1
|
Su X, He J, Liu Y, Liu K, Lei B, Zhong Y. Protocol for labeling and manipulating "home"-activated neurons in mice. STAR Protoc 2025; 6:103704. [PMID: 40121664 DOI: 10.1016/j.xpro.2025.103704] [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: 12/09/2024] [Revised: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Home-related cues, such as home bedding, influence affective behaviors in mice, including fear relief, anxiety reduction, and defensive aggression. Here, we present a protocol for labeling and manipulating "home"-activated neurons in the lateral hypothalamus (LH) using a Tet-Off inducible system. We describe steps for virus injection, postoperative recovery, and combining c-Fos-driven activity labeling with chemogenetics. This protocol integrates neuronal targeting with behavioral assays to provide insights into the functional role of "home"-activated neurons in home-driven behavioral adaptations. For complete details on the use and execution of this protocol, please refer to Su et al.1.
Collapse
Affiliation(s)
- Xiaoya Su
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China; School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Junyue He
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; McGovern Institute of Brain Research, Beijing 100084, P.R. China; Peking University, Tsinghua University, National Institute Biological Science Joint Graduate Program, Beijing, P.R. China
| | - Yunlong Liu
- Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Keyu Liu
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Bo Lei
- Beijing Academy of Artificial Intelligence, Beijing 100084, P.R. China.
| | - Yi Zhong
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China; School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; McGovern Institute of Brain Research, Beijing 100084, P.R. China; MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, P.R. China.
| |
Collapse
|
2
|
Franceschini A, Jin M, Chen CW, Silvestri L, Mastrodonato A, Denny CA. Brain-wide immunolabeling and tissue clearing applications for engram research. Neurobiol Learn Mem 2025; 218:108032. [PMID: 39922482 DOI: 10.1016/j.nlm.2025.108032] [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: 10/28/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
In recent years, there has been significant progress in memory research, driven by genetic and imaging technological advances that have given unprecedented access to individual memory traces or engrams. Although Karl Lashley argued since the 1930s that an engram is not confined to a particular area but rather distributed across the entire brain, most current studies have focused exclusively on a single or few brain regions. However, this compartmentalized approach overlooks the interactions between multiple brain regions, limiting our understanding of engram mechanisms. More recently, several studies have begun to investigate engrams across the brain, but research is still limited by a lack of standardized techniques capable of reconstructing multiple ensembles at single-cell resolution across the entire brain. In this review, we guide researchers through the latest technological advancements and discoveries in immediate early gene (IEG) techniques, tissue clearing methods, microscope modalities, and automated large-scale analysis. These innovations could propel the field forward in building brain-wide engram maps of normal and disease states, thus, providing unprecedented new insights. Ultimately, this review aims to bridge the gap between research focused on single brain regions and the need for a comprehensive understanding of whole-brain engrams, revealing new approaches for exploring the neuronal mechanisms underlying engrams.
Collapse
Affiliation(s)
- Alessandra Franceschini
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, 50019 Italy
| | - Michelle Jin
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY 10027, USA
| | - Claire W Chen
- Cellular, Molecular, and Biomedical Sciences Graduate Program, Columbia University, New York, NY 10027, USA
| | - Ludovico Silvestri
- European Laboratory for Non-linear Spectroscopy (LENS), Sesto Fiorentino, 50019 Italy; Department of Physics and Astronomy, University of Florence, Sesto Fiorentino 50019, Italy
| | - Alessia Mastrodonato
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY 10032, USA.
| | - Christine Ann Denny
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY 10032, USA; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY 10032, USA.
| |
Collapse
|
3
|
Nemat P, Semenova S, van der Loo RJ, Smit AB, Spijker S, van den Oever MC, Rao-Ruiz P. Structural synaptic signatures of contextual memory retrieval-reactivated hippocampal engram cells. Neurobiol Learn Mem 2025; 218:108033. [PMID: 39923960 DOI: 10.1016/j.nlm.2025.108033] [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: 09/29/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
Learning enhances hippocampal engram cell synaptic connectivity which is crucial for engram reactivation and recall to natural cues. Memory retrieval engages only a subset of the learning-activated ensemble, indicating potential differences in synaptic connectivity signatures of reactivated and non-reactivated cells. We probed these differences in structural synaptic connectivity patterns after recent memory retrieval, 72 h after either neutral Context Exploration (CE) or aversive Contextual Fear Conditioning (CFC). Using a combination of eGRASP (enhanced green fluorescent protein (GFP) reconstitution across synaptic partners) and viral-TRAP (targeted recombination in activated populations) to label CA3 synapses onto CA1 engram cells, we investigated differences in spine density, clusters, and morphology between the reactivated and non-reactivated population of the learning ensemble. In doing so, we developed a pipeline for reconstruction and analysis of dendrites and spines, taking nested data structure into account. Our data demonstrate an interplay between reactivation status, context valence or both factors on the number, distribution, and morphology of CA1 engram cell synapses. Despite a lack of differences in spine density, reactivated engram cells encoding an aversive context were characterised by a higher probability of forming spine clusters and a more dynamic spine type signature compared to their non-reactivated counterparts or engram cells encoding a neutral context. Together, our data indicate that the learning-activated ensemble undergoes different trajectories in structural synaptic connectivity during engram refinement.
Collapse
Affiliation(s)
- Panthea Nemat
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Salimat Semenova
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit 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, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Sabine Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit 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, The Netherlands.
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Parkins S, Song Y, Jaoui Y, Gala A, Konda KT, Richardson C, Lee HK. Spatial Mapping of Activity Changes across Sensory Areas Following Visual Deprivation in Adults. J Neurosci 2025; 45:e0969242024. [PMID: 39592237 PMCID: PMC11756622 DOI: 10.1523/jneurosci.0969-24.2024] [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: 05/21/2024] [Revised: 10/22/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Loss of a sensory modality triggers global adaptation across brain areas, allowing the remaining senses to guide behavior more effectively. There are specific synaptic and circuit plasticity observed across many sensory areas, which suggests potential widespread changes in activity. Here we used a cFosTRAP2 mouse line to drive tdTomato (tdT) expression in active cells to spatially map the extent of activity changes in various sensory areas in adult mice of both sexes following two modes of visual deprivation. We found that in the primary visual cortex (V1), both dark exposure (DE) and enucleation (EN) caused an initial loss of active cells followed by a partial rebound, which occurred relatively more in the superficial layers. A similar pattern was observed in the secondary visual cortex, especially in the lateral areas (V2L). The spared primary sensory cortices adapted distinctly. In the primary somatosensory barrel cortex (S1BF), there was a change in the density of active cells dependent on the duration and the mode of visual deprivation. In the primary auditory cortex (A1), there was a relative reduction in the density of active cells in the superficial layers without a significant change in the overall density. There were minimal changes in the active cell density in the secondary cortices of the spared senses and the multisensory retrosplenial cortex (RSP). Our results are consistent with cross-modal recruitment of the deprived visual cortex and compensatory plasticity in the spared primary sensory cortices that can support enhanced processing and refinement of the spared senses.
Collapse
Affiliation(s)
- Samuel Parkins
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Cell Molecular Developmental Biology and Biophysics Graduate Program, Johns Hopkins University, Baltimore, Maryland 21218
| | - Yidong Song
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Yanis Jaoui
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Aryan Gala
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Kaven T Konda
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Cell Molecular Developmental Biology and Biophysics Graduate Program, Johns Hopkins University, Baltimore, Maryland 21218
| | - Crispo Richardson
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Hey-Kyoung Lee
- Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
- Cell Molecular Developmental Biology and Biophysics Graduate Program, Johns Hopkins University, Baltimore, Maryland 21218
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland 21218
| |
Collapse
|
5
|
Tregub PP, Komleva YK, Kukla MV, Averchuk AS, Vetchinova AS, Rozanova NA, Illarioshkin SN, Salmina AB. Brain Plasticity and Cell Competition: Immediate Early Genes Are the Focus. Cells 2025; 14:143. [PMID: 39851571 PMCID: PMC11763428 DOI: 10.3390/cells14020143] [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: 11/09/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Brain plasticity is at the basis of many cognitive functions, including learning and memory. It includes several mechanisms of synaptic and extrasynaptic changes, neurogenesis, and the formation and elimination of synapses. The plasticity of synaptic transmission involves the expression of immediate early genes (IEGs) that regulate neuronal activity, thereby supporting learning and memory. In addition, IEGs are involved in the regulation of brain cells' metabolism, proliferation, and survival, in the establishment of multicellular ensembles, and, presumably, in cell competition in the tissue. In this review, we analyze the current understanding of the role of IEGs (c-Fos, c-Myc, Arg3.1/Arc) in controlling brain plasticity in physiological and pathological conditions, including brain aging and neurodegeneration. This work might inspire new gene therapy strategies targeting IEGs to regulate synaptic plasticity, and potentially prevent or mitigate neurodegenerative diseases.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Research Center of Neurology, 125367 Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | | | | | - Anna S. Vetchinova
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | | | | |
Collapse
|
6
|
Griego E, Cerna C, Sollozo-Dupont I, Fuenzalida M, Galván EJ. Maternal immune activation alters temporal Precision of spike generation of CA1 pyramidal neurons by Unbalancing GABAergic inhibition intheOffspring. Brain Behav Immun 2025; 123:211-228. [PMID: 39293693 DOI: 10.1016/j.bbi.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
Infection during pregnancy represents a risk factor for neuropsychiatric disorders associated with neurodevelopmental alterations. A growing body of evidence from rodents and non-human primates shows that maternal inflammation induced by viral or bacterial infections results in several neurobiological alterations in the offspring. These changes may play an important role in the pathophysiology of psychiatric disorders like schizophrenia and autism spectrum disorders, whose clinical features include impairments in cognitive processing and social performance. Such alterations are causally associated with the maternal inflammatory response to infection rather than with the infection itself. Previously, we reported that CA1 pyramidal neurons of mice exposed to MIA exhibit increased excitability accompanied by a reduction in dendritic complexity. However, potential alterations in cellular and synaptic rules that shape the neuronal computational properties of the offspring remain to be determined. In this study, using mice as subjects, we identified a series of cellular and synaptic alterations endured by CA1 pyramidal neurons of the dorsal hippocampus in a lipopolysaccharide-induced maternal immune activation (MIA) model. Our data indicate that MIA reshapes the excitation-inhibition balance by decreasing the perisomatic GABAergic inhibition predominantly mediated by cholecystokinin-expressing Interneurons but not parvalbumin-expressing interneurons impinging on CA1 pyramidal neurons. These alterations yield a dysregulated amplification of the temporal and spatial synaptic integration. In addition, MIA-exposed offspring displayed social and anxiety-like abnormalities. These findings collectively contribute to understanding the cellular and synaptic alterations underlying the behavioral symptoms present in neurodevelopmental disorders associated with MIA.
Collapse
Affiliation(s)
- Ernesto Griego
- Departamento de Farmacobiología, Cinvestav, Ciudad de México, México; Current Address: Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Camila Cerna
- Centro de Neurobiología y Fisiopatología Integrativa, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Isabel Sollozo-Dupont
- Instituto Nacional de Perinatología, Isidro Espinosa de los Reyes. Ciudad de México, México
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Emilio J Galván
- Departamento de Farmacobiología, Cinvestav, Ciudad de México, México; Centro de Investigaciones sobre el Envejecimiento, CIE-Cinvestav, Ciudad de México, México.
| |
Collapse
|
7
|
Li Z, Lu W, Yang L, Lai N, Wang Y, Chen Z. Decade of TRAP progress: Insights and future prospects for advancing functional network research in epilepsy. Prog Neurobiol 2025; 244:102707. [PMID: 39725016 DOI: 10.1016/j.pneurobio.2024.102707] [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: 08/01/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Targeted Recombination in Active Populations (TRAP) represents an effective and extensively applied technique that has earned significant utilization in neuroscience over the past decade, primarily for identifying and modulating functionally activated neuronal ensembles associated with diverse behaviors. As epilepsy is a neurological disorder characterized by pathological hyper-excitatory networks, TRAP has already been widely applied in epilepsy research. However, the deployment of TRAP in this field remains underexplored, and there is significant potential for further application and development in epilepsy-related investigations. In this review, we embark on a concise examination of the mechanisms behind several TRAP tools, introduce the current applications of TRAP in epilepsy research, and collate the key advantages as well as limitations of TRAP. Furthermore, we sketch out perspectives on potential applications of TRAP in future epilepsy research, grounded in the present landscape and challenges of the field, as well as the ways TRAP has been embraced in other neuroscience domains.
Collapse
Affiliation(s)
- Zhisheng Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wangjialu Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Yang
- key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Nanxi Lai
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
8
|
Zheng Z, Liu Y, Mu R, Guo X, Feng Y, Guo C, Yang L, Qiu W, Zhang Q, Yang W, Dong Z, Qiu S, Dong Y, Cui Y. A small population of stress-responsive neurons in the hypothalamus-habenula circuit mediates development of depression-like behavior in mice. Neuron 2024; 112:3924-3939.e5. [PMID: 39389052 DOI: 10.1016/j.neuron.2024.09.012] [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: 12/11/2023] [Revised: 06/25/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Accumulating evidence has shown that various brain functions are associated with experience-activated neuronal ensembles. However, whether such neuronal ensembles are engaged in the pathogenesis of stress-induced depression remains elusive. Utilizing activity-dependent viral strategies in mice, we identified a small population of stress-responsive neurons, primarily located in the middle part of the lateral hypothalamus (mLH) and the medial part of the lateral habenula (LHbM). These neurons serve as "starter cells" to transmit stress-related information and mediate the development of depression-like behaviors during chronic stress. Starter cells in the mLH and LHbM form dominant connections, which are selectively potentiated by chronic stress. Silencing these connections during chronic stress prevents the development of depression-like behaviors, whereas activating these connections directly elicits depression-like behaviors without stress experience. Collectively, our findings dissect a core functional unit within the LH-LHb circuit that mediates the development of depression-like behaviors in mice.
Collapse
Affiliation(s)
- Zhiwei Zheng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiqin Liu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Ruiqi Mu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Xiaonan Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yirong Feng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Chen Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Liang Yang
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Wenxi Qiu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Qi Zhang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhaoqi Dong
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shuang Qiu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiyan Dong
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| | - Yihui Cui
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
9
|
Tozzi F, Guglielmo S, Paraciani C, van den Oever MC, Mainardi M, Cattaneo A, Origlia N. Involvement of a lateral entorhinal cortex engram in episodic-like memory recall. Cell Rep 2024; 43:114795. [PMID: 39325619 DOI: 10.1016/j.celrep.2024.114795] [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: 12/22/2023] [Revised: 07/16/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Episodic memory relies on the entorhinal cortex (EC), a crucial hub connecting the hippocampus and sensory processing regions. This study investigates the role of the lateral EC (LEC) in episodic-like memory in mice. Here, we employ the object-place-context-recognition task (OPCRT), a behavioral test used to study episodic-like memory in rodents. Electrophysiology in brain slices reveals that OPCRT specifically induces a shift in the threshold for the induction of synaptic plasticity in LEC superficial layer II. Additionally, a dual viral system is used to express chemogenetic receptors coupled to the c-Fos promoter in neurons recruited during the learning. We demonstrate that the inhibition of LEC neurons impairs the performance of the mice in the memory task, while their stimulation significantly facilitates memory recall. Our findings provide evidence for an episodic-like memory engram in the LEC and emphasize its role in memory processing within the broader network of episodic memory.
Collapse
Affiliation(s)
- Francesca Tozzi
- BIO@SNS Laboratory, Scuola Normale Superiore, Via Moruzzi 1, 56124 Pisa, Italy; Institute of Neuroscience, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Stefano Guglielmo
- BIO@SNS Laboratory, Scuola Normale Superiore, Via Moruzzi 1, 56124 Pisa, Italy; Institute of Neuroscience, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Camilla Paraciani
- Institute of Neuroscience, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Michel C van den Oever
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Marco Mainardi
- Institute of Neuroscience, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; Department of Biomedical Sciences University of Padova, 35122 Padova, Italy
| | - Antonino Cattaneo
- BIO@SNS Laboratory, Scuola Normale Superiore, Via Moruzzi 1, 56124 Pisa, Italy; European Brain Research Institute Rita Levi-Montalcini, Via del Fosso di Fiorano 64/65, 00143 Rome, Italy
| | - Nicola Origlia
- Institute of Neuroscience, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
10
|
Griffith EC, West AE, Greenberg ME. Neuronal enhancers fine-tune adaptive circuit plasticity. Neuron 2024; 112:3043-3057. [PMID: 39208805 PMCID: PMC11550865 DOI: 10.1016/j.neuron.2024.08.002] [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: 05/01/2023] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Neuronal activity-regulated gene expression plays a crucial role in sculpting neural circuits that underpin adaptive brain function. Transcriptional enhancers are now recognized as key components of gene regulation that orchestrate spatiotemporally precise patterns of gene transcription. We propose that the dynamics of enhancer activation uniquely position these genomic elements to finely tune activity-dependent cellular plasticity. Enhancer specificity and modularity can be exploited to gain selective genetic access to specific cell states, and the precise modulation of target gene expression within restricted cellular contexts enabled by targeted enhancer manipulation allows for fine-grained evaluation of gene function. Mounting evidence also suggests that enduring stimulus-induced changes in enhancer states can modify target gene activation upon restimulation, thereby contributing to a form of cell-wide metaplasticity. We advocate for focused exploration of activity-dependent enhancer function to gain new insight into the mechanisms underlying brain plasticity and cognitive dysfunction.
Collapse
Affiliation(s)
- Eric C Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | | |
Collapse
|
11
|
Megagiannis P, Mei Y, Yan RE, Yuan L, Wilde JJ, Eckersberg H, Suresh R, Tan X, Chen H, Farmer WT, Cha K, Le PU, Catoire H, Rochefort D, Kwan T, Yee BA, Dion P, Krishnaswamy A, Cloutier JF, Stifani S, Petrecca K, Yeo GW, Murai KK, Feng G, Rouleau GA, Ideker T, Sanjana NE, Zhou Y. Autism-associated CHD8 controls reactive gliosis and neuroinflammation via remodeling chromatin in astrocytes. Cell Rep 2024; 43:114637. [PMID: 39154337 DOI: 10.1016/j.celrep.2024.114637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 06/11/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
Reactive changes of glial cells during neuroinflammation impact brain disorders and disease progression. Elucidating the mechanisms that control reactive gliosis may help us to understand brain pathophysiology and improve outcomes. Here, we report that adult ablation of autism spectrum disorder (ASD)-associated CHD8 in astrocytes attenuates reactive gliosis via remodeling chromatin accessibility, changing gene expression. Conditional Chd8 deletion in astrocytes, but not microglia, suppresses reactive gliosis by impeding astrocyte proliferation and morphological elaboration. Astrocyte Chd8 ablation alleviates lipopolysaccharide-induced neuroinflammation and septic-associated hypothermia in mice. Astrocytic CHD8 plays an important role in neuroinflammation by altering the chromatin landscape, regulating metabolic and lipid-associated pathways, and astrocyte-microglia crosstalk. Moreover, we show that reactive gliosis can be directly mitigated in vivo using an adeno-associated virus (AAV)-mediated Chd8 gene editing strategy. These findings uncover a role of ASD-associated CHD8 in the adult brain, which may warrant future exploration of targeting chromatin remodelers in reactive gliosis and neuroinflammation in injury and neurological diseases.
Collapse
Affiliation(s)
- Platon Megagiannis
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Yuan Mei
- Division of Genetics, Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Rachel E Yan
- New York Genome Center, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Lin Yuan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Jonathan J Wilde
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailey Eckersberg
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Rahul Suresh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Xinzhu Tan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Hong Chen
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - W Todd Farmer
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Kuwook Cha
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Phuong Uyen Le
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Helene Catoire
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Daniel Rochefort
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tony Kwan
- McGill Genome Center and Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Brian A Yee
- Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patrick Dion
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Jean-Francois Cloutier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Kevin Petrecca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Guoping Feng
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| | - Neville E Sanjana
- New York Genome Center, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Yang Zhou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
12
|
Hou WH, Jariwala M, Wang KY, Seewald A, Lin YL, Liou YC, Ricci A, Ferraguti F, Lien CC, Capogna M. Inhibitory fear memory engram in the mouse central lateral amygdala. Cell Rep 2024; 43:114468. [PMID: 39106862 DOI: 10.1016/j.celrep.2024.114468] [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: 01/04/2022] [Revised: 04/29/2024] [Accepted: 06/24/2024] [Indexed: 08/09/2024] Open
Abstract
Engrams, which are cellular substrates of memory traces, have been identified in various brain areas, including the amygdala. While most identified engrams are composed of excitatory, glutamatergic neurons, GABAergic inhibitory engrams have been relatively overlooked. Here, we report the identification of an inhibitory engram in the central lateral amygdala (CeL), a key area for auditory fear conditioning. This engram is primarily composed of GABAergic somatostatin-expressing (SST(+)) and, to a lesser extent, protein kinase C-δ-expressing (PKC-δ(+)) neurons. Fear memory is accompanied by a preferential enhancement of synaptic inhibition onto PKC-δ(+) neurons. Silencing this CeL GABAergic engram disinhibits the activity of targeted extra-amygdaloid areas, selectively increasing the expression of fear. Our findings define the behavioral function of an engram formed exclusively by GABAergic inhibitory neurons in the mammalian brain.
Collapse
Affiliation(s)
- Wen-Hsien Hou
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark; Center for Proteins in Memory - PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| | - Meet Jariwala
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Proteins in Memory - PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| | - Kai-Yi Wang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Anna Seewald
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Yu-Ling Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chen Liou
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Alessia Ricci
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Marco Capogna
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark; Center for Proteins in Memory - PROMEMO, Danish National Research Foundation, Aarhus University, Aarhus, Denmark
| |
Collapse
|
13
|
Kveim VA, Salm L, Ulmer T, Lahr M, Kandler S, Imhof F, Donato F. Divergent recruitment of developmentally defined neuronal ensembles supports memory dynamics. Science 2024; 385:eadk0997. [PMID: 39146420 DOI: 10.1126/science.adk0997] [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: 11/03/2023] [Accepted: 06/24/2024] [Indexed: 08/17/2024]
Abstract
Memories are dynamic constructs whose properties change with time and experience. The biological mechanisms underpinning these dynamics remain elusive, particularly concerning how shifts in the composition of memory-encoding neuronal ensembles influence the evolution of a memory over time. By targeting developmentally distinct subpopulations of principal neurons, we discovered that memory encoding resulted in the concurrent establishment of multiple memory traces in the mouse hippocampus. Two of these traces were instantiated in subpopulations of early- and late-born neurons and followed distinct reactivation trajectories after encoding. The divergent recruitment of these subpopulations underpinned gradual reorganization of memory ensembles and modulated memory persistence and plasticity across multiple learning episodes. Thus, our findings reveal profound and intricate relationships between ensemble dynamics and the progression of memories over time.
Collapse
Affiliation(s)
- Vilde A Kveim
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Laurenz Salm
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Talia Ulmer
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Maria Lahr
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | | | - Fabia Imhof
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| | - Flavio Donato
- Biozentrum, Universität Basel, 4056 Basel, Switzerland
| |
Collapse
|
14
|
Méndez P, de la Vega-Ruiz R, Montes-Mellado A. Estrogenic regulation of hippocampal inhibitory system across lifespan. J Neuroendocrinol 2024:e13441. [PMID: 39143852 DOI: 10.1111/jne.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Estrogens produced in peripheral tissues and locally in the brain are potent neuromodulators. The function of the hippocampus, a brain region essential for episodic memory and spatial navigation, relies on the activity of ensembles of excitatory neurons whose activity is temporally and spatially coordinated by a wide diversity of inhibitory neurons (INs) types. Over the last years, we have accumulated evidence that indicates that estrogens regulate the function of hippocampal INs through different mechanisms, including transcriptional regulation and rapid nongenomic signaling. Here, we argue that the well-documented influence of estrogens on episodic memory may be related to the actions of local and peripheral estrogens on the heterogenous populations of hippocampal INs. We discuss how physiological changes in peripheral sex hormone levels throughout lifespan may interact with local brain sources to regulate IN function at different stages of life, from early hippocampal development to the aging brain. We conclude that considering INs as mediators of sex hormone actions in the hippocampus across the healthy life span will benefit our understanding of sex-biased neurodevelopmental disorders and physiological aging.
Collapse
|
15
|
Hughes BW, Huebschman JL, Tsvetkov E, Siemsen BM, Snyder KK, Akiki RM, Wood DJ, Penrod RD, Scofield MD, Berto S, Taniguchi M, Cowan CW. NPAS4 supports cocaine-conditioned cues in rodents by controlling the cell type-specific activation balance in the nucleus accumbens. Nat Commun 2024; 15:5971. [PMID: 39117647 PMCID: PMC11310321 DOI: 10.1038/s41467-024-50099-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Powerful associations that link drugs of abuse with cues in the drug-paired environment often serve as prepotent relapse triggers. Drug-associated contexts and cues activate ensembles of nucleus accumbens (NAc) neurons, including D1-class medium spiny neurons (MSNs) that typically promote, and D2-class MSNs that typically oppose, drug seeking. We found that in mice, cocaine conditioning upregulated transiently the activity-regulated transcription factor, Neuronal PAS Domain Protein 4 (NPAS4), in a small subset of NAc neurons. The NPAS4+ NAc ensemble was required for cocaine conditioned place preference. We also observed that NPAS4 functions within NAc D2-, but not D1-, MSNs to support cocaine-context associations and cue-induced cocaine, but not sucrose, seeking. Together, our data show that the NPAS4+ ensemble of NAc neurons is essential for cocaine-context associations in mice, and that NPAS4 itself functions in NAc D2-MSNs to support cocaine-context associations by suppressing drug-induced counteradaptations that oppose relapse-related behaviour.
Collapse
Affiliation(s)
- Brandon W Hughes
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica L Huebschman
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Benjamin M Siemsen
- Department of Anesthesiology, Medical University of South Carolina, Charleston, SC, USA
| | - Kirsten K Snyder
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Rose Marie Akiki
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel J Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, USA
| | - Rachel D Penrod
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Michael D Scofield
- Department of Anesthesiology, Medical University of South Carolina, Charleston, SC, USA
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
16
|
Wu X, Yang L, Li Z, Gu C, Jin K, Luo A, Rasheed NF, Fiutak I, Chao K, Chen A, Mao J, Chen Q, Ding W, Shen S. Aging-associated decrease of PGC-1α promotes pain chronification. Aging Cell 2024; 23:e14177. [PMID: 38760908 PMCID: PMC11320346 DOI: 10.1111/acel.14177] [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: 08/24/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 05/20/2024] Open
Abstract
Aging is generally associated with declining somatosensory function, which seems at odds with the high prevalence of chronic pain in older people. This discrepancy is partly related to the high prevalence of degenerative diseases such as osteoarthritis in older people. However, whether aging alters pain processing in the primary somatosensory cortex (S1), and if so, whether it promotes pain chronification is largely unknown. Herein, we report that older mice displayed prolonged nociceptive behavior following nerve injury when compared with mature adult mice. The expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) in S1 was decreased in older mice, whereas PGC-1α haploinsufficiency promoted prolonged nociceptive behavior after nerve injury. Both aging and PGC-1α haploinsufficiency led to abnormal S1 neural dynamics, revealed by intravital two-photon calcium imaging. Manipulating S1 neural dynamics affected nociceptive behavior after nerve injury: chemogenetic inhibition of S1 interneurons aggravated nociceptive behavior in naive mice; chemogenetic activation of S1 interneurons alleviated nociceptive behavior in older mice. More interestingly, adeno-associated virus-mediated expression of PGC-1α in S1 interneurons ameliorated aging-associated chronification of nociceptive behavior as well as aging-related S1 neural dynamic changes. Taken together, our results showed that aging-associated decrease of PGC-1α promotes pain chronification, which might be harnessed to alleviate the burden of chronic pain in older individuals.
Collapse
Affiliation(s)
- Xinbo Wu
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Present address:
Shanghai 10th HospitalTongji University School of MedicineShanghaiChina
| | - Liuyue Yang
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Zihua Li
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Chenzheng Gu
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Kaiyan Jin
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Andrew Luo
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | | | | - Kristina Chao
- Summer Intern ProgramMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Amy Chen
- Summer Intern ProgramMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jianren Mao
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Qian Chen
- Chinese Academy of SciencesZhongshan Institute for Drug Discovery, Shanghai Institute of Materia MedicaShanghaiChina
| | - Weihua Ding
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
17
|
Onishi T, Hirose K, Sakaba T. Molecular tools to capture active neural circuits. Front Neural Circuits 2024; 18:1449459. [PMID: 39100199 PMCID: PMC11294111 DOI: 10.3389/fncir.2024.1449459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
To understand how neurons and neural circuits function during behaviors, it is essential to record neuronal activity in the brain in vivo. Among the various technologies developed for recording neuronal activity, molecular tools that induce gene expression in an activity-dependent manner have attracted particular attention for their ability to clarify the causal relationships between neuronal activity and behavior. In this review, we summarize recently developed activity-dependent gene expression tools and their potential contributions to the study of neural circuits.
Collapse
Affiliation(s)
- Taichi Onishi
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo City, Bunkyo, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo City, Bunkyo, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto, Japan
| |
Collapse
|
18
|
Lee J, Jeong Y, Park S, Kim S, Oh H, Jin JA, Sohn JW, Kim D, Shin HS, Do Heo W. Phospholipase C beta 1 in the dentate gyrus gates fear memory formation through regulation of neuronal excitability. SCIENCE ADVANCES 2024; 10:eadj4433. [PMID: 38959322 PMCID: PMC11221510 DOI: 10.1126/sciadv.adj4433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 05/28/2024] [Indexed: 07/05/2024]
Abstract
Memory processes rely on a molecular signaling system that balances the interplay between positive and negative modulators. Recent research has focused on identifying memory-regulating genes and their mechanisms. Phospholipase C beta 1 (PLCβ1), highly expressed in the hippocampus, reportedly serves as a convergence point for signal transduction through G protein-coupled receptors. However, the detailed role of PLCβ1 in memory function has not been elucidated. Here, we demonstrate that PLCβ1 in the dentate gyrus functions as a memory suppressor. We reveal that mice lacking PLCβ1 in the dentate gyrus exhibit a heightened fear response and impaired memory extinction, and this excessive fear response is repressed by upregulation of PLCβ1 through its overexpression or activation using a newly developed optogenetic system. Last, our results demonstrate that PLCβ1 overexpression partially inhibits exaggerated fear response caused by traumatic experience. Together, PLCβ1 is crucial in regulating contextual fear memory formation and potentially enhancing the resilience to trauma-related conditions.
Collapse
Affiliation(s)
- Jinsu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yeonji Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Seahyung Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Sungsoo Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Hyunsik Oh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Ju-Ae Jin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
19
|
Hadler MD, Alle H, Geiger JRP. Parvalbumin interneuron cell-to-network plasticity: mechanisms and therapeutic avenues. Trends Pharmacol Sci 2024; 45:586-601. [PMID: 38763836 DOI: 10.1016/j.tips.2024.04.003] [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: 03/31/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Alzheimer's disease (AD) and schizophrenia (SCZ) represent two major neuropathological conditions with a high disease burden. Despite their distinct etiologies, patients suffering from AD or SCZ share a common burden of disrupted memory functions unattended by current therapies. Recent preclinical analyses highlight cell-type-specific contributions of parvalbumin interneurons (PVIs), particularly the plasticity of their cellular excitability, towards intact neuronal network function (cell-to-network plasticity) and memory performance. Here we argue that deficits of PVI cell-to-network plasticity may underlie memory deficits in AD and SCZ, and we explore two therapeutic avenues: the targeting of PVI-specific neuromodulation, including by neuropeptides, and the recruitment of network synchrony in the gamma frequency range (40 Hz) by external stimulation. We finally propose that these approaches be merged under consideration of recent insights into human brain physiology.
Collapse
Affiliation(s)
- Michael D Hadler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R P Geiger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
20
|
Cui K, Qi X, Liu Z, Sun W, Jiao P, Liu C, Tong J, Sun X, Sun H, Fu S, Wang J, Zheng Y, Liu T, Cui S, Liu F, Mao J, Zheng J, Wan Y, Yi M. Dominant activities of fear engram cells in the dorsal dentate gyrus underlie fear generalization in mice. PLoS Biol 2024; 22:e3002679. [PMID: 38995985 PMCID: PMC11244812 DOI: 10.1371/journal.pbio.3002679] [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: 10/18/2023] [Accepted: 05/16/2024] [Indexed: 07/14/2024] Open
Abstract
Over-generalized fear is a maladaptive response to harmless stimuli or situations characteristic of posttraumatic stress disorder (PTSD) and other anxiety disorders. The dorsal dentate gyrus (dDG) contains engram cells that play a crucial role in accurate memory retrieval. However, the coordination mechanism of neuronal subpopulations within the dDG network during fear generalization is not well understood. Here, with the Tet-off system combined with immunostaining and two-photon calcium imaging, we report that dDG fear engram cells labeled in the conditioned context constitutes a significantly higher proportion of dDG neurons activated in a similar context where mice show generalized fear. The activation of these dDG fear engram cells encoding the conditioned context is both sufficient and necessary for inducing fear generalization in the similar context. Activities of mossy cells in the ventral dentate gyrus (vMCs) are significantly suppressed in mice showing fear generalization in a similar context, and activating the vMCs-dDG pathway suppresses generalized but not conditioned fear. Finally, modifying fear memory engrams in the dDG with "safety" signals effectively rescues fear generalization. These findings reveal that the competitive advantage of dDG engram cells underlies fear generalization, which can be rescued by activating the vMCs-dDG pathway or modifying fear memory engrams, and provide novel insights into the dDG network as the neuronal basis of fear generalization.
Collapse
Affiliation(s)
- Kun Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
| | - Xuetao Qi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zilong Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Weiqi Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Peijie Jiao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chang Liu
- Beijing Life Science Academy, Beijing, China
| | - Jifu Tong
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaoyan Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Haojie Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- UCL School of Pharmacy, University College London, London, United Kingdom
| | - Su Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaxin Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yawen Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tianyu Liu
- Department of Anesthesiology, Peking University People's Hospital, Beijing, China
| | - Shuang Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Jian Mao
- Beijing Life Science Academy, Beijing, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ming Yi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| |
Collapse
|
21
|
Brito DVC, Kupke J, Sokolov R, Cambridge S, Both M, Bengtson CP, Rozov A, Oliveira AMM. Biphasic Npas4 expression promotes inhibitory plasticity and suppression of fear memory consolidation in mice. Mol Psychiatry 2024; 29:1929-1940. [PMID: 38347124 PMCID: PMC11408256 DOI: 10.1038/s41380-024-02454-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/19/2024]
Abstract
Long-term memories are believed to be encoded by unique transcriptional signatures in the brain. The expression of immediate early genes (IEG) promotes structural and molecular changes required for memory consolidation. Recent evidence has shown that the brain is equipped with mechanisms that not only promote, but actively constrict memory formation. However, it remains unknown whether IEG expression may play a role in memory suppression. Here we uncovered a novel function of the IEG neuronal PAS domain protein 4 (Npas4), as an inducible memory suppressor gene of highly salient aversive experiences. Using a contextual fear conditioning paradigm, we found that low stimulus salience leads to monophasic Npas4 expression, while highly salient learning induces a biphasic expression of Npas4 in the hippocampus. The later phase requires N-methyl-D-aspartate (NMDA) receptor activity and is independent of dopaminergic neurotransmission. Our in vivo pharmacological and genetic manipulation experiments suggested that the later phase of Npas4 expression restricts the consolidation of a fear memory and promote behavioral flexibility, by facilitating fear extinction and the contextual specificity of fear responses. Moreover, immunofluorescence and electrophysiological analysis revealed a concomitant increase in synaptic input from cholecystokinin (CCK)-expressing interneurons. Our results demonstrate how salient experiences evoke unique temporal patterns of IEG expression that fine-tune memory consolidation. Moreover, our study provides evidence for inducible gene expression associated with memory suppression as a possible mechanism to balance the consolidation of highly salient memories, and thereby to evade the formation of maladaptive behavior.
Collapse
Affiliation(s)
- David V C Brito
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
- ABC-RI, Algarve Biomedical Center Research Institute, 8005-139, Faro, Portugal
- Faculdade de Medicina e Ciências Biomédicas, Universidade do Algarve, 8005-139, Faro, Portugal
| | - Janina Kupke
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, the Netherlands
| | - Rostilav Sokolov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia
- Federal Center of Brain Research and Neurotechnology, 117513, Moscow, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny, Novgorod, Russia
| | - Sidney Cambridge
- Anatomy II, Dr. Senckenberg Anatomy, Goethe-University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Martin Both
- Institute of Physiology and Pathophysiology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
| | - Andrei Rozov
- Federal Center of Brain Research and Neurotechnology, 117513, Moscow, Russia
- Institute of Physiology and Pathophysiology, Medical Faculty, Heidelberg University, 69120, Heidelberg, Germany
- OpenLab of Neurobiology, Kazan Federal University, 420008, Kazan, Russia
| | - Ana M M Oliveira
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany.
- Department of Molecular and Cellular Cognition Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany.
| |
Collapse
|
22
|
Huang M, Pieraut S, Cao J, de Souza Polli F, Roncace V, Shen G, Ramos-Medina C, Lee H, Maximov A. Nr4a1 regulates cell-specific transcriptional programs in inhibitory GABAergic interneurons. Neuron 2024; 112:2031-2044.e7. [PMID: 38754414 PMCID: PMC11189749 DOI: 10.1016/j.neuron.2024.03.018] [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: 01/12/2023] [Revised: 01/29/2024] [Accepted: 03/14/2024] [Indexed: 05/18/2024]
Abstract
The patterns of synaptic connectivity and physiological properties of diverse neuron types are shaped by distinct gene sets. Our study demonstrates that, in the mouse forebrain, the transcriptional profiles of inhibitory GABAergic interneurons are regulated by Nr4a1, an orphan nuclear receptor whose expression is transiently induced by sensory experiences and is required for normal learning. Nr4a1 exerts contrasting effects on the local axonal wiring of parvalbumin- and somatostatin-positive interneurons, which innervate different subcellular domains of their postsynaptic partners. The loss of Nr4a1 activity in these interneurons results in bidirectional, cell-type-specific transcriptional switches across multiple gene families, including those involved in surface adhesion and repulsion. Our findings reveal that combinatorial synaptic organizing codes are surprisingly flexible and highlight a mechanism by which inducible transcription factors can influence neural circuit structure and function.
Collapse
Affiliation(s)
- Min Huang
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; The Kellogg School of Science and Technology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Simon Pieraut
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jasmine Cao
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Filip de Souza Polli
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vincenzo Roncace
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gloria Shen
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Carlos Ramos-Medina
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - HeeYang Lee
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; The Kellogg School of Science and Technology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Anton Maximov
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, USA; The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
23
|
Nakajima N, Kamijo T, Hayakawa H, Sugisaki E, Aihara T. Modification of temporal pattern sensitivity for inputs from medial entorhinal cortex by lateral inputs in hippocampal granule cells. Cogn Neurodyn 2024; 18:1047-1059. [PMID: 38826655 PMCID: PMC11143144 DOI: 10.1007/s11571-023-09964-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/08/2023] [Accepted: 03/23/2023] [Indexed: 06/04/2024] Open
Abstract
The medial dendrites (MDs) of granule cells (GCs) receive spatial information through the medial entorhinal cortex (MEC) from the entorhinal cortex in the rat hippocampus while the distal dendrites (DDs) of GCs receive non-spatial information (sensory inputs) through the lateral entorhinal cortex (LEC). However, it is unclear how information processing through the two pathways is managed in GCs. In this study, we investigated associative information processing between two independent inputs to MDs and DDs. First, in physiological experiments, to compare response characteristics between MDs and DDs, electrical stimuli comprising five pulses were applied to the MPP or LPP in rat hippocampal slices. These stimuli transiently decreased the excitatory postsynaptic potentials (EPSPs) of successive input pulses to MDs, whereas EPSPs to DDs showed sustained responses. Next, in computational experiments using a local network model obtained by fitting of the physiological experimental data, we compared associative information processing between DDs and MDs. The results showed that the temporal pattern sensitivity for burst inputs to MDs depended on the frequency of the random pulse inputs applied to DDs. On the other hand, with lateral inhibition to GCs from interneurons, the temporal pattern sensitivity for burst inputs to MDs was enhanced or tuned up according to the frequency of the random pulse inputs to the other cells. Thus, our results suggest that the temporal pattern sensitivity of spatial information depends on the non-spatial inputs to GCs.
Collapse
Affiliation(s)
- Naoki Nakajima
- Graduated School of Engineering, Tamagawa University, Tokyo, Japan
| | - Tadanobu Kamijo
- Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | | | - Eriko Sugisaki
- Brain Science Institute, Tamagawa University, Tokyo, Japan
| | - Takeshi Aihara
- Graduated School of Engineering, Tamagawa University, Tokyo, Japan
- Brain Science Institute, Tamagawa University, Tokyo, Japan
| |
Collapse
|
24
|
Liu Y, Ye S, Li XN, Li WG. Memory Trace for Fear Extinction: Fragile yet Reinforceable. Neurosci Bull 2024; 40:777-794. [PMID: 37812300 PMCID: PMC11178705 DOI: 10.1007/s12264-023-01129-3] [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: 04/02/2023] [Accepted: 06/08/2023] [Indexed: 10/10/2023] Open
Abstract
Fear extinction is a biological process in which learned fear behavior diminishes without anticipated reinforcement, allowing the organism to re-adapt to ever-changing situations. Based on the behavioral hypothesis that extinction is new learning and forms an extinction memory, this new memory is more readily forgettable than the original fear memory. The brain's cellular and synaptic traces underpinning this inherently fragile yet reinforceable extinction memory remain unclear. Intriguing questions are about the whereabouts of the engram neurons that emerged during extinction learning and how they constitute a dynamically evolving functional construct that works in concert to store and express the extinction memory. In this review, we discuss recent advances in the engram circuits and their neural connectivity plasticity for fear extinction, aiming to establish a conceptual framework for understanding the dynamic competition between fear and extinction memories in adaptive control of conditioned fear responses.
Collapse
Affiliation(s)
- Ying Liu
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Shuai Ye
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Xin-Ni Li
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
25
|
Barti B, Dudok B, Kenesei K, Zöldi M, Miczán V, Balla GY, Zala D, Tasso M, Sagheddu C, Kisfali M, Tóth B, Ledri M, Vizi ES, Melis M, Barna L, Lenkei Z, Soltész I, Katona I. Presynaptic nanoscale components of retrograde synaptic signaling. SCIENCE ADVANCES 2024; 10:eado0077. [PMID: 38809980 PMCID: PMC11135421 DOI: 10.1126/sciadv.ado0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
While our understanding of the nanoscale architecture of anterograde synaptic transmission is rapidly expanding, the qualitative and quantitative molecular principles underlying distinct mechanisms of retrograde synaptic communication remain elusive. We show that a particular form of tonic cannabinoid signaling is essential for setting target cell-dependent synaptic variability. It does not require the activity of the two major endocannabinoid-producing enzymes. Instead, by developing a workflow for physiological, anatomical, and molecular measurements at the same unitary synapse, we demonstrate that the nanoscale stoichiometric ratio of type 1 cannabinoid receptors (CB1Rs) to the release machinery is sufficient to predict synapse-specific release probability. Accordingly, selective decrease of extrasynaptic CB1Rs does not affect synaptic transmission, whereas in vivo exposure to the phytocannabinoid Δ9-tetrahydrocannabinol disrupts the intrasynaptic nanoscale stoichiometry and reduces synaptic variability. These findings imply that synapses leverage the nanoscale stoichiometry of presynaptic receptor coupling to the release machinery to establish synaptic strength in a target cell-dependent manner.
Collapse
Affiliation(s)
- Benjámin Barti
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Barna Dudok
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Departments of Neurology and Neuroscience, Baylor College of Medicine, 1 Baylor Plz, Houston, TX 77030, USA
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - Kata Kenesei
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miklós Zöldi
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Vivien Miczán
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Center, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Gyula Y. Balla
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Diana Zala
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Mariana Tasso
- Institute of Nanosystems, School of Bio and Nanotechnologies, National University of San Martín - CONICET, 25 de Mayo Ave., 1021 San Martín, Argentina
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - Máté Kisfali
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- BiTrial Ltd., Tállya st 23, H-1121 Budapest, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Szt. Gellért square 4, H-1111 Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Marco Ledri
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 17, BMC A11, 221 84 Lund, Sweden
| | - E. Sylvester Vizi
- Molecular Pharmacology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - László Barna
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
| | - Zsolt Lenkei
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Iván Soltész
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - István Katona
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| |
Collapse
|
26
|
Pérez-Sisqués L, Bhatt SU, Matuleviciute R, Gileadi TE, Kramar E, Graham A, Garcia FG, Keiser A, Matheos DP, Cain JA, Pittman AM, Andreae LC, Fernandes C, Wood MA, Giese KP, Basson MA. The Intellectual Disability Risk Gene Kdm5b Regulates Long-Term Memory Consolidation in the Hippocampus. J Neurosci 2024; 44:e1544232024. [PMID: 38575342 PMCID: PMC11079963 DOI: 10.1523/jneurosci.1544-23.2024] [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: 02/18/2024] [Revised: 03/21/2024] [Accepted: 03/30/2024] [Indexed: 04/06/2024] Open
Abstract
The histone lysine demethylase KDM5B is implicated in recessive intellectual disability disorders, and heterozygous, protein-truncating variants in KDM5B are associated with reduced cognitive function in the population. The KDM5 family of lysine demethylases has developmental and homeostatic functions in the brain, some of which appear to be independent of lysine demethylase activity. To determine the functions of KDM5B in hippocampus-dependent learning and memory, we first studied male and female mice homozygous for a Kdm5b Δ ARID allele that lacks demethylase activity. Kdm5b Δ ARID/ Δ ARID mice exhibited hyperactivity and long-term memory deficits in hippocampus-dependent learning tasks. The expression of immediate early, activity-dependent genes was downregulated in these mice and hyperactivated upon a learning stimulus compared with wild-type (WT) mice. A number of other learning-associated genes were also significantly dysregulated in the Kdm5b Δ ARID/ Δ ARID hippocampus. Next, we knocked down Kdm5b specifically in the adult, WT mouse hippocampus with shRNA. Kdm5b knockdown resulted in spontaneous seizures, hyperactivity, and hippocampus-dependent long-term memory and long-term potentiation deficits. These findings identify KDM5B as a critical regulator of gene expression and synaptic plasticity in the adult hippocampus and suggest that at least some of the cognitive phenotypes associated with KDM5B gene variants are caused by direct effects on memory consolidation mechanisms.
Collapse
Affiliation(s)
- Leticia Pérez-Sisqués
- Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - Shail U Bhatt
- Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
| | - Rugile Matuleviciute
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - Talia E Gileadi
- Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
| | - Eniko Kramar
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California Irvine, Irvine, California, California 92697
| | - Andrew Graham
- Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
| | - Franklin G Garcia
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California Irvine, Irvine, California, California 92697
| | - Ashley Keiser
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California Irvine, Irvine, California, California 92697
| | - Dina P Matheos
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California Irvine, Irvine, California, California 92697
| | - James A Cain
- Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
| | - Alan M Pittman
- St. George's University of London, London SW17 0RE, United Kingdom
| | - Laura C Andreae
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - Cathy Fernandes
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AB, United Kingdom
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California Irvine, Irvine, California, California 92697
| | - K Peter Giese
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, United Kingdom
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, Guy's Hospital, King's College London, London SE1 9RT, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Hatherly Laboratories, Exeter EX4 4PS, United Kingdom
| |
Collapse
|
27
|
Jeong M, Jang JH, Oh SJ, Park J, Lee J, Hwang S, Oh YS. Maladaptation of dentate gyrus mossy cells mediates contextual discrimination deficit after traumatic stress. Cell Rep 2024; 43:114000. [PMID: 38527063 DOI: 10.1016/j.celrep.2024.114000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 03/27/2024] Open
Abstract
Fear overgeneralization is a maladaptive response to traumatic stress that is associated with the inability to discriminate between threat and safety contexts, a hallmark feature of post-traumatic stress disorder (PTSD). However, the neural mechanisms underlying this deficit remain unclear. Here, we show that traumatic stress exposure impairs contextual discrimination between threat and safety contexts in the learned helplessness (LH) model. Mossy cells (MCs) in the dorsal hippocampus are suppressed in response to traumatic stress. Bidirectional manipulation of MC activity in the LH model reveals that MC inhibition is causally linked to impaired contextual discrimination. Mechanistically, MC inhibition increases the number of active granule cells in a given context, significantly overlapping context-specific ensembles. Our study demonstrates that maladaptive inhibition of MCs after traumatic stress is a substantial mechanism underlying fear overgeneralization with contextual discrimination deficit, suggesting a potential therapeutic target for cognitive symptoms of PTSD.
Collapse
Affiliation(s)
- Minseok Jeong
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Jin-Hyeok Jang
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Seo-Jin Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Jeongrak Park
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Junseop Lee
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Sehyeon Hwang
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Yong-Seok Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea; Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, 61 Cheomdan-ro, Daegu 41062, Republic of Korea.
| |
Collapse
|
28
|
Shmal D, Mantero G, Floss T, Benfenati F, Maya-Vetencourt JF. Restoring vision in adult amblyopia by enhancing plasticity through deletion of the transcriptional repressor REST. iScience 2024; 27:109507. [PMID: 38591011 PMCID: PMC11000024 DOI: 10.1016/j.isci.2024.109507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/30/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Visual cortical plasticity is high during early life, but gradually decreases with development. This is due to the Otx2-driven maturation of intracortical inhibition that parallels the condensation of extracellular matrix components into perineuronal nets mainly around parvalbumin-positive GABAergic neurons. Repressor Element 1 Silencing Transcription (REST) epigenetically controls the expression of a plethora of neuron-specific genes. We demonstrate that the conditional knockout of REST in the primary visual cortex of adult mice induces a shift of ocular dominance after short-term monocular deprivation and promotes the recovery of vision in long-term deprived animals after reverse suture. These phenomena paralleled a reduction of perineuronal net density and increased expression of REST target genes, but not of the homeoprotein Otx2 in the visual cortex contralateral to the deprived eye. This shows that REST regulates adult visual cortical plasticity and is a potential therapeutic target to restore vision in adult amblyopia by enhancing V1 plasticity.
Collapse
Affiliation(s)
- Dmytro Shmal
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Giulia Mantero
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Thomas Floss
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - José Fernando Maya-Vetencourt
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
29
|
Wang L, Park L, Wu W, King D, Vega-Medina A, Raven F, Martinez J, Ensing A, McDonald K, Yang Z, Jiang S, Aton SJ. Sleep-dependent engram reactivation during hippocampal memory consolidation associated with subregion-specific biosynthetic changes. iScience 2024; 27:109408. [PMID: 38523798 PMCID: PMC10957462 DOI: 10.1016/j.isci.2024.109408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/14/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Post-learning sleep is essential for hippocampal memory processing, including contextual fear memory consolidation. We labeled context-encoding engram neurons in the hippocampal dentate gyrus (DG) and assessed reactivation of these neurons after fear learning. Post-learning sleep deprivation (SD) selectively disrupted reactivation of inferior blade DG engram neurons, linked to SD-induced suppression of neuronal activity in the inferior, but not superior DG blade. Subregion-specific spatial profiling of transcripts revealed that transcriptomic responses to SD differed greatly between hippocampal CA1, CA3, and DG inferior blade, superior blade, and hilus. Activity-driven transcripts, and those associated with cytoskeletal remodeling, were selectively suppressed in the inferior blade. Critically, learning-driven transcriptomic changes differed dramatically between the DG blades and were absent from all other regions. Together, these data suggest that the DG is critical for sleep-dependent memory consolidation, and that the effects of sleep loss on the hippocampus are highly subregion-specific.
Collapse
Affiliation(s)
- Lijing Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lauren Park
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Weisheng Wu
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dana King
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexis Vega-Medina
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Frank Raven
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jessy Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amy Ensing
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katherine McDonald
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongying Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
30
|
Lopez MR, Wasberg SMH, Gagliardi CM, Normandin ME, Muzzio IA. Mystery of the memory engram: History, current knowledge, and unanswered questions. Neurosci Biobehav Rev 2024; 159:105574. [PMID: 38331127 DOI: 10.1016/j.neubiorev.2024.105574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/22/2023] [Accepted: 02/03/2024] [Indexed: 02/10/2024]
Abstract
The quest to understand the memory engram has intrigued humans for centuries. Recent technological advances, including genetic labelling, imaging, optogenetic and chemogenetic techniques, have propelled the field of memory research forward. These tools have enabled researchers to create and erase memory components. While these innovative techniques have yielded invaluable insights, they often focus on specific elements of the memory trace. Genetic labelling may rely on a particular immediate early gene as a marker of activity, optogenetics may activate or inhibit one specific type of neuron, and imaging may capture activity snapshots in a given brain region at specific times. Yet, memories are multifaceted, involving diverse arrays of neuronal subpopulations, circuits, and regions that work in concert to create, store, and retrieve information. Consideration of contributions of both excitatory and inhibitory neurons, micro and macro circuits across brain regions, the dynamic nature of active ensembles, and representational drift is crucial for a comprehensive understanding of the complex nature of memory.
Collapse
Affiliation(s)
- M R Lopez
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - S M H Wasberg
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - C M Gagliardi
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - M E Normandin
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - I A Muzzio
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
31
|
Eom K, Jung J, Kim B, Hyun JH. Molecular tools for recording and intervention of neuronal activity. Mol Cells 2024; 47:100048. [PMID: 38521352 PMCID: PMC11021360 DOI: 10.1016/j.mocell.2024.100048] [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: 12/15/2023] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024] Open
Abstract
Observing the activity of neural networks is critical for the identification of learning and memory processes, as well as abnormal activities of neural circuits in disease, particularly for the purpose of tracking disease progression. Methodologies for describing the activity history of neural networks using molecular biology techniques first utilized genes expressed by active neurons, followed by the application of recently developed techniques including optogenetics and incorporation of insights garnered from other disciplines, including chemistry and physics. In this review, we will discuss ways in which molecular biological techniques used to describe the activity of neural networks have evolved along with the potential for future development.
Collapse
Affiliation(s)
- Kisang Eom
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jinhwan Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Byungsoo Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, Republic of Korea.
| |
Collapse
|
32
|
Jovasevic V, Wood EM, Cicvaric A, Zhang H, Petrovic Z, Carboncino A, Parker KK, Bassett TE, Moltesen M, Yamawaki N, Login H, Kalucka J, Sananbenesi F, Zhang X, Fischer A, Radulovic J. Formation of memory assemblies through the DNA-sensing TLR9 pathway. Nature 2024; 628:145-153. [PMID: 38538785 PMCID: PMC10990941 DOI: 10.1038/s41586-024-07220-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
As hippocampal neurons respond to diverse types of information1, a subset assembles into microcircuits representing a memory2. Those neurons typically undergo energy-intensive molecular adaptations, occasionally resulting in transient DNA damage3-5. Here we found discrete clusters of excitatory hippocampal CA1 neurons with persistent double-stranded DNA (dsDNA) breaks, nuclear envelope ruptures and perinuclear release of histone and dsDNA fragments hours after learning. Following these early events, some neurons acquired an inflammatory phenotype involving activation of TLR9 signalling and accumulation of centrosomal DNA damage repair complexes6. Neuron-specific knockdown of Tlr9 impaired memory while blunting contextual fear conditioning-induced changes of gene expression in specific clusters of excitatory CA1 neurons. Notably, TLR9 had an essential role in centrosome function, including DNA damage repair, ciliogenesis and build-up of perineuronal nets. We demonstrate a novel cascade of learning-induced molecular events in discrete neuronal clusters undergoing dsDNA damage and TLR9-mediated repair, resulting in their recruitment to memory circuits. With compromised TLR9 function, this fundamental memory mechanism becomes a gateway to genomic instability and cognitive impairments implicated in accelerated senescence, psychiatric disorders and neurodegenerative disorders. Maintaining the integrity of TLR9 inflammatory signalling thus emerges as a promising preventive strategy for neurocognitive deficits.
Collapse
Affiliation(s)
- Vladimir Jovasevic
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M Wood
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana Cicvaric
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hui Zhang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zorica Petrovic
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anna Carboncino
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kendra K Parker
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Thomas E Bassett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Moltesen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Naoki Yamawaki
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Hande Login
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Farahnaz Sananbenesi
- Department for Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medical Center, Göttingen, Germany
- Cluster of Excellence MBExC, University of Göttingen, Göttingen, Germany
| | - Xusheng Zhang
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andre Fischer
- Department for Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medical Center, Göttingen, Germany
- Cluster of Excellence MBExC, University of Göttingen, Göttingen, Germany
| | - Jelena Radulovic
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- PROMEMO, Aarhus University, Aarhus, Denmark.
- DANDRITE, Aarhus University, Aarhus, Denmark.
- Department of Psychiatry and Behavioral Sciences, Psychiatry Research Institute Montefiore Einstein (PRIME), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
33
|
Suthard RL, Senne RA, Buzharsky MD, Diep AH, Pyo AY, Ramirez S. Engram reactivation mimics cellular signatures of fear. Cell Rep 2024; 43:113850. [PMID: 38401120 DOI: 10.1016/j.celrep.2024.113850] [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: 12/14/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/26/2024] Open
Abstract
Engrams, or the physical substrate of memory, recruit heterogeneous cell types. Targeted reactivation of neurons processing discrete memories drives the behavioral expression of memory, though the underlying landscape of recruited cells and their real-time responses remain elusive. To understand how artificial stimulation of fear affects intra-hippocampal neuron-astrocyte dynamics as well as their behavioral consequences, we express channelrhodopsin-2 in an activity-dependent manner within dentate gyrus neurons while recording both cell types with fiber photometry in hippocampal ventral CA1 across learning and memory. Both cell types exhibit shock responsiveness, with astrocytic calcium events uniquely modulated by fear conditioning. Optogenetic stimulation of a hippocampus-mediated engram recapitulates coordinated calcium signatures time locked to freezing, mirroring those observed during natural fear memory recall. Our findings reveal cell-type-specific dynamics in the hippocampus during freezing behavior, emphasizing neuronal-astrocytic coupling as a shared mechanism enabling both natural and artificially induced memory retrieval and the behavioral expression of fear.
Collapse
Affiliation(s)
- Rebecca L Suthard
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Ryan A Senne
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Michelle D Buzharsky
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Anh H Diep
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Angela Y Pyo
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA
| | - Steve Ramirez
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Neurophotonics Center, and Photonics Center, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
34
|
Hu Y, Du W, Qi J, Luo H, Zhang Z, Luo M, Wang Y. Comparative brain-wide mapping of ketamine- and isoflurane-activated nuclei and functional networks in the mouse brain. eLife 2024; 12:RP88420. [PMID: 38512722 PMCID: PMC10957177 DOI: 10.7554/elife.88420] [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] [Indexed: 03/23/2024] Open
Abstract
Ketamine (KET) and isoflurane (ISO) are two widely used general anesthetics, yet their distinct and shared neurophysiological mechanisms remain elusive. In this study, we conducted a comparative analysis of the effects of KET and ISO on c-Fos expression across the mouse brain, utilizing hierarchical clustering and c-Fos-based functional network analysis to evaluate the responses of individual brain regions to each anesthetic. Our findings reveal that KET activates a wide range of brain regions, notably in the cortical and subcortical nuclei involved in sensory, motor, emotional, and reward processing, with the temporal association areas (TEa) as a strong hub, suggesting a top-down mechanism affecting consciousness by primarily targeting higher order cortical networks. In contrast, ISO predominantly influences brain regions in the hypothalamus, impacting neuroendocrine control, autonomic function, and homeostasis, with the locus coeruleus (LC) as a connector hub, indicating a bottom-up mechanism in anesthetic-induced unconsciousness. KET and ISO both activate brain areas involved in sensory processing, memory and cognition, reward and motivation, as well as autonomic and homeostatic control, highlighting their shared effects on various neural pathways. In conclusion, our results highlight the distinct but overlapping effects of KET and ISO, enriching our understanding of the mechanisms underlying general anesthesia.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Jiangtao Qi
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
| | - Zhao Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
35
|
Sun W, Liu Z, Jiang X, Chen MB, Dong H, Liu J, Südhof TC, Quake SR. Spatial transcriptomics reveal neuron-astrocyte synergy in long-term memory. Nature 2024; 627:374-381. [PMID: 38326616 PMCID: PMC10937396 DOI: 10.1038/s41586-023-07011-6] [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: 03/16/2023] [Accepted: 12/21/2023] [Indexed: 02/09/2024]
Abstract
Memory encodes past experiences, thereby enabling future plans. The basolateral amygdala is a centre of salience networks that underlie emotional experiences and thus has a key role in long-term fear memory formation1. Here we used spatial and single-cell transcriptomics to illuminate the cellular and molecular architecture of the role of the basolateral amygdala in long-term memory. We identified transcriptional signatures in subpopulations of neurons and astrocytes that were memory-specific and persisted for weeks. These transcriptional signatures implicate neuropeptide and BDNF signalling, MAPK and CREB activation, ubiquitination pathways, and synaptic connectivity as key components of long-term memory. Notably, upon long-term memory formation, a neuronal subpopulation defined by increased Penk and decreased Tac expression constituted the most prominent component of the memory engram of the basolateral amygdala. These transcriptional changes were observed both with single-cell RNA sequencing and with single-molecule spatial transcriptomics in intact slices, thereby providing a rich spatial map of a memory engram. The spatial data enabled us to determine that this neuronal subpopulation interacts with adjacent astrocytes, and functional experiments show that neurons require interactions with astrocytes to encode long-term memory.
Collapse
Affiliation(s)
- Wenfei Sun
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhihui Liu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Xian Jiang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle B Chen
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Hua Dong
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan Liu
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Initiative, Redwood City, CA, USA.
| |
Collapse
|
36
|
Tomé DF, Zhang Y, Aida T, Mosto O, Lu Y, Chen M, Sadeh S, Roy DS, Clopath C. Dynamic and selective engrams emerge with memory consolidation. Nat Neurosci 2024; 27:561-572. [PMID: 38243089 PMCID: PMC10917686 DOI: 10.1038/s41593-023-01551-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/12/2023] [Indexed: 01/21/2024]
Abstract
Episodic memories are encoded by experience-activated neuronal ensembles that remain necessary and sufficient for recall. However, the temporal evolution of memory engrams after initial encoding is unclear. In this study, we employed computational and experimental approaches to examine how the neural composition and selectivity of engrams change with memory consolidation. Our spiking neural network model yielded testable predictions: memories transition from unselective to selective as neurons drop out of and drop into engrams; inhibitory activity during recall is essential for memory selectivity; and inhibitory synaptic plasticity during memory consolidation is critical for engrams to become selective. Using activity-dependent labeling, longitudinal calcium imaging and a combination of optogenetic and chemogenetic manipulations in mouse dentate gyrus, we conducted contextual fear conditioning experiments that supported our model's predictions. Our results reveal that memory engrams are dynamic and that changes in engram composition mediated by inhibitory plasticity are crucial for the emergence of memory selectivity.
Collapse
Affiliation(s)
- Douglas Feitosa Tomé
- Department of Bioengineering, Imperial College London, London, UK.
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | - Ying Zhang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Center for Life Sciences & IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Tomomi Aida
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Olivia Mosto
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yifeng Lu
- Center for Life Sciences & IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Mandy Chen
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sadra Sadeh
- Department of Bioengineering, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Dheeraj S Roy
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Claudia Clopath
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
37
|
Han KA, Yoon TH, Kim J, Lee J, Lee JY, Jang G, Um JW, Kim JK, Ko J. Specification of neural circuit architecture shaped by context-dependent patterned LAR-RPTP microexons. Nat Commun 2024; 15:1624. [PMID: 38388459 PMCID: PMC10883964 DOI: 10.1038/s41467-024-45695-0] [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: 07/13/2023] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
LAR-RPTPs are evolutionarily conserved presynaptic cell-adhesion molecules that orchestrate multifarious synaptic adhesion pathways. Extensive alternative splicing of LAR-RPTP mRNAs may produce innumerable LAR-RPTP isoforms that act as regulatory "codes" for determining the identity and strength of specific synapse signaling. However, no direct evidence for this hypothesis exists. Here, using targeted RNA sequencing, we detected LAR-RPTP mRNAs in diverse cell types across adult male mouse brain areas. We found pronounced cell-type-specific patterns of two microexons, meA and meB, in Ptprd mRNAs. Moreover, diverse neural circuits targeting the same neuronal populations were dictated by the expression of different Ptprd variants with distinct inclusion patterns of microexons. Furthermore, conditional ablation of Ptprd meA+ variants at presynaptic loci of distinct hippocampal circuits impaired distinct modes of synaptic transmission and objection-location memory. Activity-triggered alterations of the presynaptic Ptprd meA code in subicular neurons mediates NMDA receptor-mediated postsynaptic responses in CA1 neurons and objection-location memory. Our data provide the evidence of cell-type- and/or circuit-specific expression patterns in vivo and physiological functions of LAR-RPTP microexons that are dynamically regulated.
Collapse
Affiliation(s)
- Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Taek-Han Yoon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Jinhu Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
| | - Jusung Lee
- Department of New Biology, DGIST, Daegu, 42988, Korea
| | - Ju Yeon Lee
- Korea Basic Science Institute, Research Center for Bioconvergence Analysis, Cheongju, 28119, Korea
| | - Gyubin Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, Daegu, 42988, Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Korea.
- Center for Synapse Diversity and Specificity, DGIST, Daegu, 42988, Korea.
| |
Collapse
|
38
|
Chapman DP, Power SD, Vicini S, Ryan TJ, Burns MP. Amnesia after Repeated Head Impact Is Caused by Impaired Synaptic Plasticity in the Memory Engram. J Neurosci 2024; 44:e1560232024. [PMID: 38228367 PMCID: PMC10883615 DOI: 10.1523/jneurosci.1560-23.2024] [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: 08/16/2023] [Revised: 12/15/2023] [Accepted: 01/06/2024] [Indexed: 01/18/2024] Open
Abstract
Subconcussive head impacts are associated with the development of acute and chronic cognitive deficits. We recently reported that high-frequency head impact (HFHI) causes chronic cognitive deficits in mice through synaptic changes. To better understand the mechanisms underlying HFHI-induced memory decline, we used TRAP2/Ai32 transgenic mice to enable visualization and manipulation of memory engrams. We labeled the fear memory engram in male and female mice exposed to an aversive experience and subjected them to sham or HFHI. Upon subsequent exposure to natural memory recall cues, sham, but not HFHI, mice successfully retrieved fearful memories. In sham mice the hippocampal engram neurons exhibited synaptic plasticity, evident in amplified AMPA:NMDA ratio, enhanced AMPA-weighted tau, and increased dendritic spine volume compared with nonengram neurons. In contrast, although HFHI mice retained a comparable number of hippocampal engram neurons, these neurons did not undergo synaptic plasticity. This lack of plasticity coincided with impaired activation of the engram network, leading to retrograde amnesia in HFHI mice. We validated that the memory deficits induced by HFHI stem from synaptic plasticity impairments by artificially activating the engram using optogenetics and found that stimulated memory recall was identical in both sham and HFHI mice. Our work shows that chronic cognitive impairment after HFHI is a result of deficiencies in synaptic plasticity instead of a loss in neuronal infrastructure, and we can reinstate a forgotten memory in the amnestic brain by stimulating the memory engram. Targeting synaptic plasticity may have therapeutic potential for treating memory impairments caused by repeated head impacts.
Collapse
Affiliation(s)
- Daniel P Chapman
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Sarah D Power
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, D02 PN40 Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, D02 PN40 Ireland
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195 Berlin, Germany
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Departments of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, D02 PN40 Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, D02 PN40 Ireland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, Victoria 3052, Australia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, MSG IMI, Canada
| | - Mark P Burns
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057
| |
Collapse
|
39
|
Vingan I, Phatarpekar S, Tung VSK, Hernández AI, Evgrafov OV, Alarcon JM. Spatially Resolved Transcriptomic Signatures of Hippocampal Subregions and Arc-Expressing Ensembles in Active Place Avoidance Memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573225. [PMID: 38260257 PMCID: PMC10802250 DOI: 10.1101/2023.12.30.573225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc+ and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc+ and Arc- spatial transcriptomic spots.
Collapse
Affiliation(s)
- Isaac Vingan
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Shwetha Phatarpekar
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Victoria Sook Keng Tung
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - A. Iván Hernández
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Oleg V. Evgrafov
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Juan Marcos Alarcon
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
40
|
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
|
41
|
Xue J, Brawner AT, Thompson JR, Yelhekar TD, Newmaster KT, Qiu Q, Cooper YA, Yu CR, Ahmed-Braima YH, Kim Y, Lin Y. Spatiotemporal Mapping and Molecular Basis of Whole-brain Circuit Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.572456. [PMID: 38260331 PMCID: PMC10802351 DOI: 10.1101/2024.01.03.572456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Brain development is highly dynamic and asynchronous, marked by the sequential maturation of functional circuits across the brain. The timing and mechanisms driving circuit maturation remain elusive due to an inability to identify and map maturing neuronal populations. Here we create DevATLAS (Developmental Activation Timing-based Longitudinal Acquisition System) to overcome this obstacle. We develop whole-brain mapping methods to construct the first longitudinal, spatiotemporal map of circuit maturation in early postnatal mouse brains. Moreover, we uncover dramatic impairments within the deep cortical layers in a neurodevelopmental disorders (NDDs) model, demonstrating the utility of this resource to pinpoint when and where circuit maturation is disrupted. Using DevATLAS, we reveal that early experiences accelerate the development of hippocampus-dependent learning by increasing the synaptically mature granule cell population in the dentate gyrus. Finally, DevATLAS enables the discovery of molecular mechanisms driving activity-dependent circuit maturation.
Collapse
Affiliation(s)
- Jian Xue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew T. Brawner
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Equal contribution
| | - Jacqueline R. Thompson
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Equal contribution
| | - Tushar D. Yelhekar
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kyra T. Newmaster
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Qiang Qiu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, MO 66160, USA
| | - Yonatan A. Cooper
- Current address: Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, MO 66160, USA
| | | | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Yingxi Lin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Lead contact
| |
Collapse
|
42
|
Jung H, Han D, Lee C, Kaang BK. Synaptic Engram. ADVANCES IN NEUROBIOLOGY 2024; 38:131-145. [PMID: 39008014 DOI: 10.1007/978-3-031-62983-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The concept of the engram refers to structural and/or physiological changes that underlie memory associations during learning. However, the precise biological basis of the engram remains elusive, with ongoing controversy regarding whether it resides at the cellular level or within the synaptic connections between activated cells. Here, we briefly review the studies investigating the cellular engram and the challenges they encounter. Subsequently, we delve into the biological basis of the engram within synaptic connections. In this regard, we introduce the history of synaptic engrams and discuss recent findings suggesting that synaptic plasticity serves as a substrate for memory. Additionally, we provide an overview of key technologies utilized in the study of synaptic plasticity.
Collapse
Affiliation(s)
- Hyunsu Jung
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon, South Korea
- Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Daehee Han
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon, South Korea
- Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Chaery Lee
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Bong-Kiun Kaang
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon, South Korea.
- Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, South Korea.
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
43
|
Yelhekar TD, Meng M, Doupe J, Lin Y. All IEGs Are Not Created Equal-Molecular Sorting Within the Memory Engram. ADVANCES IN NEUROBIOLOGY 2024; 38:81-109. [PMID: 39008012 DOI: 10.1007/978-3-031-62983-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
When neurons are recruited to form the memory engram, they are driven to activate the expression of a series of immediate-early genes (IEGs). While these IEGs have been used relatively indiscriminately to identify the so-called engram neurons, recent research has demonstrated that different IEG ensembles can be physically and functionally distinct within the memory engram. This inherent heterogeneity of the memory engram is driven by the diversity in the functions and distributions of different IEGs. This process, which we call molecular sorting, is analogous to sorting the entire population of engram neurons into different sub-engrams molecularly defined by different IEGs. In this chapter, we will describe the molecular sorting process by systematically reviewing published work on engram ensemble cells defined by the following four major IEGs: Fos, Npas4, Arc, and Egr1. By comparing and contrasting these likely different components of the memory engram, we hope to gain a better understanding of the logic and significance behind the molecular sorting process for memory functions.
Collapse
Affiliation(s)
- Tushar D Yelhekar
- Department of Psychiatry, O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meizhen Meng
- Department of Psychiatry, O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Neuroscience Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joslyn Doupe
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yingxi Lin
- Department of Psychiatry, O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
44
|
Fuentes-Ramos M, Barco Á. Unveiling Transcriptional and Epigenetic Mechanisms Within Engram Cells: Insights into Memory Formation and Stability. ADVANCES IN NEUROBIOLOGY 2024; 38:111-129. [PMID: 39008013 DOI: 10.1007/978-3-031-62983-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Memory traces for behavioral experiences, such as fear conditioning or taste aversion, are believed to be stored through biophysical and molecular changes in distributed neuronal ensembles across various brain regions. These ensembles are known as engrams, and the cells that constitute them are referred to as engram cells. Recent advancements in techniques for labeling and manipulating neural activity have facilitated the study of engram cells throughout different memory phases, including acquisition, allocation, long-term storage, retrieval, and erasure. In this chapter, we will explore the application of next-generation sequencing methods to engram research, shedding new light on the contribution of transcriptional and epigenetic mechanisms to engram formation and stability.
Collapse
Affiliation(s)
- Miguel Fuentes-Ramos
- Instituto de Neurociencias, Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas, Alicante, Spain
| | - Ángel Barco
- Instituto de Neurociencias, Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas, Alicante, Spain.
| |
Collapse
|
45
|
Kitamura T, Ramesh K, Terranova JI. Understanding Others' Distress Through Past Experiences: The Role of Memory Engram Cells in Observational Fear. ADVANCES IN NEUROBIOLOGY 2024; 38:215-234. [PMID: 39008018 DOI: 10.1007/978-3-031-62983-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
For individuals to survive and function in society, it is essential that they recognize, interact with, and learn from other conspecifics. Observational fear (OF) is the well-conserved empathic ability of individuals to understand the other's aversive situation. While it is widely known that factors such as prior similar aversive experience and social familiarity with the demonstrator facilitate OF, the neural circuit mechanisms that explicitly regulate experience-dependent OF (Exp OF) were unclear. In this review, we examine the neural circuit mechanisms that regulate OF, with an emphasis on rodent models, and then discuss emerging evidence for the role of fear memory engram cells in the regulation of Exp OF. First, we examine the neural circuit mechanisms that underlie Naive OF, which is when an observer lacks prior experiences relevant to OF. In particular, the anterior cingulate cortex to basolateral amygdala (BLA) neural circuit is essential for Naive OF. Next, we discuss a recent study that developed a behavioral paradigm in mice to examine the neural circuit mechanisms that underlie Exp OF. This study found that fear memory engram cells in the BLA of observers, which form during a prior similar aversive experience with shock, are reactivated by ventral hippocampal neurons in response to shock delivery to the familiar demonstrator to elicit Exp OF. Finally, we discuss the implications of fear memory engram cells in Exp OF and directions of future research that are of both translational and basic interest.
Collapse
Affiliation(s)
- Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Kritika Ramesh
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
46
|
O'Sullivan FM, Ryan TJ. If Engrams Are the Answer, What Is the Question? ADVANCES IN NEUROBIOLOGY 2024; 38:273-302. [PMID: 39008021 DOI: 10.1007/978-3-031-62983-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Engram labelling and manipulation methodologies are now a staple of contemporary neuroscientific practice, giving the impression that the physical basis of engrams has been discovered. Despite enormous progress, engrams have not been clearly identified, and it is unclear what they should look like. There is an epistemic bias in engram neuroscience toward characterizing biological changes while neglecting the development of theory. However, the tools of engram biology are exciting precisely because they are not just an incremental step forward in understanding the mechanisms of plasticity and learning but because they can be leveraged to inform theory on one of the fundamental mysteries in neuroscience-how and in what format the brain stores information. We do not propose such a theory here, as we first require an appreciation for what is lacking. We outline a selection of issues in four sections from theoretical biology and philosophy that engram biology and systems neuroscience generally should engage with in order to construct useful future theoretical frameworks. Specifically, what is it that engrams are supposed to explain? How do the different building blocks of the brain-wide engram come together? What exactly are these component parts? And what information do they carry, if they carry anything at all? Asking these questions is not purely the privilege of philosophy but a key to informing scientific hypotheses that make the most of the experimental tools at our disposal. The risk for not engaging with these issues is high. Without a theory of what engrams are, what they do, and the wider computational processes they fit into, we may never know when they have been found.
Collapse
Affiliation(s)
- Fionn M O'Sullivan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland.
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia.
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, Canada.
| |
Collapse
|
47
|
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
|
48
|
Li R, Huang J, Li L, Zhao Z, Liang S, Liang S, Wang M, Liao X, Lyu J, Zhou Z, Wang S, Jin W, Chen H, Holder D, Liu H, Zhang J, Li M, Tang Y, Remy S, Pakan JMP, Chen X, Jia H. Holistic bursting cells store long-term memory in auditory cortex. Nat Commun 2023; 14:8090. [PMID: 38062015 PMCID: PMC10703882 DOI: 10.1038/s41467-023-43620-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
The sensory neocortex has been suggested to be a substrate for long-term memory storage, yet which exact single cells could be specific candidates underlying such long-term memory storage remained neither known nor visible for over a century. Here, using a combination of day-by-day two-photon Ca2+ imaging and targeted single-cell loose-patch recording in an auditory associative learning paradigm with composite sounds in male mice, we reveal sparsely distributed neurons in layer 2/3 of auditory cortex emerged step-wise from quiescence into bursting mode, which then invariably expressed holistic information of the learned composite sounds, referred to as holistic bursting (HB) cells. Notably, it was not shuffled populations but the same sparse HB cells that embodied the behavioral relevance of the learned composite sounds, pinpointing HB cells as physiologically-defined single-cell candidates of an engram underlying long-term memory storage in auditory cortex.
Collapse
Affiliation(s)
- Ruijie Li
- Advanced Institute for Brain and Intelligence and School of Physical Science and Technology, Guangxi University, Nanning, 530004, China
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Junjie Huang
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China
- Leibniz Institute for Neurobiology (LIN), 39118, Magdeburg, Germany
| | - Longhui Li
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Zhikai Zhao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Susu Liang
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Shanshan Liang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Meng Wang
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Jing Lyu
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Zhenqiao Zhou
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Sibo Wang
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Wenjun Jin
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, 400038, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China
| | - Haiyang Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Damaris Holder
- Leibniz Institute for Neurobiology (LIN), 39118, Magdeburg, Germany
| | - Hongbang Liu
- Advanced Institute for Brain and Intelligence and School of Physical Science and Technology, Guangxi University, Nanning, 530004, China
| | - Jianxiong Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Min Li
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Yuguo Tang
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Stefan Remy
- Leibniz Institute for Neurobiology (LIN), 39118, Magdeburg, Germany
- Center for Behavioral and Brain Science (CBBS), Otto von Guericke University, 39120, Magdeburg, Germany
| | - Janelle M P Pakan
- Center for Behavioral and Brain Science (CBBS), Otto von Guericke University, 39120, Magdeburg, Germany.
- Institute for Cognitive Neurology and Dementia Research, Otto von Guericke University, 39120, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany.
| | - Xiaowei Chen
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, 400038, China.
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
| | - Hongbo Jia
- Advanced Institute for Brain and Intelligence and School of Physical Science and Technology, Guangxi University, Nanning, 530004, China.
- Leibniz Institute for Neurobiology (LIN), 39118, Magdeburg, Germany.
- Brain Research Instrument Innovation Center, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China.
- Institute of Neuroscience and the SyNergy Cluster, Technical University of Munich, 80802, Munich, Germany.
| |
Collapse
|
49
|
Grieco SF, Johnston KG, Gao P, Garduño BM, Tang B, Yi E, Sun Y, Horwitz GD, Yu Z, Holmes TC, Xu X. Anatomical and molecular characterization of parvalbumin-cholecystokinin co-expressing inhibitory interneurons: implications for neuropsychiatric conditions. Mol Psychiatry 2023; 28:5293-5308. [PMID: 37443194 DOI: 10.1038/s41380-023-02153-5] [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: 03/17/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023]
Abstract
Inhibitory interneurons are crucial to brain function and their dysfunction is implicated in neuropsychiatric conditions. Emerging evidence indicates that cholecystokinin (CCK)-expressing interneurons (CCK+) are highly heterogenous. We find that a large subset of parvalbumin-expressing (PV+) interneurons express CCK strongly; between 40 and 56% of PV+ interneurons in mouse hippocampal CA1 express CCK. Primate interneurons also exhibit substantial PV/CCK co-expression. Mouse PV+/CCK+ and PV+/CCK- cells show distinguishable electrophysiological and molecular characteristics. Analysis of single nuclei RNA-seq and ATAC-seq data shows that PV+/CCK+ cells are a subset of PV+ cells, not of synuclein gamma positive (SNCG+) cells, and that they strongly express oxidative phosphorylation (OXPHOS) genes. We find that mitochondrial complex I and IV-associated OXPHOS gene expression is strongly correlated with CCK expression in PV+ interneurons at both the transcriptomic and protein levels. Both PV+ interneurons and dysregulation of OXPHOS processes are implicated in neuropsychiatric conditions, including autism spectrum (ASD) disorder and schizophrenia (SCZ). Analysis of human brain samples from patients with these conditions shows alterations in OXPHOS gene expression. Together these data reveal important molecular characteristics of PV-CCK co-expressing interneurons and support their implication in neuropsychiatric conditions.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department of Mathematics, School of Physical Sciences, University of California, Irvine, CA, 92697, USA
| | - Pan Gao
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - B Maximiliano Garduño
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Bryan Tang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Elsie Yi
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Yanjun Sun
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Gregory D Horwitz
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA
| | - Zhaoxia Yu
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Department of Statistics, Donald Bren School of Information and Computer Sciences, University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
- Department of Computer Science, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
50
|
Zhou X, Wu X, Wu Y, Yang L, Shi E, Ding W, Chen L, Shi X, Feng X, Su C, You Z, Xia J, Chen C, Yeliseyev V, Bry L, Xia S, Huang P, Meng J, Houle T, Akeju O, Mao J, Gerszten R, Chen Q, Xie Z, Shen S. Indole-3-Propionic Acid, a Gut Microbiota Metabolite, Protects Against the Development of Postoperative Delirium. Ann Surg 2023; 278:e1164-e1174. [PMID: 37185230 PMCID: PMC10603211 DOI: 10.1097/sla.0000000000005886] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
OBJECTIVE The aim was to determine preoperative gut microbiota metabolites that may be associated with postoperative delirium (POD) development in patients and further study in rodents. SUMMARY BACKGROUND DATA POD occurs in 9% to 50% of older patients undergoing anesthesia/surgery but lacks effective treatments or prevention. High-throughput metabolomics using liquid chromatography with tandem mass spectrometry has accelerated disease-related biomarkers discovery. We performed metabolomic studies in humans to identify potential metabolite biomarkers linked to POD and examined potential mechanisms in rodents. METHODS We performed a prospective observational cohort study to examine the metabolomic changes that were associated with the development of POD. Then the gut microbiota-related metabolomic changes were recapitulated by gut microbiota perturbation in rodents. POD was assessed in mice using a battery of behavioral tests including novel objective test, Y-maze test, open-field test, and buried food test. The mechanisms through which gut microbiota-related metabolomic changes influenced POD were examined using chemogenetics. RESULTS Indole-3-propionic acid (IPA) is a gut microbiota metabolite that belongs to the indole family. Baseline plasma levels of IPA were significantly inversely correlated with the onset of POD in 103 (17 cases) human individuals. This relationship was validated in preclinical mouse models for POD: reducing IPA levels through gut microbiota perturbation promoted POD-like behavior. More importantly, IPA administration deterred POD-like behavior. Colonization of germ-free mice with mutant Clostridium sporogenes that did not produce IPA-promoted POD-like behavior. Chemogenetic studies revealed that the protective effect of IPA in mice was mediated, in part, by peroxisome proliferator-activated receptor gamma coactivator 1-alpha in hippocampal interneurons. CONCLUSIONS Gut microbiota-derived IPA is an important molecule implicated in the pathogenesis of POD, which could potentially be harnessed for POD prevention.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Xinbo Wu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Orthopedics, Shanghai Tenth Hospital, Tongji University School of Medicine, Shanghai
| | - Yan Wu
- Department of Anesthesiology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liuyue Yang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Eleanor Shi
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Weihua Ding
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Liang Chen
- Center for Discovery and Innovation, Hackensack Health Care, Nutley, NJ
| | - Xu Shi
- Department of Cardiovascular Medicine, Beth Israel Deaconess Medical Center
| | - Xia Feng
- Department of Anesthesiology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chienwen Su
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Zerong You
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jianguo Xia
- Department of Parasitology, McGill University, Montreal, Canada
| | - Cynthia Chen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Lynn Bry
- Department of Pathology, Brigham and Women’s Hospital
| | - Suyun Xia
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Peigen Huang
- The Steele Lab, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jiawei Meng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Timothy Houle
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jianren Mao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Robert Gerszten
- Department of Cardiovascular Medicine, Beth Israel Deaconess Medical Center
| | - Qian Chen
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|