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
|
Brida KL, Day JJ. Molecular and genetic mechanisms of plasticity in addiction. Curr Opin Neurobiol 2025; 93:103032. [PMID: 40311544 DOI: 10.1016/j.conb.2025.103032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
Drugs of abuse result in well-characterized changes in synapse function and number in brain reward regions such as the nucleus accumbens. However, recent reports demonstrate that only a small fraction of neurons in the nucleus accumbens are activated in response to psychostimulants such as cocaine. While these "ensemble" neurons are marked by drug-related transcriptional changes in immediate early genes, the mechanisms that ultimately link these early changes to enduring molecular alterations in the same neurons are less clear. In this review, we 1) describe potential mechanisms underlying regulation of diverse plasticity-related gene programs across drug-activated ensembles, 2) discuss factors conferring ensemble recruitment bias within seemingly homogeneous populations, and 3) speculate on the role of chromatin and epigenetic modifiers in gating metaplastic state transitions that contribute to addiction.
Collapse
Affiliation(s)
- Kasey L Brida
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd, SHEL 910, Birmingham, AL 35294, USA
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Blvd, SHEL 910, Birmingham, AL 35294, USA.
| |
Collapse
|
3
|
Brida KL, Jorgensen ET, Phillips RA, Newman CE, Tuscher JJ, Morring EK, Zipperly ME, Ianov L, Montgomery KD, Tippani M, Hyde TM, Maynard KR, Martinowich K, Day JJ. Reelin marks cocaine-activated striatal neurons, promotes neuronal excitability, and regulates cocaine reward. SCIENCE ADVANCES 2025; 11:eads4441. [PMID: 40138397 PMCID: PMC12076537 DOI: 10.1126/sciadv.ads4441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025]
Abstract
Drugs of abuse activate defined neuronal populations in reward structures such as the nucleus accumbens (NAc), which promote the enduring synaptic, circuit, and behavioral consequences of drug exposure. While the molecular and cellular effects arising from experience with drugs like cocaine are increasingly well understood, mechanisms that dictate NAc neuronal recruitment remain unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling and targeted in situ detection to identify Reln (encoding the secreted glycoprotein, Reelin) as a marker of cocaine-activated neuronal populations within the rat NAc. A CRISPR interference approach enabling selective Reln knockdown in the adult NAc altered expression of calcium signaling genes, promoted a transcriptional trajectory consistent with loss of cocaine sensitivity, and decreased MSN excitability. Behaviorally, Reln knockdown prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. These results identify Reelin as a critical mechanistic link between neuronal activation and cocaine-induced behavioral adaptations.
Collapse
Affiliation(s)
- Kasey L. Brida
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily T. Jorgensen
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert A. Phillips
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Catherine E. Newman
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Emily K. Morring
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center,
University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kelsey D. Montgomery
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neurology, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development,
Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral
Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205,
USA
- Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD 21205, USA
- The Kavli Neuroscience Discovery
Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of
Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
4
|
Thibeault KC, Leonard MZ, Kondev V, Emerson SD, Bethi R, Lopez AJ, Sens JP, Nabit BP, Elam HB, Winder DG, Patel S, Kiraly DD, Grueter BA, Calipari ES. A Cocaine-Activated Ensemble Exerts Increased Control Over Behavior While Decreasing in Size. Biol Psychiatry 2025; 97:590-601. [PMID: 38901723 PMCID: PMC11995305 DOI: 10.1016/j.biopsych.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Substance use disorder is characterized by long-lasting changes in reward-related brain regions, such as the nucleus accumbens. Previous work has shown that cocaine exposure induces plasticity in broad, genetically defined cell types in the nucleus accumbens; however, in response to a stimulus, only a small percentage of neurons are transcriptionally active-termed an ensemble. Here, we identify an Arc-expressing neuronal ensemble that has a unique trajectory of recruitment and causally controls drug self-administration after repeated, but not acute, cocaine exposure. METHODS Using Arc-CreERT2 transgenic mice, we expressed transgenes in Arc+ ensembles activated by cocaine exposure (either acute [1 × 10 mg/kg intraperitoneally] or repeated [10 × 10 mg/kg intraperitoneally]). Using genetic, optical, and physiological recording and manipulation strategies, we assessed the contribution of these ensembles to behaviors associated with substance use disorder. RESULTS Repeated cocaine exposure reduced the size of the ensemble while simultaneously increasing its control over behavior. Neurons within the repeated cocaine ensemble were hyperexcitable, and their optogenetic excitation was sufficient for reinforcement. Finally, lesioning the repeated cocaine, but not the acute cocaine, ensemble blunted cocaine self-administration. Thus, repeated cocaine exposure reduced the size of the ensemble while simultaneously increasing its contributions to drug reinforcement. CONCLUSIONS We showed that repeated, but not acute, cocaine exposure induced a physiologically distinct ensemble characterized by the expression of the immediate early gene Arc, which was uniquely capable of modulating reinforcement behavior.
Collapse
Affiliation(s)
- Kimberly C Thibeault
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee
| | - Michael Z Leonard
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Veronika Kondev
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee
| | - Soren D Emerson
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee
| | - Rishik Bethi
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee
| | - Alberto J Lopez
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Jonathon P Sens
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Brett P Nabit
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Hannah B Elam
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Danny G Winder
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee; Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt JF Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Sachin Patel
- Department of Psychiatry, Northwestern University, Chicago, Illinois
| | - Drew D Kiraly
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Brad A Grueter
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee; Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Erin S Calipari
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee; Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt JF Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee.
| |
Collapse
|
5
|
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
|
6
|
Balmer GL, Guha S, Poll S. Engrams across diseases: Different pathologies - unifying mechanisms? Neurobiol Learn Mem 2025; 219:108036. [PMID: 40023216 DOI: 10.1016/j.nlm.2025.108036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Memories are our reservoir of knowledge and thus, are crucial for guiding decisions and defining our self. The physical correlate of a memory in the brain is termed an engram and since decades helps researchers to elucidate the intricate nature of our imprinted experiences and knowledge. Given the importance that memories have for our lives, their impairment can present a tremendous burden. In this review we aim to discuss engram malfunctioning across diseases, covering dementia-associated pathologies, epilepsy, chronic pain and psychiatric disorders. Current neuroscientific tools allow to witness the emergence and fate of engram cells and enable their manipulation. We further suggest that specific mechanisms of mnemonic malfunction can be derived from engram cell readouts. While depicting the way diseases act on the mnemonic component - specifically, on the cellular engram - we emphasize a differentiation between forms of amnesia and hypermnesia. Finally, we highlight commonalities and distinctions of engram impairments on the cellular level across diseases independent of their pathogenic origins and discuss prospective therapeutic measures.
Collapse
Affiliation(s)
- Greta Leonore Balmer
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Shuvrangshu Guha
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Stefanie Poll
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE) Bonn, Germany.
| |
Collapse
|
7
|
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
|
8
|
Fujikawa R, Ramsaran AI, Guskjolen A, de la Parra J, Zou Y, Mocle AJ, Josselyn SA, Frankland PW. Neurogenesis-dependent remodeling of hippocampal circuits reduces PTSD-like behaviors in adult mice. Mol Psychiatry 2024; 29:3316-3329. [PMID: 38719894 DOI: 10.1038/s41380-024-02585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 11/08/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a hypermnesic condition that develops in a subset of individuals following exposure to severe trauma. PTSD symptoms are debilitating, and include increased anxiety, abnormal threat generalization, and impaired extinction. In developing treatment strategies for PTSD, preclinical studies in rodents have largely focused on interventions that target post-encoding memory processes such as reconsolidation and extinction. Instead, here we focus on forgetting, another post-encoding process that regulates memory expression. Using a double trauma murine model for PTSD, we asked whether promoting neurogenesis-mediated forgetting can weaken trauma memories and associated PTSD-relevant behavioral phenotypes. In the double trauma paradigm, consecutive aversive experiences lead to a constellation of behavioral phenotypes associated with PTSD including increases in anxiety-like behavior, abnormal threat generalization, and deficient extinction. We found that post-training interventions that elevate hippocampal neurogenesis weakened the original trauma memory and decreased these PTSD-relevant phenotypes. These effects were observed using multiple methods to manipulate hippocampal neurogenesis, including interventions restricted to neural progenitor cells that selectively promoted integration of adult-generated granule cells into hippocampal circuits. The same interventions also weakened cocaine place preference memories, suggesting that promoting hippocampal neurogenesis may represent a broadly useful approach in hypermnesic conditions such as PTSD and substance abuse disorders.
Collapse
Affiliation(s)
- Risako Fujikawa
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Adam I Ramsaran
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Axel Guskjolen
- 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, M5G 1X8, Canada
| | - Juan de la Parra
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Yi Zou
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Andrew J Mocle
- 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, M5G 1X8, Canada
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Child & Brain Development Program, Canadian Institute for Advanced Research, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
9
|
Li H, Liuha X, Chen R, Xiao Y, Xu W, Zhou Y, Bai L, Zhang J, Zhao Y, Zhao Y, Wang L, Qin F, Chen Y, Han S, Wei Q, Li S, Zhang D, Bu Q, Wang X, Jiang L, Dai Y, Zhang N, Kuang W, Qin M, Wang H, Tian J, Zhao Y, Cen X. Pyruvate dehydrogenase complex E1 subunit α crotonylation modulates cocaine-associated memory through hippocampal neuron activation. Cell Rep 2024; 43:114529. [PMID: 39046876 DOI: 10.1016/j.celrep.2024.114529] [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: 01/08/2024] [Revised: 06/04/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Neuronal activation is required for the formation of drug-associated memory, which is critical for the development, persistence, and relapse of drug addiction. Nevertheless, the metabolic mechanisms underlying energy production for neuronal activation remain poorly understood. In the study, a large-scale proteomics analysis of lysine crotonylation (Kcr), a type of protein posttranslational modification (PTM), reveals that cocaine promoted protein Kcr in the hippocampal dorsal dentate gyrus (dDG). We find that Kcr is predominantly discovered in a few enzymes critical for mitochondrial energy metabolism; in particular, pyruvate dehydrogenase (PDH) complex E1 subunit α (PDHA1) is crotonylated at the lysine 39 (K39) residue through P300 catalysis. Crotonylated PDHA1 promotes pyruvate metabolism by activating PDH to increase ATP production, thus providing energy for hippocampal neuronal activation and promoting cocaine-associated memory recall. Our findings identify Kcr of PDHA1 as a PTM that promotes pyruvate metabolism to enhance neuronal activity for cocaine-associated memory.
Collapse
Affiliation(s)
- Hongchun Li
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoyu Liuha
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Rong Chen
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhou Xiao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Xu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Yuanyi Zhou
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Bai
- Histology and Imaging Platform, Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhang
- Histology and Imaging Platform, Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Zhao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Zhao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Wang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Qin
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaxing Chen
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuang Han
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingfan Wei
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Li
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dingwen Zhang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qian Bu
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; West China-Frontier PharmaTech Co., Ltd., Chengdu 610041, China
| | - Xiaojie Wang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linhong Jiang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Dai
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ni Zhang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weihong Kuang
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Qin
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongbo Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jingwei Tian
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yinglan Zhao
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaobo Cen
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Park S, Ko SY, Frankland PW, Josselyn SA. Comparing behaviours induced by natural memory retrieval and optogenetic reactivation of an engram ensemble in mice. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230227. [PMID: 38853560 PMCID: PMC11343273 DOI: 10.1098/rstb.2023.0227] [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: 11/14/2023] [Revised: 03/06/2024] [Accepted: 04/09/2024] [Indexed: 06/11/2024] Open
Abstract
Memories are thought to be stored within sparse collections of neurons known as engram ensembles. Neurons active during a training episode are allocated to an engram ensemble ('engram neurons'). Memory retrieval is initiated by external sensory or internal cues present at the time of training reactivating engram neurons. Interestingly, optogenetic reactivation of engram ensemble neurons alone in the absence of external sensory cues is sufficient to induce behaviour consistent with memory retrieval in mice. However, there may exist differences between the behaviours induced by natural retrieval cues or artificial engram reactivation. Here, we compared two defensive behaviours (freezing and the syllable structure of ultrasonic vocalizations, USVs) induced by sensory cues present at training (natural memory retrieval) and optogenetic engram ensemble reactivation (artificial memory retrieval) in a threat conditioning paradigm in the same mice. During natural memory recall, we observed a strong positive correlation between freezing levels and distinct USV syllable features (characterized by an unsupervised algorithm, MUPET (Mouse Ultrasonic Profile ExTraction)). Moreover, we observed strikingly similar behavioural profiles in terms of freezing and USV characteristics between natural memory recall and artificial memory recall in the absence of sensory retrieval cues. Although our analysis focused on two behavioural measures of threat memory (freezing and USV characteristics), these results underscore the similarities between threat memory recall triggered naturally and through optogenetic reactivation of engram ensembles. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Sungmo Park
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, OntarioM5G 1X8, Canada
| | - Sang Yoon Ko
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, OntarioM5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, OntarioM5G 1X8, Canada
| | - Paul W. Frankland
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, OntarioM5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, OntarioM5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, OntarioM5G 1X8, Canada
| | - Sheena A. Josselyn
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, OntarioM5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, OntarioM5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, OntarioM5G 1X8, Canada
| |
Collapse
|
11
|
Brida KL, Jorgensen ET, Phillips RA, Newman CE, Tuscher JJ, Morring EK, Zipperly ME, Ianov L, Montgomery KD, Tippani M, Hyde TM, Maynard KR, Martinowich K, Day JJ. Reelin marks cocaine-activated striatal ensembles, promotes neuronal excitability, and regulates cocaine reward. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599348. [PMID: 38948801 PMCID: PMC11212904 DOI: 10.1101/2024.06.17.599348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Drugs of abuse activate defined neuronal ensembles in brain reward structures such as the nucleus accumbens (NAc), which are thought to promote the enduring synaptic, circuit, and behavioral consequences of drug exposure. While the molecular and cellular effects arising from experience with drugs like cocaine are increasingly well understood, the mechanisms that sculpt NAc ensemble participation are largely unknown. Here, we leveraged unbiased single-nucleus transcriptional profiling to identify expression of the secreted glycoprotein Reelin (encoded by the Reln gene) as a marker of cocaine-activated neuronal ensembles within the rat NAc. Multiplexed in situ detection confirmed selective expression of the immediate early gene Fos in Reln+ neurons after cocaine experience, and also revealed enrichment of Reln mRNA in Drd1 + medium spiny neurons (MSNs) in both the rat and human brain. Using a novel CRISPR interference strategy enabling selective Reln knockdown in the adult NAc, we observed altered expression of genes linked to calcium signaling, emergence of a transcriptional trajectory consistent with loss of cocaine sensitivity, and a striking decrease in MSN intrinsic excitability. At the behavioral level, loss of Reln prevented cocaine locomotor sensitization, abolished cocaine place preference memory, and decreased cocaine self-administration behavior. Together, these results identify Reelin as a critical mechanistic link between ensemble participation and cocaine-induced behavioral adaptations.
Collapse
|
12
|
Liu X, Lu T, Chen X, Huang S, Zheng W, Zhang W, Meng S, Yan W, Shi L, Bao Y, Xue Y, Shi J, Yuan K, Han Y, Lu L. Memory consolidation drives the enhancement of remote cocaine memory via prefrontal circuit. Mol Psychiatry 2024; 29:730-741. [PMID: 38221548 DOI: 10.1038/s41380-023-02364-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024]
Abstract
Remote memory usually decreases over time, whereas remote drug-cue associated memory exhibits enhancement, increasing the risk of relapse during abstinence. Memory system consolidation is a prerequisite for remote memory formation, but neurobiological underpinnings of the role of consolidation in the enhancement of remote drug memory are unclear. Here, we found that remote cocaine-cue associated memory was enhanced in rats that underwent self-administration training, together with a progressive increase in the response of prelimbic cortex (PrL) CaMKII neurons to cues. System consolidation was required for the enhancement of remote cocaine memory through PrL CaMKII neurons during the early period post-training. Furthermore, dendritic spine maturation in the PrL relied on the basolateral amygdala (BLA) input during the early period of consolidation, contributing to remote memory enhancement. These findings indicate that memory consolidation drives the enhancement of remote cocaine memory through a time-dependent increase in activity and maturation of PrL CaMKII neurons receiving a sustained BLA input.
Collapse
Affiliation(s)
- Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Xuan Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Wei Zheng
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Wei Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Le Shi
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China.
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Dongcheng, Beijing, China.
| |
Collapse
|
13
|
Xia F, Fascianelli V, Vishwakarma N, Ghinger FG, Fusi S, Kheirbek MA. Identifying and modulating neural signatures of stress susceptibility and resilience enables control of anhedonia. RESEARCH SQUARE 2024:rs.3.rs-3581329. [PMID: 38343839 PMCID: PMC10854313 DOI: 10.21203/rs.3.rs-3581329/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Anhedonia is a core aspect of major depressive disorder. Traditionally viewed as a blunted emotional state in which individuals are unable to experience joy, anhedonia also diminishes the drive to seek rewards and the ability to value and learn about them 1-4.The neural underpinnings of anhedonia and how this emotional state drives related behavioral changes remain unclear. Here, we investigated these questions by taking advantage of the fact that when mice are exposed to traumatic social stress, susceptible animals become socially withdrawn and anhedonic, where they cease to seek high-value rewards, while others remain resilient. By performing high density electrophysiological recordings and comparing neural activity patterns of these groups in the basolateral amygdala (BLA) and ventral CA1 (vCA1) of awake behaving animals, we identified neural signatures of susceptibility and resilience to anhedonia. When animals actively sought rewards, BLA activity in resilient mice showed stronger discrimination between upcoming reward choices. In contrast, susceptible mice displayed a rumination-like signature, where BLA neurons encoded the intention to switch or stay on a previously chosen reward. When animals were at rest, the spontaneous BLA activity of susceptible mice was higher dimensional than in controls, reflecting a greater number of distinct neural population states. Notably, this spontaneous activity allowed us to decode group identity and to infer if a mouse had a history of stress better than behavioral outcomes alone. Finally, targeted manipulation of vCA1 inputs to the BLA in susceptible mice rescued dysfunctional neural dynamics, amplified dynamics associated with resilience, and reversed their anhedonic behavior. This work reveals population-level neural signatures that explain individual differences in responses to traumatic stress, and suggests that modulating vCA1-BLA inputs can enhance resilience by regulating these dynamics.
Collapse
Affiliation(s)
- Frances Xia
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
| | - Valeria Fascianelli
- Center for Theoretical Neuroscience, Columbia University, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, NY, USA
| | - Nina Vishwakarma
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, USA
| | - Frances Grace Ghinger
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
| | - Stefano Fusi
- Center for Theoretical Neuroscience, Columbia University, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, NY, USA
- Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, NY, USA
- Kavli Institute for Brain Science, Columbia University Irving Medical Center, NY, USA
| | - Mazen A Kheirbek
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
14
|
Jiang C, Huang H, Yang X, Le Q, Liu X, Ma L, Wang F. Targeting mitochondrial dynamics of morphine-responsive dopaminergic neurons ameliorates opiate withdrawal. J Clin Invest 2024; 134:e171995. [PMID: 38236644 PMCID: PMC10904060 DOI: 10.1172/jci171995] [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: 05/03/2023] [Accepted: 01/11/2024] [Indexed: 03/02/2024] Open
Abstract
Converging studies demonstrate the dysfunction of the dopaminergic neurons following chronic opioid administration. However, the therapeutic strategies targeting opioid-responsive dopaminergic ensembles that contribute to the development of opioid withdrawal remain to be elucidated. Here, we used the neuronal activity-dependent Tet-Off system to label dopaminergic ensembles in response to initial morphine exposure (Mor-Ens) in the ventral tegmental area (VTA). Fiber optic photometry recording and transcriptome analysis revealed downregulated spontaneous activity and dysregulated mitochondrial respiratory, ultrastructure, and oxidoreductase signal pathways after chronic morphine administration in these dopaminergic ensembles. Mitochondrial fragmentation and the decreased mitochondrial fusion gene mitofusin 1 (Mfn1) were found in these ensembles after prolonged opioid withdrawal. Restoration of Mfn1 in the dopaminergic Mor-Ens attenuated excessive oxidative stress and the development of opioid withdrawal. Administration of Mdivi-1, a mitochondrial fission inhibitor, ameliorated the mitochondrial fragmentation and maladaptation of the neuronal plasticity in these Mor-Ens, accompanied by attenuated development of opioid withdrawal after chronic morphine administration, without affecting the analgesic effect of morphine. These findings highlighted the plastic architecture of mitochondria as a potential therapeutic target for opioid analgesic-induced substance use disorders.
Collapse
Affiliation(s)
- Changyou Jiang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Basic Medical Sciences, Departments of Neurosurgery and Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Han Huang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Basic Medical Sciences, Departments of Neurosurgery and Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Xiao Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Basic Medical Sciences, Departments of Neurosurgery and Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Qiumin Le
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Basic Medical Sciences, Departments of Neurosurgery and Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Xing Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Basic Medical Sciences, Departments of Neurosurgery and Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Lan Ma
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Basic Medical Sciences, Departments of Neurosurgery and Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Feifei Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and School of Basic Medical Sciences, Departments of Neurosurgery and Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| |
Collapse
|
15
|
Clarke-Williams CJ, Lopes-Dos-Santos V, Lefèvre L, Brizee D, Causse AA, Rothaermel R, Hartwich K, Perestenko PV, Toth R, McNamara CG, Sharott A, Dupret D. Coordinating brain-distributed network activities in memory resistant to extinction. Cell 2024; 187:409-427.e19. [PMID: 38242086 PMCID: PMC7615560 DOI: 10.1016/j.cell.2023.12.018] [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: 10/12/2022] [Revised: 07/13/2023] [Accepted: 12/13/2023] [Indexed: 01/21/2024]
Abstract
Certain memories resist extinction to continue invigorating maladaptive actions. The robustness of these memories could depend on their widely distributed implementation across populations of neurons in multiple brain regions. However, how dispersed neuronal activities are collectively organized to underpin a persistent memory-guided behavior remains unknown. To investigate this, we simultaneously monitored the prefrontal cortex, nucleus accumbens, amygdala, hippocampus, and ventral tegmental area (VTA) of the mouse brain from initial recall to post-extinction renewal of a memory involving cocaine experience. We uncover a higher-order pattern of short-lived beta-frequency (15-25 Hz) activities that are transiently coordinated across these networks during memory retrieval. The output of a divergent pathway from upstream VTA glutamatergic neurons, paced by a slower (4-Hz) oscillation, actuates this multi-network beta-band coactivation; its closed-loop phase-informed suppression prevents renewal of cocaine-biased behavior. Binding brain-distributed neural activities in this temporally structured manner may constitute an organizational principle of robust memory expression.
Collapse
Affiliation(s)
- Charlie J Clarke-Williams
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| | - Vítor Lopes-Dos-Santos
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Laura Lefèvre
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Demi Brizee
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Adrien A Causse
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Roman Rothaermel
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Katja Hartwich
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Pavel V Perestenko
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Robert Toth
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Colin G McNamara
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - Andrew Sharott
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK
| | - David Dupret
- Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3TH, UK.
| |
Collapse
|
16
|
Pang B, Wu X, Chen H, Yan Y, Du Z, Yu Z, Yang X, Wang W, Lu K. Exploring the memory: existing activity-dependent tools to tag and manipulate engram cells. Front Cell Neurosci 2024; 17:1279032. [PMID: 38259503 PMCID: PMC10800721 DOI: 10.3389/fncel.2023.1279032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/17/2023] [Indexed: 01/24/2024] Open
Abstract
The theory of engrams, proposed several years ago, is highly crucial to understanding the progress of memory. Although it significantly contributes to identifying new treatments for cognitive disorders, it is limited by a lack of technology. Several scientists have attempted to validate this theory but failed. With the increasing availability of activity-dependent tools, several researchers have found traces of engram cells. Activity-dependent tools are based on the mechanisms underlying neuronal activity and use a combination of emerging molecular biological and genetic technology. Scientists have used these tools to tag and manipulate engram neurons and identified numerous internal connections between engram neurons and memory. In this review, we provide the background, principles, and selected examples of applications of existing activity-dependent tools. Using a combination of traditional definitions and concepts of engram cells, we discuss the applications and limitations of these tools and propose certain developmental directions to further explore the functions of engram cells.
Collapse
Affiliation(s)
- Bo Pang
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wu
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Hailun Chen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yiwen Yan
- School of Basic Medicine Science, Southern Medical University, Guangzhou, China
| | - Zibo Du
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Zihan Yu
- School of Basic Medicine Science, Southern Medical University, Guangzhou, China
| | - Xiai Yang
- Department of Neurology, Ankang Central Hospital, Ankang, China
| | - Wanshan Wang
- Laboratory Animal Management Center, Southern Medical University, Guangzhou, China
- Guangzhou Southern Medical Laboratory Animal Sci. and Tech. Co., Ltd., Guangzhou, China
| | - Kangrong Lu
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
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
|
18
|
Salery M, Godino A, Xu YQ, Fullard JF, Durand-de Cuttoli R, LaBanca AR, Holt LM, Russo SJ, Roussos P, Nestler EJ. Transcriptional correlates of cocaine-associated learning in striatal ARC ensembles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571585. [PMID: 38168167 PMCID: PMC10760161 DOI: 10.1101/2023.12.13.571585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Learned associations between the rewarding effects of drugs and the context in which they are experienced underlie context-induced relapse. Previous work demonstrates the importance of sparse neuronal populations - called neuronal ensembles - in associative learning and cocaine seeking, but it remains unknown whether the encoding vs. retrieval of cocaine-associated memories involves similar or distinct mechanisms of ensemble activation and reactivation in nucleus accumbens (NAc). We use ArcCreER T2 mice to establish that mostly distinct NAc ensembles are recruited by initial vs. repeated exposures to cocaine, which are then differentially reactivated and exert distinct effects during cocaine-related memory retrieval. Single-nuclei RNA-sequencing of these ensembles demonstrates predominant recruitment of D1 medium spiny neurons and identifies transcriptional properties that are selective to cocaine-recruited NAc neurons and could explain distinct excitability features. These findings fundamentally advance our understanding of how cocaine drives pathological memory formation during repeated exposures.
Collapse
|
19
|
Liu X, Wang F, Le Q, Ma L. Cellular and molecular basis of drug addiction: The role of neuronal ensembles in addiction. Curr Opin Neurobiol 2023; 83:102813. [PMID: 37972536 DOI: 10.1016/j.conb.2023.102813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
Addiction has been conceptualized as a disease of learning and memory. Learned associations between environmental cues and unconditioned rewards induced by drug administration, which play a critical role in addiction, have been shown to be encoded in sparsely distributed populations of neurons called neuronal ensembles. This review aims to highlight how synaptic remodeling and alterations in signaling pathways that occur specifically in neuronal ensembles contribute to the pathogenesis of addiction. Furthermore, a causal link between transcriptional and epigenetic modifications in neuronal ensembles and the development of the addictive state is proposed. Translational studies of molecular and cellular changes in neuronal ensembles that contribute to drug-seeking behavior, will allow the identification of molecular and circuit targets and interventions for substance use disorders.
Collapse
Affiliation(s)
- Xing Liu
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Feifei Wang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China.
| | - Qiumin Le
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| | - Lan Ma
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, China
| |
Collapse
|
20
|
Xia F, Fascianelli V, Vishwakarma N, Ghinger FG, Fusi S, Kheirbek MA. Neural signatures of stress susceptibility and resilience in the amygdala-hippocampal network. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563652. [PMID: 37961124 PMCID: PMC10634760 DOI: 10.1101/2023.10.23.563652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The neural dynamics that underlie divergent anhedonic responses to stress remain unclear. Here, we identified neuronal dynamics in an amygdala-hippocampal circuit that distinguish stress resilience and susceptibility. In a reward-choice task, basolateral amygdala (BLA) activity in resilient mice showed enhanced discrimination of upcoming reward choices. In contrast, a rumination-like signature emerged in the BLA of susceptible mice; a linear decoder could classify the intention to switch or stay on a previously chosen reward. Spontaneous activity in the BLA of susceptible mice was higher dimensional than controls, reflecting the exploration of a larger number of distinct neural states. Manipulation of vCA1-BLA inputs rescued dysfunctional neural dynamics and anhedonia in susceptible mice, suggesting that targeting this pathway can enhance BLA circuit function and ameliorate of depression-related behaviors.
Collapse
Affiliation(s)
- Frances Xia
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
| | - Valeria Fascianelli
- Center for Theoretical Neuroscience, Columbia University, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, NY, USA
| | - Nina Vishwakarma
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, USA
| | - Frances Grace Ghinger
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
| | - Stefano Fusi
- Center for Theoretical Neuroscience, Columbia University, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, NY, USA
- Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, NY, USA
- Kavli Institute for Brain Science, Columbia University Irving Medical Center, NY, USA
| | - Mazen A Kheirbek
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, USA
- Kavli Institute for Brain Science, Columbia University Irving Medical Center, NY, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
21
|
Balouek JA, Mclain CA, Minerva AR, Rashford RL, Bennett SN, Rogers FD, Peña CJ. Reactivation of Early-Life Stress-Sensitive Neuronal Ensembles Contributes to Lifelong Stress Hypersensitivity. J Neurosci 2023; 43:5996-6009. [PMID: 37429717 PMCID: PMC10451005 DOI: 10.1523/jneurosci.0016-23.2023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/01/2023] [Accepted: 06/17/2023] [Indexed: 07/12/2023] Open
Abstract
Early-life stress (ELS) is one of the strongest lifetime risk factors for depression, anxiety, suicide, and other psychiatric disorders, particularly after facing additional stressful events later in life. Human and animal studies demonstrate that ELS sensitizes individuals to subsequent stress. However, the neurobiological basis of such stress sensitization remains largely unexplored. We hypothesized that ELS-induced stress sensitization would be detectable at the level of neuronal ensembles, such that cells activated by ELS would be more reactive to adult stress. To test this, we leveraged transgenic mice to genetically tag, track, and manipulate experience-activated neurons. We found that in both male and female mice, ELS-activated neurons within the nucleus accumbens (NAc), and to a lesser extent the medial prefrontal cortex, were preferentially reactivated by adult stress. To test whether reactivation of ELS-activated ensembles in the NAc contributes to stress hypersensitivity, we expressed hM4Dis receptor in control or ELS-activated neurons of pups and chemogenetically inhibited their activity during experience of adult stress. Inhibition of ELS-activated NAc neurons, but not control-tagged neurons, ameliorated social avoidance behavior following chronic social defeat stress in males. These data provide evidence that ELS-induced stress hypersensitivity is encoded at the level of corticolimbic neuronal ensembles.SIGNIFICANCE STATEMENT Early-life stress enhances sensitivity to stress later in life, yet the mechanisms of such stress sensitization are largely unknown. Here, we show that neuronal ensembles in corticolimbic brain regions remain hypersensitive to stress across the life span, and quieting these ensembles during experience of adult stress rescues stress hypersensitivity.
Collapse
Affiliation(s)
- Julie-Anne Balouek
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Christabel A Mclain
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Adelaide R Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Rebekah L Rashford
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Shannon N Bennett
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Forrest D Rogers
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | | |
Collapse
|
22
|
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: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
23
|
Park A, Jacob AD, Hsiang HLL, Frankland PW, Howland JG, Josselyn SA. Formation and fate of an engram in the lateral amygdala supporting a rewarding memory in mice. Neuropsychopharmacology 2023; 48:724-733. [PMID: 36261624 PMCID: PMC10066178 DOI: 10.1038/s41386-022-01472-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/08/2022]
Abstract
Memories allow past experiences to guide future decision making and behavior. Sparse ensembles of neurons, known as engrams, are thought to store memories in the brain. Most previous research has focused on engrams supporting threatening or fearful memories where results show that neurons involved in a particular engram ("engram neurons") are both necessary and sufficient for memory expression. Far less is understood about engrams supporting appetitive or rewarding memories. As circumstances and environments are dynamic, the fate of a previously acquired engram with changing circumstances is unknown. Here we examined how engrams supporting a rewarding cue-cocaine memory are formed and whether this original engram is important in reinstatement of memory-guided behavior following extinction. Using a variety of techniques, we show that neurons in the lateral amygdala are allocated to an engram based on relative neuronal excitability at training. Furthermore, once allocated, these neurons become both necessary and sufficient for behavior consistent with recall of that rewarding memory. Allocated neurons are also critical for cocaine-primed reinstatement of memory-guided behavior following extinction. Moreover, artificial reactivation of initially allocated neurons supports reinstatement-like behavior following extinction even in the absence of cocaine-priming. Together, these findings suggest that cocaine priming after extinction reactivates the original engram, and that memory-guided reinstatement behavior does not occur in the absence of this reactivation. Although we focused on neurons in one brain region only, our findings that manipulations of lateral amygdala engram neurons alone were sufficient to impact memory-guided behavior indicate that the lateral amygdala is a critical hub region in what may be a larger brain-wide engram.
Collapse
Affiliation(s)
- Albert Park
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., 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
- Department of Psychology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Hwa-Lin Liz Hsiang
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., 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
- Department of Psychology, University of Toronto, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, M5G 1M1, Canada
| | - John G Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada.
- Department of Psychology, University of Toronto, Toronto, ON, M5G 1X8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
24
|
Chen L, Francisco TR, Baggetta AM, Zaki Y, Ramirez S, Clem RL, Shuman T, Cai DJ. Ensemble-specific deficit in neuronal intrinsic excitability in aged mice. Neurobiol Aging 2023; 123:92-97. [PMID: 36652783 PMCID: PMC9892234 DOI: 10.1016/j.neurobiolaging.2022.12.007] [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: 03/30/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
With the prevalence of age-related cognitive deficits on the rise, it is essential to identify cellular and circuit alterations that contribute to age-related memory impairment. Increased intrinsic neuronal excitability after learning is important for memory consolidation, and changes to this process could underlie memory impairment in old age. Some studies find age-related deficits in hippocampal neuronal excitability that correlate with memory impairment but others do not, possibly due to selective changes only in activated neural ensembles. Thus, we tagged CA1 neurons activated during learning and recorded their intrinsic excitability 5 hours or 7 days post-training. Adult mice exhibited increased neuronal excitability 5 hours after learning, specifically in ensemble (learning-activated) CA1 neurons. As expected, ensemble excitability returned to baseline 7 days post-training. In aged mice, there was no ensemble-specific excitability increase after learning, which was associated with impaired hippocampal memory performance. These results suggest that CA1 may be susceptible to age-related impairments in post-learning ensemble excitability and underscore the need to selectively measure ensemble-specific changes in the brain.
Collapse
Affiliation(s)
- Lingxuan Chen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Taylor R Francisco
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Austin M Baggetta
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yosif Zaki
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steve Ramirez
- Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Roger L Clem
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tristan Shuman
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Denise J Cai
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
25
|
Murthy BKB, Somatakis S, Ulivi AF, Klimmt H, Castello-Waldow TP, Haynes N, Huettl RE, Chen A, Attardo A. Arc-driven mGRASP highlights CA1 to CA3 synaptic engrams. Front Behav Neurosci 2023; 16:1072571. [PMID: 36793796 PMCID: PMC9924068 DOI: 10.3389/fnbeh.2022.1072571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/29/2022] [Indexed: 02/03/2023] Open
Abstract
Subpopulations of neurons display increased activity during memory encoding and manipulating the activity of these neurons can induce artificial formation or erasure of memories. Thus, these neurons are thought to be cellular engrams. Moreover, correlated activity between pre- and postsynaptic engram neurons is thought to lead to strengthening of their synaptic connections, thus increasing the probability of neural activity patterns occurring during encoding to reoccur at recall. Therefore, synapses between engram neurons can also be considered as a substrate of memory, or a synaptic engram. One can label synaptic engrams by targeting two complementary, non-fluorescent, synapse-targeted GFP fragments separately to the pre- and postsynaptic compartment of engram neurons; the two GFP fragments reconstitute a fluorescent GFP at the synaptic cleft between the engram neurons, thereby highlighting synaptic engrams. In this work we explored a transsynaptic GFP reconstitution system (mGRASP) to label synaptic engrams between hippocampal CA1 and CA3 engram neurons identified by different Immediate-Early Genes: cFos and Arc. We characterized the expression of the cellular and synaptic labels of the mGRASP system upon exposure to a novel environment or learning of a hippocampal-dependent memory task. We found that mGRASP under the control of transgenic ArcCreERT2 labeled synaptic engrams more efficiently than when controlled by viral cFostTA, possibly due to differences in the genetic systems rather than the specific IEG promoters.
Collapse
Affiliation(s)
- B. K. B. Murthy
- Leibniz Institute for Neurobiology, Magdeburg, Germany,Graduate School of Systemic Neurosciences, Munich, Germany,Max Planck Institute of Psychiatry, Munich, Germany
| | - S. Somatakis
- Max Planck Institute of Psychiatry, Munich, Germany
| | - A. F. Ulivi
- Leibniz Institute for Neurobiology, Magdeburg, Germany,Max Planck Institute of Psychiatry, Munich, Germany
| | - H. Klimmt
- Leibniz Institute for Neurobiology, Magdeburg, Germany,Max Planck Institute of Psychiatry, Munich, Germany,International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | | | - N. Haynes
- Max Planck Institute of Psychiatry, Munich, Germany
| | - R. E. Huettl
- Max Planck Institute of Psychiatry, Munich, Germany
| | - A. Chen
- Graduate School of Systemic Neurosciences, Munich, Germany,Max Planck Institute of Psychiatry, Munich, Germany,International Max Planck Research School for Translational Psychiatry, Munich, Germany,Weizmann Institute of Science, Rehovot, Israel
| | - Alessio Attardo
- Leibniz Institute for Neurobiology, Magdeburg, Germany,Graduate School of Systemic Neurosciences, Munich, Germany,Max Planck Institute of Psychiatry, Munich, Germany,International Max Planck Research School for Translational Psychiatry, Munich, Germany,*Correspondence: Alessio Attardo,
| |
Collapse
|
26
|
Persistent increase of accumbens cocaine ensemble excitability induced by IRK downregulation after withdrawal mediates the incubation of cocaine craving. Mol Psychiatry 2023; 28:448-462. [PMID: 36481931 PMCID: PMC9812793 DOI: 10.1038/s41380-022-01884-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 10/30/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022]
Abstract
The incubation phenomenon, cue-induced drug craving progressively increasing over prolonged withdrawal, accounts for persistent relapse, leading to a dilemma in the treatment of cocaine addiction. The role of neuronal ensembles activated by initial cocaine experience in the incubation phenomenon was unclear. In this study, with cocaine self-administration (SA) models, we found that neuronal ensembles in the nucleus accumbens shell (NAcSh) showed increasing activation induced by cue-induced drug-seeking after 30-day withdrawal. Inhibition or activation of NAcSh cocaine-ensembles suppressed or promoted craving for cocaine, demonstrating a critical role of NAcSh cocaine-ensembles in incubation for cocaine craving. NAcSh cocaine-ensembles showed a specific increase of membrane excitability and a decrease of inward rectifying channels Kir2.1 currents after 30-day withdrawal. Overexpression of Kir2.1 in NAcSh cocaine-ensembles restored neuronal membrane excitability and suppressed cue-induced drug-seeking after 30-day withdrawal. Expression of dominant-negative Kir2.1 in NAcSh cocaine-ensembles enhanced neuronal membrane excitability and accelerated incubation of cocaine craving. Our results provide a cellular mechanism that the downregulation of Kir2.1 functions in NAcSh cocaine-ensembles induced by prolonged withdrawal mediates the enhancement of ensemble membrane excitability, leading to incubation of cocaine craving.
Collapse
|
27
|
Park S, Jung JH, Karimi SA, Jacob AD, Josselyn SA. Opto-extinction of a threat memory in mice. Brain Res Bull 2022; 191:61-68. [DOI: 10.1016/j.brainresbull.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/02/2022]
|
28
|
Bechay KR, Abduljawad N, Latifi S, Suzuki K, Iwashita H, Carmichael ST. PDE2A Inhibition Enhances Axonal Sprouting, Functional Connectivity, and Recovery after Stroke. J Neurosci 2022; 42:8225-8236. [PMID: 36163142 PMCID: PMC9653274 DOI: 10.1523/jneurosci.0730-22.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 11/21/2022] Open
Abstract
Phosphodiesterase (PDE) inhibitors have been safely and effectively used in the clinic and increase the concentration of intracellular cyclic nucleotides (cAMP/cGMP). These molecules activate downstream mediators, including the cAMP response element-binding protein (CREB), which controls neuronal excitability and growth responses. CREB gain of function enhances learning and allocates neurons into memory engrams. CREB also controls recovery after stroke. PDE inhibitors are linked to recovery from neural damage and to stroke recovery in specific sites within the brain. PDE2A is enriched in cortex. In the present study, we use a mouse cortical stroke model in young adult and aged male mice to test the effect of PDE2A inhibition on functional recovery, and on downstream mechanisms of axonal sprouting, tissue repair, and the functional connectivity of neurons in recovering cortex. Stroke causes deficits in use of the contralateral forelimb, loss of axonal projections in cortex adjacent to the infarct, and functional disconnection of neuronal networks. PDE2A inhibition enhances functional recovery, increases axonal projections in peri-infarct cortex, and, through two-photon in vivo imaging, enhances the functional connectivity of motor system excitatory neurons. PDE2A inhibition after stroke does not have an effect on other aspects of tissue repair, such as angiogenesis, gliogenesis, neurogenesis, and inflammatory responses. These data suggest that PDE2A inhibition is an effective therapeutic approach for stroke recovery in the rodent and that it simultaneously enhances connectivity in peri-infarct neuronal populations.SIGNIFICANCE STATEMENT Inhibition of PDE2A enhances motor recovery, axonal projections, and functional connectivity of neurons in peri-infarct tissue. This represents an avenue for a pharmacological therapy for stroke recovery.
Collapse
Affiliation(s)
- Kirollos Raouf Bechay
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Nora Abduljawad
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Shahrzad Latifi
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Kazunori Suzuki
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa 251-8555, Japan
| | - Hiroki Iwashita
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa 251-8555, Japan
| | - S Thomas Carmichael
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
29
|
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
|
30
|
Anderson EM, Taniguchi M. Epigenetic Effects of Addictive Drugs in the Nucleus Accumbens. Front Mol Neurosci 2022; 15:828055. [PMID: 35813068 PMCID: PMC9260254 DOI: 10.3389/fnmol.2022.828055] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
Substance use induces long-lasting behavioral changes and drug craving. Increasing evidence suggests that epigenetic gene regulation contributes to the development and expression of these long-lasting behavioral alterations. Here we systematically review extensive evidence from rodent models of drug-induced changes in epigenetic regulation and epigenetic regulator proteins. We focus on histone acetylation and histone methylation in a brain region important for drug-related behaviors: the nucleus accumbens. We also discuss how experimentally altering these epigenetic regulators via systemically administered compounds or nucleus accumbens-specific manipulations demonstrate the importance of these proteins in the behavioral effects of drugs and suggest potential therapeutic value to treat people with substance use disorder. Finally, we discuss limitations and future directions for the field of epigenetic studies in the behavioral effects of addictive drugs and suggest how to use these insights to develop efficacious treatments.
Collapse
|
31
|
Dissociated Role of Thalamic and Cortical Input to the Lateral Amygdala for Consolidation of Long-Term Fear Memory. J Neurosci 2021; 41:9561-9570. [PMID: 34667069 DOI: 10.1523/jneurosci.1167-21.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/25/2022] Open
Abstract
Post-encoding coordinated reactivation of memory traces distributed throughout interconnected brain regions is thought to be critical for consolidation of memories. However, little is known about the role of neural circuit pathways during post-learning periods for consolidation of memories. To investigate this question, we optogenetically silenced the inputs from both auditory cortex and thalamus in the lateral amygdala (LA) for 15 min immediately following auditory fear conditioning (FC) and examined its effect on fear memory formation in mice of both sexes. Optogenetic inhibition of both inputs disrupted long-term fear memory formation tested 24 h after FC. This effect was specific such that the same inhibition did not affect short-term memory and context-dependent memory. Moreover, long-term memory was intact if the inputs were inhibited at much later time points after FC (3 h or 1 d after FC), indicating that optical inhibition for 15 min itself does not produce any nonspecific deleterious effect on fear memory retrieval. Selective inhibition of thalamic input was sufficient to impair consolidation of auditory fear memory. In contrast, selective inhibition of cortical input disrupted remote fear memory without affecting recent memory. These results reveal a dissociated role of thalamic and cortical input to the LA during early post-learning periods for consolidation of long-term fear memory.SIGNIFICANCE STATEMENT Coordinated communications between brain regions are thought to be essential during post-learning periods for consolidation of memories. However, the role of specific neural circuit pathways in this process has been scarcely explored. Using a precise optogenetic inhibition of auditory input pathways, either thalamic or cortical or both, to the LA during post-training periods, we here show that thalamic input is required for consolidation of both recent and remote fear memory, whereas cortical input is crucial for consolidation of remote fear memory. These results reveal a dissociated role of auditory input pathways to the LA for consolidation of long-term fear memory.
Collapse
|
32
|
4R Tau Modulates Cocaine-Associated Memory through Adult Dorsal Hippocampal Neurogenesis. J Neurosci 2021; 41:6753-6774. [PMID: 34099513 DOI: 10.1523/jneurosci.2848-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/28/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
The development, persistence and relapse of drug addiction require drug memory that generally develops with drug administration-paired contextual stimuli. Adult hippocampal neurogenesis (AHN) contributes to cocaine memory formation; however, the underlying mechanism remains unclear. Male mice hippocampal expression of Tau was significantly decreased during the cocaine-associated memory formation. Genetic overexpression of four microtubule-binding repeats Tau (4R Tau) in the mice hippocampus disrupted cocaine memory by suppressing AHN. Furthermore, 4R Tau directly interacted with phosphoinositide 3-kinase (PI3K)-p85 and impaired its nuclear translocation and PI3K-AKT signaling, processes required for hippocampal neuron proliferation. Collectively, 4R Tau modulates cocaine memory formation by disrupting AHN, suggesting a novel mechanism underlying cocaine memory formation and provide a new strategy for the treatment of cocaine addiction.SIGNIFICANCE STATEMENT Drug memory that generally develops with drug-paired contextual stimuli and drug administration is critical for the development, persistence and relapse of drug addiction. Previous studies have suggested that adult hippocampal neurogenesis (AHN) plays a role in cocaine memory formation. Here, we showed that Tau was significantly downregulated in the hippocampus in the cocaine memory formation. Tau knock-out (KO) promoted AHN in the hippocampal dentate gyrus (DG), resulting in the enhanced memory formation evoked by cocaine-cue stimuli. In contrast, genetically overexpressed 4R Tau in the hippocampus disrupted cocaine-cue memory by suppressing AHN. In addition, 4R Tau interacted directly with phosphoinositide 3-kinase (PI3K)-p85 and hindered its nuclear translocation, eventually repressing PI3K-AKT signaling, which is essential for hippocampal neuronal proliferation.
Collapse
|
33
|
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
|
34
|
Neuronal ensembles in memory processes. Semin Cell Dev Biol 2021; 125:136-143. [PMID: 33858772 DOI: 10.1016/j.semcdb.2021.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/19/2022]
Abstract
A neuronal ensemble represents the concomitant activity of a specific group of neurons that could encompass a broad repertoire of brain functions such as motor, perceptual, memory or cognitive states. On the other hand, a memory engram portrays the physical manifestation of memory or the changes that enable learning and retrieval. Engram studies focused for many years on finding where memories are stored as in, which cells or brain regions represent a memory trace, and disregarded the investigation of how neuronal activity patterns give rise to such memories. Recent experiments suggest that the association and reactivation of specific neuronal groups could be the main mechanism underlying the brain's ability to remember past experiences and envision future actions. Thus, the growing consensus is that the interaction between neuronal ensembles could allow sequential activity patterns to become memories and recurrent memories to compose complex behaviors. The goal of this review is to propose how the neuronal ensemble framework could be translated and useful to understand memory processes.
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
|
Salery M, Godino A, Nestler EJ. Drug-activated cells: From immediate early genes to neuronal ensembles in addiction. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 90:173-216. [PMID: 33706932 DOI: 10.1016/bs.apha.2020.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Beyond their rapid rewarding effects, drugs of abuse can durably alter an individual's response to their environment as illustrated by the compulsive drug seeking and risk of relapse triggered by drug-associated stimuli. The persistence of these associations even long after cessation of drug use demonstrates the enduring mark left by drugs on brain reward circuits. However, within these circuits, neuronal populations are differently affected by drug exposure and growing evidence indicates that relatively small subsets of neurons might be involved in the encoding and expression of drug-mediated associations. The identification of sparse neuronal populations recruited in response to drug exposure has benefited greatly from the study of immediate early genes (IEGs) whose induction is critical in initiating plasticity programs in recently activated neurons. In particular, the development of technologies to manipulate IEG-expressing cells has been fundamental to implicate broadly distributed neuronal ensembles coincidently activated by either drugs or drug-associated stimuli and to then causally establish their involvement in drug responses. In this review, we summarize the literature regarding IEG regulation in different learning paradigms and addiction models to highlight their role as a marker of activity and plasticity. As the exploration of neuronal ensembles in addiction improves our understanding of drug-associated memory encoding, it also raises several questions regarding the cellular and molecular characteristics of these discrete neuronal populations as they become incorporated in drug-associated neuronal ensembles. We review recent efforts towards this goal and discuss how they will offer a more comprehensive understanding of addiction pathophysiology.
Collapse
Affiliation(s)
- Marine Salery
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Arthur Godino
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
37
|
Koob GF. A Basolateral Amygdala Microcircuit for Drug Craving: Is There a Craving Engram? Biol Psychiatry 2021; 89:323-325. [PMID: 33478679 DOI: 10.1016/j.biopsych.2020.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022]
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism and the National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
38
|
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: 28] [Impact Index Per Article: 5.6] [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
|
39
|
Savell KE, Tuscher JJ, Zipperly ME, Duke CG, Phillips RA, Bauman AJ, Thukral S, Sultan FA, Goska NA, Ianov L, Day JJ. A dopamine-induced gene expression signature regulates neuronal function and cocaine response. SCIENCE ADVANCES 2020; 6:eaba4221. [PMID: 32637607 PMCID: PMC7314536 DOI: 10.1126/sciadv.aba4221] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/08/2020] [Indexed: 05/21/2023]
Abstract
Drugs of abuse elevate dopamine levels in the nucleus accumbens (NAc) and alter transcriptional programs believed to promote long-lasting synaptic and behavioral adaptations. Here, we leveraged single-nucleus RNA-sequencing to generate a comprehensive molecular atlas of cell subtypes in the NAc, defining both sex-specific and cell type-specific responses to acute cocaine experience in a rat model system. Using this transcriptional map, we identified an immediate early gene expression program that is up-regulated following cocaine experience in vivo and dopamine receptor activation in vitro. Multiplexed induction of this gene program with a large-scale CRISPR-dCas9 activation strategy initiated a secondary synapse-centric transcriptional profile, altered striatal physiology in vitro, and enhanced cocaine sensitization in vivo. Together, these results define the transcriptional response to cocaine with cellular precision and demonstrate that drug-responsive gene programs can potentiate both physiological and behavioral adaptations to drugs of abuse.
Collapse
Affiliation(s)
- Katherine E. Savell
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G. Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert A. Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Allison J. Bauman
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Saakshi Thukral
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Faraz A. Sultan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nicholas A. Goska
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
40
|
A time-dependent role for the transcription factor CREB in neuronal allocation to an engram underlying a fear memory revealed using a novel in vivo optogenetic tool to modulate CREB function. Neuropsychopharmacology 2020; 45:916-924. [PMID: 31837649 PMCID: PMC7162924 DOI: 10.1038/s41386-019-0588-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/08/2019] [Accepted: 12/04/2019] [Indexed: 12/21/2022]
Abstract
The internal representation of an experience is thought to be encoded by long-lasting physical changes to the brain ("engrams") . Previously, we and others showed within the lateral amygdala (LA), a region critical for auditory conditioned fear, eligible neurons compete against one other for allocation to an engram. Neurons with relatively higher function of the transcription factor CREB were more likely to be allocated to the engram. In these studies, though, CREB function was artificially increased for several days before training. Precisely when increased CREB function is important for allocation remains an unanswered question. Here, we took advantage of a novel optogenetic tool (opto-DN-CREB) to gain spatial and temporal control of CREB function in freely behaving mice. We found increasing CREB function in a small, random population of LA principal neurons in the minutes, but not 24 h, before training was sufficient to enhance memory, likely because these neurons were preferentially allocated to the underlying engram. However, similarly increasing CREB activity in a small population of random LA neurons immediately after training disrupted subsequent memory retrieval, likely by disrupting the precise spatial and temporal patterns of offline post-training neuronal activity and/or function required for consolidation. These findings reveal the importance of the timing of CREB activity in regulating allocation and subsequent memory retrieval, and further, highlight the potential of optogenetic approaches to control protein function with temporal specificity in behaving animals.
Collapse
|
41
|
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: 543] [Impact Index Per Article: 108.6] [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
|
42
|
Acute restraint stress augments the rewarding memory of cocaine through activation of α1 adrenoceptors in the medial prefrontal cortex of mice. Neuropharmacology 2020; 166:107968. [DOI: 10.1016/j.neuropharm.2020.107968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/11/2020] [Accepted: 01/15/2020] [Indexed: 01/17/2023]
|
43
|
Aqrabawi AJ, Kim JC. Olfactory memory representations are stored in the anterior olfactory nucleus. Nat Commun 2020; 11:1246. [PMID: 32144256 PMCID: PMC7060254 DOI: 10.1038/s41467-020-15032-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 02/15/2020] [Indexed: 11/26/2022] Open
Abstract
The anterior olfactory nucleus (AON) is the initial recipient of odour information from the olfactory bulb, and the target of dense innervation conveying spatiotemporal cues from the hippocampus. We hypothesized that the AON detects the coincidence of these inputs, generating patterns of activity reflective of episodic odour engrams. Using activity-dependent tagging combined with neural manipulation techniques, we reveal that contextually-relevant odour engrams are stored within the AON and that their activity is necessary and sufficient for the behavioural expression of odour memory. Our findings offer a new model for studying the mechanisms underlying memory representations. Odours are powerful stimuli used by most organisms to guide behaviour. Here, the authors identify populations of neurons within the anterior olfactory nucleus (AON) which are necessary and sufficient for the behavioural expression of odour memory.
Collapse
Affiliation(s)
- Afif J Aqrabawi
- Department of Cell and Systems Biology, University of Toronto, Toronto, M5S 3G5, Canada. .,Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Jun Chul Kim
- Department of Cell and Systems Biology, University of Toronto, Toronto, M5S 3G5, Canada. .,Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada.
| |
Collapse
|
44
|
Rich MT, Huang YH, Torregrossa MM. Plasticity at Thalamo-amygdala Synapses Regulates Cocaine-Cue Memory Formation and Extinction. Cell Rep 2020; 26:1010-1020.e5. [PMID: 30673597 DOI: 10.1016/j.celrep.2018.12.105] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/27/2018] [Accepted: 12/26/2018] [Indexed: 01/02/2023] Open
Abstract
Repeated drug use has long-lasting effects on plasticity throughout the brain's reward and memory systems. Environmental cues that are associated with drugs of abuse can elicit craving and relapse, but the neural circuits responsible for driving drug-cue-related behaviors have not been well delineated, creating a hurdle for the development of effective relapse prevention therapies. In this study, we used a cocaine+cue self-administration paradigm followed by cue re-exposure to establish that the strength of the drug cue association corresponds to the strength of synapses between the medial geniculate nucleus (MGN) of the thalamus and the lateral amygdala (LA). Furthermore, we demonstrate, via optogenetically induced LTD of MGN-LA synapses, that reversing cocaine-induced potentiation of this pathway is sufficient to inhibit cue-induced relapse-like behavior.
Collapse
Affiliation(s)
- Matthew T Rich
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mary M Torregrossa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
45
|
Rich MT, Huang YH, Torregrossa MM. Calcineurin Promotes Neuroplastic Changes in the Amygdala Associated with Weakened Cocaine-Cue Memories. J Neurosci 2020; 40:1344-1354. [PMID: 31862855 PMCID: PMC7002139 DOI: 10.1523/jneurosci.0453-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 11/23/2019] [Accepted: 12/10/2019] [Indexed: 01/03/2023] Open
Abstract
Interfering with memory reconsolidation or inducing memory extinction are two approaches for weakening maladaptive memories in disorders such as addiction and post-traumatic stress disorder. Both extinction and reconsolidation are regulated by intracellular protein kinases and phosphatases, and interfering with these signaling molecules can alter memory strength. The calcium-dependent protein phosphatase, calcineurin (CaN), has been implicated in both the consolidation and extinction of fear memories. However, the role of CaN in regulating drug-cue associative memories has not been investigated. Prior studies have demonstrated that plasticity at thalamo-lateral amygdala (T-LA) synapses is critically involved in the regulation of cocaine-cue memories. Therefore, in the present study, we tested the effects of LA administration of an activator of CaN, chlorogenic acid (CGA), on behavioral and electrophysiological indices of cocaine cue memory reconsolidation and extinction. Male, Sprague-Dawley rats were trained to self-administer cocaine paired with an audiovisual cue. The cue memory was then either briefly reactivated, extinguished, or not manipulated, followed immediately by LA infusion of CGA. Rats were tested 24 h later for cue-induced reinstatement, or LA slices were prepared for electrophysiological recordings. We found that intra-LA infusions of CGA following cue extinction or reconsolidation reduced cue-induced reinstatement, which was blocked by co-infusion of the CaN inhibitor, FK-506. Similarly, CGA infusions following cue re-exposure significantly attenuated EPSC amplitude at T-LA synapses, suggesting that CaN affects cocaine-cue memory reconsolidation and extinction by altering T-LA synaptic strength. Therefore, CaN signaling in the LA may represent a novel target for disrupting cocaine-associated memories to reduce relapse.SIGNIFICANCE STATEMENT Repetitive drug use induces synaptic plasticity that underlies the formation of long-lasting associative memories for environmental cues paired with the drug. We previously identified thalamo-amygdala synapses (T-LA) that project via the interal capsule, as an important locus for the regulation of cocaine-cue memories. These synapses are strengthened by repeated cocaine-cue pairings, but this is reversed by extinction training or by optogenetic induction of in vivo long-term depression (LTD). Here, we demonstrate that activating calcineurin, a calcium-dependent phosphatase, following the reactivation or extinction of a cocaine-cue memory, induces LTD-like changes at T-LA synapses, and a corresponding decrease in cue-induced reinstatement, suggesting that calcineurin may be a potential therapeutic target for relapse prevention.
Collapse
Affiliation(s)
- Matthew T Rich
- Department of Psychiatry
- Center for Neuroscience, and
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | | | | |
Collapse
|
46
|
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
|
47
|
Armio RL, Laurikainen H, Ilonen T, Walta M, Salokangas RKR, Koutsouleris N, Hietala J, Tuominen L. Amygdala subnucleus volumes in psychosis high-risk state and first-episode psychosis. Schizophr Res 2020; 215:284-292. [PMID: 31744752 DOI: 10.1016/j.schres.2019.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/17/2019] [Accepted: 10/08/2019] [Indexed: 12/13/2022]
Abstract
Structural and functional abnormalities of the amygdala in schizophrenia have been well documented. Post-mortem studies suggest that the lateral nucleus is particularly affected in schizophrenia. It is not known whether the amygdala subnuclei are differently affected at the time of the first-episode psychosis or already at high-risk state. 75 first-episode psychosis patients (FEP), 45 clinical high-risk patients (CHR) and 76 population controls participated in this cross-sectional case-control study. Participants underwent T1-weighted 3T MRI scans, from which the amygdala was segmented using a newly developed automated algorithm. Because early adverse events increase risk for psychosis and affect the amygdala, we also tested whether experiences of childhood maltreatment associate with the putative amygdala subnuclei abnormalities. Compared to the population controls, FEP had smaller volumes of the lateral, and basal nuclei. In CHR, only the lateral nucleus was significantly smaller compared to the control subjects. Experience of childhood maltreatment was inversely associated with lateral nucleus volumes in FEP but not in CHR. These results show that the lateral and basal nuclei of the amygdala are already affected in FEP. These volumetric changes may reflect specific cellular abnormalities that have been observed in post-mortem studies in schizophrenia in the same subnuclei. Decreased volume of the lateral nucleus in CHR suggest that a smaller lateral nucleus could serve as a potential biomarker for psychosis risk. Finally, we found that the lateral nucleus volumes in FEP may be sensitive to the effects of childhood maltreatment.
Collapse
Affiliation(s)
- Reetta-Liina Armio
- PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland; Department of Psychiatry, University of Turku, Kunnallissairaalantie 20, building 9, 20700, Turku, Finland; Department of Psychiatry, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Heikki Laurikainen
- PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland; Department of Psychiatry, University of Turku, Kunnallissairaalantie 20, building 9, 20700, Turku, Finland; Department of Psychiatry, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Tuula Ilonen
- Department of Psychiatry, University of Turku, Kunnallissairaalantie 20, building 9, 20700, Turku, Finland
| | - Maija Walta
- Department of Psychiatry, University of Turku, Kunnallissairaalantie 20, building 9, 20700, Turku, Finland; Department of Psychiatry, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Raimo K R Salokangas
- Department of Psychiatry, University of Turku, Kunnallissairaalantie 20, building 9, 20700, Turku, Finland
| | - Nikolaos Koutsouleris
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian University, Nussbaumstr. 7, D-80336, Munich, Germany
| | - Jarmo Hietala
- PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland; Department of Psychiatry, University of Turku, Kunnallissairaalantie 20, building 9, 20700, Turku, Finland; Department of Psychiatry, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland
| | - Lauri Tuominen
- PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland; Department of Psychiatry, University of Turku, Kunnallissairaalantie 20, building 9, 20700, Turku, Finland; University of Ottawa Institute of Mental Health Research, Ottawa, ON, K1Z 8N3, Canada
| |
Collapse
|
48
|
Zhang T, Yanagida J, Kamii H, Wada S, Domoto M, Sasase H, Deyama S, Takarada T, Hinoi E, Sakimura K, Yamanaka A, Maejima T, Mieda M, Sakurai T, Nishitani N, Nagayasu K, Kaneko S, Minami M, Kaneda K. Glutamatergic neurons in the medial prefrontal cortex mediate the formation and retrieval of cocaine-associated memories in mice. Addict Biol 2020; 25:e12723. [PMID: 30734456 DOI: 10.1111/adb.12723] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 12/04/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
In drug addiction, environmental stimuli previously associated with cocaine use readily elicit cocaine-associated memories, which persist long after abstinence and trigger cocaine craving and consumption. Although previous studies suggest that the medial prefrontal cortex (mPFC) is involved in the expression of cocaine-addictive behaviors, it remains unclear whether excitatory and inhibitory neurons in the mPFC are causally related to the formation and retrieval of cocaine-associated memories. To address this issue, we used the designer receptors exclusively activated by designer drugs (DREADD) technology combined with a cocaine-induced conditioned place preference (CPP) paradigm. We suppressed mPFC neuronal activity in a cell-type- and timing-dependent manner. C57BL/6J wild-type mice received bilateral intra-mPFC infusion of an adeno-associated virus (AAV) expressing inhibitory DREADD (hM4Di) under the control of CaMKII promotor to selectively suppress mPFC pyramidal neurons. GAD67-Cre mice received bilateral intra-mPFC infusion of a Cre-dependent AAV expressing hM4Di to specifically silence GABAergic neurons. Chemogenetic suppression of mPFC pyramidal neurons significantly attenuated both the acquisition and expression of cocaine CPP, while suppression of mPFC GABAergic neurons affected neither the acquisition nor expression of cocaine CPP. Moreover, chemogenetic inhibition of mPFC glutamatergic neurons did not affect the acquisition and expression of lithium chloride-induced conditioned place aversion. These results suggest that the activation of glutamatergic, but not GABAergic, neurons in the mPFC mediates both the formation and retrieval of cocaine-associated memories.
Collapse
Affiliation(s)
- Tong Zhang
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| | - Junko Yanagida
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| | - Hironori Kamii
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
- Department of Pharmacology, Graduate School of Pharmaceutical SciencesHokkaido University Sapporo Japan
| | - Shintaro Wada
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| | - Masaki Domoto
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| | - Hitoki Sasase
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| | - Takeshi Takarada
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
- Department of Regenerative ScienceOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama Japan
| | - Eiichi Hinoi
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research InstituteNiigata University Niigata Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental MedicineNagoya University Nagoya Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical SciencesKanazawa University Kanazawa Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical SciencesKanazawa University Kanazawa Japan
| | - Takeshi Sakurai
- Department of Integrative Neurophysiology, Graduate School of Medical SciencesKanazawa University Kanazawa Japan
- International Institute for Integrative Sleep MedicineUniversity of Tsukuba Tsukuba Japan
| | - Naoya Nishitani
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical SciencesKyoto University Kyoto Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical SciencesKyoto University Kyoto Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical SciencesKyoto University Kyoto Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical SciencesHokkaido University Sapporo Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health SciencesKanazawa University Kanazawa Japan
| |
Collapse
|
49
|
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: 110] [Impact Index Per Article: 18.3] [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
|
50
|
Matos MR, Visser E, Kramvis I, van der Loo RJ, Gebuis T, Zalm R, Rao-Ruiz P, Mansvelder HD, Smit AB, van den Oever MC. Memory strength gates the involvement of a CREB-dependent cortical fear engram in remote memory. Nat Commun 2019; 10:2315. [PMID: 31127098 PMCID: PMC6534583 DOI: 10.1038/s41467-019-10266-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/29/2019] [Indexed: 12/22/2022] Open
Abstract
Encoding and retrieval of contextual memories is initially mediated by sparsely activated neurons, so-called engram cells, in the hippocampus. Subsequent memory persistence is thought to depend on network-wide changes involving progressive contribution of cortical regions, a process referred to as systems consolidation. Using a viral-based TRAP (targeted recombination in activated populations) approach, we studied whether consolidation of contextual fear memory by neurons in the medial prefrontal cortex (mPFC) is modulated by memory strength and CREB function. We demonstrate that activity of a small subset of mPFC neurons is sufficient and necessary for remote memory expression, but their involvement depends on the strength of conditioning. Furthermore, selective disruption of CREB function in mPFC engram cells after mild conditioning impairs remote memory expression. Together, our data demonstrate that memory consolidation by mPFC engram cells requires CREB-mediated transcription, with the functionality of this network hub being gated by memory strength. Little is known about mechanisms that regulate the involvement of cortical engram cells in remote memory. Here, authors demonstrate that memory consolidation by mPFC engram cells requires CREB-mediated transcription, with the functionality of this network hub being gated by memory strength.
Collapse
Affiliation(s)
- Mariana R Matos
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Esther Visser
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Ioannis Kramvis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Rolinka J van der Loo
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Titia Gebuis
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Robbert Zalm
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|