse
Affiliation(s)
- Mary T Joy
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Lau JMH, Rashid AJ, Jacob AD, Frankland PW, Schacter DL, Josselyn SA. The role of neuronal excitability, allocation to an engram and memory linking in the behavioral generation of a false memory in mice. Neurobiol Learn Mem 2020; 174:107284. [PMID: 32745601 DOI: 10.1016/j.nlm.2020.107284] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 01/23/2023]
Abstract
Memory is a constructive, not reproductive, process that is prone to errors. Errors in memory, though, may originate from normally adaptive memory processes. At the extreme of memory distortion is falsely "remembering" an event that did not occur. False memories are well-studied in cognitive psychology, but have received relatively less attention in neuroscience. Here, we took advantage of mechanistic insights into how neurons are allocated or recruited into an engram (memory trace) to generate a false memory in mice using only behavioral manipulations. At the time of an event, neurons compete for allocation to an engram supporting the memory for this event; neurons with higher excitability win this competition (Han et al., 2007). Even after the event, these allocated "engram neurons" remain temporarily (~6 h) more excitable than neighboring neurons. Should a similar event occur in this 6 h period of heightened engram neuron excitability, an overlapping population of neurons will be co-allocated to this second engram, which serves to functionally link the two memories (Rashid et al., 2016). Here, we applied this principle of co-allocation and found that mice develop a false fear memory to a neutral stimulus if exposed to this stimulus shortly (3 h), but not a longer time (24 h), after cued fear conditioning. Similar to co-allocation, the generation of this false memory depended on the post-training excitability of engram neurons such that these neurons remained more excitable during exposure to the neutral stimulus at 3 h but not 24 h. Optogenetically silencing engram neurons 3 h after cued fear conditioning impaired formation of a false fear memory to the neutral stimulus, while optogenetically activating engram neurons 24 h after cued fear conditioning created a false fear memory. These results suggest that some false memories may originate from normally adaptive mnemonic processes such as neuronal excitability-dependent allocation and memory linking.
Collapse
Affiliation(s)
- Jocelyn M H Lau
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Asim J Rashid
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5G 1X9, Canada; Dept. of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada; Dept. of Psychology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Alexander D Jacob
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Dept. of Psychology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5G 1X9, Canada; Dept. of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada; Dept. of Psychology, University of Toronto, Toronto, ON M5G 1X8, Canada; Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada
| | | | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5G 1X9, Canada; Dept. of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada; Dept. of Psychology, University of Toronto, Toronto, ON M5G 1X8, Canada; Brain, Mind & Consciousness Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
38
|
Lindquist DH. Emotion in motion: A three-stage model of aversive classical conditioning. Neurosci Biobehav Rev 2020; 115:363-377. [DOI: 10.1016/j.neubiorev.2020.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 01/12/2023]
|
39
|
Josselyn SA, Tonegawa S. Memory engrams: Recalling the past and imagining the future. Science 2020; 367:367/6473/eaaw4325. [PMID: 31896692 DOI: 10.1126/science.aaw4325] [Citation(s) in RCA: 542] [Impact Index Per Article: 108.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In 1904, Richard Semon introduced the term "engram" to describe the neural substrate for storing memories. An experience, Semon proposed, activates a subset of cells that undergo off-line, persistent chemical and/or physical changes to become an engram. Subsequent reactivation of this engram induces memory retrieval. Although Semon's contributions were largely ignored in his lifetime, new technologies that allow researchers to image and manipulate the brain at the level of individual neurons has reinvigorated engram research. We review recent progress in studying engrams, including an evaluation of evidence for the existence of engrams, the importance of intrinsic excitability and synaptic plasticity in engrams, and the lifetime of an engram. Together, these findings are beginning to define an engram as the basic unit of memory.
Collapse
Affiliation(s)
- Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada. .,Department of Psychology, University of Toronto, Toronto, Ontario M5S 3G3, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario M5G 1X8, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Brain, Mind & Consciousness Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario M5G 1M1, Canada
| | - Susumu Tonegawa
- RIKEN-MIT Laboratory for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
40
|
Nawarawong NN, Olsen CM. Within-animal comparisons of novelty and cocaine neuronal ensemble overlap in the nucleus accumbens and prefrontal cortex. Behav Brain Res 2020; 379:112275. [PMID: 31614186 DOI: 10.1016/j.bbr.2019.112275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/28/2019] [Accepted: 10/01/2019] [Indexed: 11/29/2022]
Abstract
Novelty seeking is a personality trait associated with an increased vulnerability for substance abuse. In rodents, elevated novelty seeking has been shown to be a predictor for elevated drug self-administration and compulsive use. While previous studies have shown that both novelty and drugs of abuse have actions within similar mesocorticolimbic regions, little is known as to whether the same neural ensembles are engaged by these two stimuli. Using the TetTag mouse model and Fos immunohistochemistry, we measured neurons engaged by novelty and acute cocaine exposure, respectively in the prefrontal cortex (PFC) and nucleus accumbens (NAc). While there was no significant impact of novelty exposure on the size of the EGFP+ ensemble, we found that cocaine engaged significantly more Fos+ neurons in the NAc, while stress increased the size of the Fos+ ensemble in the PFC. Analysis of ensemble reactivation was specific to the emotional valence of the second stimuli. We found that a greater proportion of the EGFP+ ensemble was reactivated in the groups that paired novelty with a positive (cocaine) or neutral (saline) experience in the NAc, while the novelty/stress paired groups exhibited significantly less ensemble overlap in the PFC. However, only in the NAc shell was this increase in ensemble overlap specific to those exposed to both novelty and cocaine. This suggests that the NAc shell, but not the NAc core or PFC, may play an important role in general reward processing by engaging a similar network of neurons.
Collapse
Affiliation(s)
- Natalie N Nawarawong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Christopher M Olsen
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
41
|
Penrod RD, Thomsen M, Taniguchi M, Guo Y, Cowan CW, Smith LN. The activity-regulated cytoskeleton-associated protein, Arc/Arg3.1, influences mouse cocaine self-administration. Pharmacol Biochem Behav 2020; 188:172818. [PMID: 31682894 PMCID: PMC7202920 DOI: 10.1016/j.pbb.2019.172818] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/01/2019] [Accepted: 10/30/2019] [Indexed: 01/04/2023]
Abstract
The activity-regulated cytoskeleton-associated protein (Arc, also known as Arg3.1), an immediate early gene and synaptic regulator, is upregulated following a single cocaine exposure. However, there is not much known regarding Arc/Arg3.1's potential contribution to addiction-relevant behaviors. Despite known learning and memory deficits in contextual fear and water-maze reversal learning tasks, we find that mice lacking Arc/Arg3.1 perform conditioned place preference and operant conditioning involving positive reinforcers (food and cocaine) with little-to-no impairment. However, following normal saline-extinction, wild type (WT) mice show a classic inverted-U dose-response function, while Arc/Arg3.1 knockout (KO) mice fail to adjust their intake across multiple doses. Importantly, Arc/Arg3.1 KO and WT mice behave comparably on an increasing cost task (FR1-FR3; acquisition dose), providing evidence that both groups find cocaine reinforcing. Differences in individuals that drive variations in use patterns and particularly, drug intake levels, are critical as they influence the likelihood of developing dependence. Our data suggest that Arc/Arg3.1 may contribute to addiction as a regulator of drug-taking vulnerability under different drug availability conditions.
Collapse
Affiliation(s)
- Rachel D Penrod
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Morgane Thomsen
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Makoto Taniguchi
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Yuhong Guo
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, United States of America; Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Christopher W Cowan
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Laura N Smith
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, United States of America; Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America.
| |
Collapse
|
42
|
Butler CW, Wilson YM, Mills SA, Gunnersen JM, Murphy M. Evidence that a defined population of neurons in lateral amygdala is directly involved in auditory fear learning and memory. Neurobiol Learn Mem 2019; 168:107139. [PMID: 31843653 DOI: 10.1016/j.nlm.2019.107139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 12/12/2019] [Indexed: 10/25/2022]
Abstract
Memory is thought to be encoded within networks of neurons within the brain, but the identity of the neurons involved and circuits they form have not been described for any memory. Previously, we used fos-tau-lacZ (FTL) transgenic mice to identify discrete populations of neurons in different regions of the brain which were specifically activated following fear conditioning. This suggested that these populations of neurons form nodes in a network that encodes fear memory. In particular, one population of learning activated neurons was found within a discrete region of the lateral amygdala (LA), a key nucleus required for fear conditioning. In order to provide evidence that this population is directly involved in fear conditioning, we have analysed the expression of a key molecular requirement for fear conditioning in LA, phosphorylated Extracellular Signal Regulated Kinase 1 and 2 (pERK1/2). The only neurons in LA that specifically expressed pERK1/2 following auditory fear conditioning were in the ventrolateral nucleus of the LA (LAvl), in the same discrete region where we found learning specific FTL+ neurons. Double labelling experiments in FTL mice showed that a substantial proportion of the learning activated neurons expressed both pERK1/2 and FTL. These experiments provide clear evidence that the learning specific neurons we identified within LAvl are directly involved in auditory fear conditioning. In addition, learning specific expression of pERK1/2 was found in a dense network of dendrites contained within the border region of the LAvl. This network of dendrites may represent an activated dendritic field involved in fear conditioning in LA.
Collapse
Affiliation(s)
- Christopher W Butler
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Yvette M Wilson
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Samuel A Mills
- Biological Optical Microscopy Platform, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Jenny M Gunnersen
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Mark Murphy
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria 3010, Australia.
| |
Collapse
|
43
|
The Emergence of a Stable Neuronal Ensemble from a Wider Pool of Activated Neurons in the Dorsal Medial Prefrontal Cortex during Appetitive Learning in Mice. J Neurosci 2019; 40:395-410. [PMID: 31727794 DOI: 10.1523/jneurosci.1496-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 11/21/2022] Open
Abstract
Animals selectively respond to environmental cues associated with food reward to optimize nutrient intake. Such appetitive conditioned stimulus-unconditioned stimulus (CS-US) associations are thought to be encoded in select, stable neuronal populations or neuronal ensembles, which undergo physiological modifications during appetitive conditioning. These ensembles in the medial prefrontal cortex (mPFC) control well-established, cue-evoked food seeking, but the mechanisms involved in the genesis of these ensembles are unclear. Here, we used male Fos-GFP mice that express green fluorescent protein (GFP) in recently behaviorally activated neurons, to reveal how dorsal mPFC neurons are recruited and modified to encode CS-US memory representations using an appetitive conditioning task. In the initial conditioning session, animals did not exhibit discriminated, cue-selective food seeking, but did so in later sessions indicating that a CS-US association was established. Using microprism-based in vivo 2-Photon imaging, we revealed that only a minority of neurons activated during the initial session was consistently activated throughout subsequent conditioning sessions and during cue-evoked memory recall. Notably, using ex vivo electrophysiology, we found that neurons activated following the initial session exhibited transient hyperexcitability. Chemogenetically enhancing the excitability of these neurons throughout subsequent conditioning sessions interfered with the development of reliable cue-selective food seeking, indicated by persistent, nondiscriminated performance. We demonstrate how appetitive learning consistently activates a subset of neurons to form a stable neuronal ensemble during the formation of a CS-US association. This ensemble may arise from a pool of hyperexcitable neurons activated during the initial conditioning session.SIGNIFICANCE STATEMENT Appetitive conditioning endows cues associated with food with the ability to guide food-seeking, through the formation of a food-cue association. Neuronal ensembles in the mPFC control established cue-evoked food-seeking. However, how neurons undergo physiological modifications and become part of an ensemble during conditioning remain unclear. We found that only a minority of dorsal mPFC neurons activated on the initial conditioning session became consistently activated during conditioning and memory recall. These initially activated neurons were also transiently hyperexcitable. We demonstrate the following: (1) how stable neuronal ensemble formation in the dorsal mPFC underlies appetitive conditioning; and (2) how this ensemble may arise from hyperexcitable neurons activated before the establishment of cue-evoked food seeking.
Collapse
|
44
|
Hoffman AN, Lam J, Hovda DA, Giza CC, Fanselow MS. Sensory sensitivity as a link between concussive traumatic brain injury and PTSD. Sci Rep 2019; 9:13841. [PMID: 31554865 PMCID: PMC6761112 DOI: 10.1038/s41598-019-50312-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most common injuries to military personnel, a population often exposed to stressful stimuli and emotional trauma. Changes in sensory processing after TBI might contribute to TBI-post traumatic stress disorder (PTSD) comorbidity. Combining an animal model of TBI with an animal model of emotional trauma, we reveal an interaction between auditory sensitivity after TBI and fear conditioning where 75 dB white noise alone evokes a phonophobia-like phenotype and when paired with footshocks, fear is robustly enhanced. TBI reduced neuronal activity in the hippocampus but increased activity in the ipsilateral lateral amygdala (LA) when exposed to white noise. The white noise effect in LA was driven by increased activity in neurons projecting from ipsilateral auditory thalamus (medial geniculate nucleus). These data suggest that altered sensory processing within subcortical sensory-emotional circuitry after TBI results in neutral stimuli adopting aversive properties with a corresponding impact on facilitating trauma memories and may contribute to TBI-PTSD comorbidity.
Collapse
Affiliation(s)
- Ann N Hoffman
- UCLA, Neurosurgery; Brain Injury Research Center, Los Angeles, USA.
- UCLA, Psychology, Los Angeles, USA.
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, USA.
- Staglin Center for Brain and Behavioral Health, Life Sciences, UCLA, Los Angeles, USA.
| | | | - David A Hovda
- UCLA, Neurosurgery; Brain Injury Research Center, Los Angeles, USA
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, USA
- UCLA, Medical and Molecular Pharmacology, Los Angeles, USA
| | - Christopher C Giza
- UCLA, Neurosurgery; Brain Injury Research Center, Los Angeles, USA
- UCLA Steve Tisch BrainSPORT Program, Los Angeles, USA
- UCLA Mattel Children's Hospital, Los Angeles, USA
| | - Michael S Fanselow
- UCLA, Psychology, Los Angeles, USA
- UCLA, Psychiatry and Biobehavioral Sciences, Los Angeles, USA
- Staglin Center for Brain and Behavioral Health, Life Sciences, UCLA, Los Angeles, USA
| |
Collapse
|
45
|
Frankland PW, Josselyn SA, Köhler S. The neurobiological foundation of memory retrieval. Nat Neurosci 2019; 22:1576-1585. [PMID: 31551594 DOI: 10.1038/s41593-019-0493-1] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
Abstract
Memory retrieval involves the interaction between external sensory or internally generated cues and stored memory traces (or engrams) in a process termed 'ecphory'. While ecphory has been examined in human cognitive neuroscience research, its neurobiological foundation is less understood. To the extent that ecphory involves 'reawakening' of engrams, leveraging recently developed technologies that can identify and manipulate engrams in rodents provides a fertile avenue for examining retrieval at the level of neuronal ensembles. Here we evaluate emerging neuroscientific research of this type, using cognitive theory as a guiding principle to organize and interpret initial findings. Our Review highlights the critical interaction between engrams and retrieval cues (environmental or artificial) for memory accessibility and retrieval success. These findings also highlight the intimate relationship between the mechanisms important in forming engrams and those important in their recovery, as captured in the cognitive notion of 'encoding specificity'. Finally, we identify several questions that currently remain unanswered.
Collapse
Affiliation(s)
- Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada. .,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada. .,Department of Psychology, University of Toronto, Toronto, Ontario, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, Canada. .,Child & Brain Development Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada.
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.,Department of Psychology, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Brain, Mind & Consciousness Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Stefan Köhler
- Department of Psychology, University of Western Ontario, London, Ontario, Canada. .,The Brain and Mind Institute, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
46
|
Troyner F, Bicca MA, Bertoglio LJ. Nucleus reuniens of the thalamus controls fear memory intensity, specificity and long-term maintenance during consolidation. Hippocampus 2019; 28:602-616. [PMID: 29747244 DOI: 10.1002/hipo.22964] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 01/05/2023]
Abstract
The thalamic nucleus reuniens (NR) has been shown to support bidirectional medial prefrontal cortex-hippocampus communication and synchronization relevant for cognitive processing. Using non-selective or prolonged inactivation of the NR, previous studies reported its activity positively modulates aversive memory consolidation. Here we examined the NR's role in consolidating contextual fear memories with varied strength, at both recent and more remote time points, using muscimol-induced temporary inactivation in rats. Results indicate the NR negatively modulates fear memory intensity, specificity, and long-term maintenance. The more intense, generalized, and enduring fear memory induced by NR inactivation during consolidation was less prone to behavioral suppression by extinction or reconsolidation disruption induced by clonidine, an alpha-2 adrenergic receptor agonist. Lastly, we used immunohistochemistry for Arc protein, which is involved in synaptic modifications underlying memory consolidation, to investigate whether treatment condition and/or conditioning status could change its levels not only in the NR, but also in the hippocampus (dorsal and ventral CA1 subregions) and the medial prefrontal cortex (anterior cingulate, prelimbic and infralimbic subregions). Results indicate a significant imbalance in the number of Arc-expressing neurons in the brain areas investigated in muscimol fear conditioned animals when compared with controls. Collectively, present results provide convergent evidence for the NR's role as a hub regulating quantitative and qualitative aspects of a contextual fear memory during its consolidation that seem to influence the subsequent susceptibility to experimental interventions aiming at attenuating its expression. They also indicate the selectivity and duration of a given inactivation approach may influence its outcomes.
Collapse
Affiliation(s)
- Fernanda Troyner
- Departmento de Farmacologia, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | - Maíra A Bicca
- Departmento de Farmacologia, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| | - Leandro J Bertoglio
- Departmento de Farmacologia, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil
| |
Collapse
|
47
|
Chandra N, Awasthi R, Ozdogan T, Johenning FW, Imbrosci B, Morris G, Schmitz D, Barkai E. A Cellular Mechanism Underlying Enhanced Capability for Complex Olfactory Discrimination Learning. eNeuro 2019; 6:ENEURO.0198-18.2019. [PMID: 30783614 PMCID: PMC6378325 DOI: 10.1523/eneuro.0198-18.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 12/26/2018] [Accepted: 01/06/2019] [Indexed: 11/21/2022] Open
Abstract
The biological mechanisms underlying complex forms of learning requiring the understanding of rules based on previous experience are not yet known. Previous studies have raised the intriguing possibility that improvement in complex learning tasks requires the long-term modulation of intrinsic neuronal excitability, induced by reducing the conductance of the slow calcium-dependent potassium current (sIAHP) simultaneously in most neurons in the relevant neuronal networks in several key brain areas. Such sIAHP reduction is expressed in attenuation of the postburst afterhyperpolarization (AHP) potential, and thus in enhanced repetitive action potential firing. Using complex olfactory discrimination (OD) learning as a model for complex learning, we show that brief activation of the GluK2 subtype glutamate receptor results in long-lasting enhancement of neuronal excitability in neurons from controls, but not from trained rats. Such an effect can be obtained by a brief tetanic synaptic stimulation or by direct application of kainate, both of which reduce the postburst AHP in pyramidal neurons. Induction of long-lasting enhancement of neuronal excitability is mediated via a metabotropic process that requires PKC and ERK activation. Intrinsic neuronal excitability cannot be modulated by synaptic activation in neurons from GluK2 knock-out mice. Accordingly, these mice are incapable of learning the complex OD task. Moreover, viral-induced overexpression of Gluk2 in piriform cortex pyramidal neurons results in remarkable enhancement of complex OD learning. Thus, signaling via kainate receptors has a central functional role in higher cognitive abilities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Edi Barkai
- University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
48
|
Meloni EG, Kaye KT, Venkataraman A, Carlezon WA. PACAP increases Arc/Arg 3.1 expression within the extended amygdala after fear conditioning in rats. Neurobiol Learn Mem 2018; 157:24-34. [PMID: 30458282 DOI: 10.1016/j.nlm.2018.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 11/09/2018] [Accepted: 11/16/2018] [Indexed: 11/24/2022]
Abstract
The stress-related neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is implicated in neuromodulation of learning and memory. PACAP can alter synaptic plasticity and has direct actions on neurons in the amygdala and hippocampus that could contribute to its acute and persistent effects on the consolidation and expression of conditioned fear. We recently demonstrated that intracerebroventricular (ICV) infusion of PACAP prior to fear conditioning (FC) results in initial amnestic-like effects followed by hyper-expression of conditioned freezing with repeated testing, and analyses of immediate-early gene c-Fos expression suggested that the central nucleus of the amygdala (CeA), but not the lateral/basolateral amygdala (LA/BLA) or hippocampus, are involved in these PACAP effects. Here, we extend that work by examining the expression of the synaptic plasticity marker activity-regulated cytoskeleton-associated protein (Arc/Arg 3.1) after PACAP administration and FC. Male Sprague-Dawley rats were implanted with cannula for ICV infusion of PACAP-38 (1.5 µg) or vehicle followed by FC and tests for conditioned freezing. One hour after FC, Arc protein expression was significantly elevated in the CeA and bed nucleus of the stria terminalis (BNST), interconnected structures that are key elements of the extended amygdala, in rats that received the combination of PACAP + FC. In contrast, Arc expression within the subdivisions of the hippocampus, or the LA/BLA, were unchanged. A subpopulation of Arc-positive cells in both the CeA and BNST also express PKCdelta, an intracellular marker that has been used to identify microcircuits that gate conditioned fear in the CeA. Consistent with our previous findings, on the following day conditioned freezing behavior was reduced in rats that had been given the combination of PACAP + FC-an amnestic-like effect-and Arc expression levels had returned to baseline. Given the established role of Arc in modifying synaptic plasticity and memory formation, our findings suggest that PACAP-induced overexpression of Arc following fear conditioning may disrupt neuroplastic changes within populations of CeA and BNST neurons normally responsible for encoding fear-related cues that, in this case, results in altered fear memory consolidation. Hence, PACAP systems may represent an axis on which stress and experience-driven neurotransmission converge to alter emotional memory, and mediate pathologies that are characteristic of psychiatric illnesses such as post-traumatic stress disorder.
Collapse
Affiliation(s)
- Edward G Meloni
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA 02478, United States.
| | - Karen T Kaye
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA 02478, United States
| | - Archana Venkataraman
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA 02478, United States
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA 02478, United States
| |
Collapse
|
49
|
Reuveni I, Barkai E. Tune it in: mechanisms and computational significance of neuron-autonomous plasticity. J Neurophysiol 2018; 120:1781-1795. [DOI: 10.1152/jn.00102.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The activity of a neural network is a result of synaptic signals that convey the communication between neurons and neuron-based intrinsic currents that determine the neuron’s input-output transfer function. Ample studies have demonstrated that cell-based excitability, and in particular intrinsic excitability, is modulated by learning and that these modifications play a key role in learning-related behavioral changes. The field of cell-based plasticity is largely growing, and it entails numerous experimental findings that demonstrate a large diversity of currents that are affected by learning. The diverse effect of learning on the neuron’s excitability emphasizes the need for a framework under which cell-based plasticity can be categorized to enable the assessment of the computational roles of the intrinsic modifications. We divide the domain of cell-based plasticity into three main categories, where the first category entails the currents that mediate the passive properties and single-spike generation, the second category entails the currents that mediate spike frequency adaptation, and the third category entails a novel learning-induced mechanism where all excitatory and inhibitory synapses double their strength. Curiously, this elementary division enables a natural categorization of the computational roles of these learning-induced plasticities. The computational roles are diverse and include modification of the neuronal mode of action, such as bursting, prolonged, and fast responsive; attention-like effect where the signal detection is improved; transfer of the network into an active state; biasing the competition for memory allocation; and transforming an environmental cue into a dominant cue and enabling a quicker formation of new memories.
Collapse
Affiliation(s)
- Iris Reuveni
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
50
|
Synaptic encoding of fear memories in the amygdala. Curr Opin Neurobiol 2018; 54:54-59. [PMID: 30216780 DOI: 10.1016/j.conb.2018.08.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/20/2018] [Indexed: 01/19/2023]
Abstract
Over the years Pavlovian fear conditioning has proved to be a powerful model to investigate the neural underpinnings of aversive associative memory formation. Although it is well appreciated that plasticity occurring at excitatory synapses within the basolateral complex of the amygdala (BLA) plays a critical role in associative memory formation, recent evidence suggests that plasticity within the amygdala is more distributed than previously appreciated. In particular, studies demonstrate that plasticity in the central nucleus (CeA) is critical for the acquisition of conditioned fear. In addition, a variety of interneuron populations within the amygdala, defined by unique neurochemical markers, contribute to distinct aspects of stimulus processing and memory formation during fear conditioning. Here, we will review and summarize recent advances in our understanding of amygdala networks and how unique players within this network contribute to synaptic plasticity associated with the acquisition of conditioned fear.
Collapse
|