1
|
Subramanian R, Bauman A, Carpenter O, Cho C, Coste G, Dam A, Drake K, Ehnstrom S, Fitzgerald N, Jenkins A, Koolpe H, Liu R, Paserman T, Petersen D, Chavez DS, Rozental S, Thompson H, Tsukuda T, Zweig S, Gall M, Zupan B, Bergstrom H. An infralimbic cortex neuronal ensemble encoded during learning attenuates fear generalization expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.18.608308. [PMID: 39229064 PMCID: PMC11370439 DOI: 10.1101/2024.08.18.608308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Generalization allows previous experience to adaptively guide behavior when conditions change. The infralimbic (IL) subregion of the ventral medial prefrontal cortex plays a known role in generalization processes, although mechanisms remain unclear. A basic physical unit of memory storage and expression in the brain are sparse, distributed groups of neurons known as ensembles (i.e., the engram). Here, we set out to determine whether neuronal ensembles established in the IL during learning contribute to generalized responses. Generalization was tested in male and female mice by presenting a novel, ambiguous, tone generalization stimulus following Pavlovian defensive (fear) conditioning. The first experiment was designed to test a role for IL in generalization using chemogenetic manipulations. Results show IL regulates defensive behavior in response to ambiguous stimuli. IL silencing led to a switch in defensive state, from vigilant scanning to generalized freezing, while IL stimulation reduced freezing in favor of scanning. Leveraging activity-dependent "tagging" technology (ArcCreER T2 x eYFP system), a neuronal ensemble, preferentially located in IL Layer 2/3, was associated with the generalization stimulus. Remarkably, in the identical discrete location, fewer reactivated neurons were associated with the generalization stimulus at the remote timepoint (30 days) following learning. When an IL neuronal ensemble established during learning was selectively chemogenetically silenced, generalization increased. Conversely, IL neuronal ensemble stimulation reduced generalization. Overall, these data identify a crucial role for IL in suppressing generalized responses. Further, an IL neuronal ensemble, formed during learning, functions to later attenuate the expression of generalization in the presence of ambiguous threat stimuli.
Collapse
|
2
|
Hille M, Kühn S, Kempermann G, Bonhoeffer T, Lindenberger U. From animal models to human individuality: Integrative approaches to the study of brain plasticity. Neuron 2024; 112:3522-3541. [PMID: 39461332 DOI: 10.1016/j.neuron.2024.10.006] [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: 04/29/2024] [Revised: 06/02/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024]
Abstract
Plasticity allows organisms to form lasting adaptive changes in neural structures in response to interactions with the environment. It serves both species-general functions and individualized skill acquisition. To better understand human plasticity, we need to strengthen the dialogue between human research and animal models. Therefore, we propose to (1) enhance the interpretability of macroscopic methods used in human research by complementing molecular and fine-structural measures used in animals with such macroscopic methods, preferably applied to the same animals, to create macroscopic metrics common to both examined species; (2) launch dedicated cross-species research programs, using either well-controlled experimental paradigms, such as motor skill acquisition, or more naturalistic environments, where individuals of either species are observed in their habitats; and (3) develop conceptual and computational models linking molecular and fine-structural events to phenomena accessible by macroscopic methods. In concert, these three component strategies can foster new insights into the nature of plastic change.
Collapse
Affiliation(s)
- Maike Hille
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany; Center for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany.
| | - Simone Kühn
- Center for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany; Clinic and Policlinic for Psychiatry and Psychotherapy, University Clinic Hamburg-Eppendorf, Hamburg, Germany; Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany, and London, UK
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Tobias Bonhoeffer
- Synapses-Circuits-Plasticity, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Ulman Lindenberger
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany; Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany, and London, UK.
| |
Collapse
|
3
|
Abstract
Memories are stored as ensembles of engram neurons and their successful recall involves the reactivation of these cellular networks. However, significant gaps remain in connecting these cell ensembles with the process of forgetting. Here, we utilized a mouse model of object memory and investigated the conditions in which a memory could be preserved, retrieved, or forgotten. Direct modulation of engram activity via optogenetic stimulation or inhibition either facilitated or prevented the recall of an object memory. In addition, through behavioral and pharmacological interventions, we successfully prevented or accelerated forgetting of an object memory. Finally, we showed that these results can be explained by a computational model in which engrams that are subjectively less relevant for adaptive behavior are more likely to be forgotten. Together, these findings suggest that forgetting may be an adaptive form of engram plasticity which allows engrams to switch from an accessible state to an inaccessible state.
Collapse
Affiliation(s)
- James D O'Leary
- School of Biochemistry and Immunology, Trinity College DublinDublinIreland
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
| | - Rasmus Bruckner
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
- Department of Education and Psychology, Freie Universität BerlinBerlinGermany
- Max Planck Research Group NeuroCode, Max Planck Institute for Human DevelopmentBerlinGermany
| | - Livia Autore
- School of Biochemistry and Immunology, Trinity College DublinDublinIreland
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College DublinDublinIreland
- Trinity College Institute of Neuroscience, Trinity College DublinDublinIreland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of MelbourneMelbourneAustralia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR)TorontoCanada
| |
Collapse
|
4
|
Zaki Y, Cai DJ. Memory engram stability and flexibility. Neuropsychopharmacology 2024; 50:285-293. [PMID: 39300271 PMCID: PMC11525749 DOI: 10.1038/s41386-024-01979-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/22/2024]
Abstract
Many studies have shown that memories are encoded in sparse neural ensembles distributed across the brain. During the post-encoding period, often during sleep, many of the cells that were active during encoding are reactivated, supporting consolidation of this memory. During memory recall, many of the same cells that were active during encoding and reactivated during consolidation are reactivated during recall. These ensembles of cells have been referred to as the memory engram cells, stably representing a specific memory. However, recent studies question the rigidity of the "stable memory engram." Here we review the past literature of how episodic-like memories are encoded, consolidated, and recalled. We also highlight more recent studies (as well as some older literature) that suggest that these stable memories and their representations are much more dynamic and flexible than previously thought. We highlight some of these processes, including memory updating, reconsolidation, forgetting, schema learning, memory-linking, and representational drift.
Collapse
Affiliation(s)
- Yosif Zaki
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Denise J Cai
- Nash Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
5
|
Fujikawa R, Ramsaran AI, Guskjolen A, de la Parra J, Zou Y, Mocle AJ, Josselyn SA, Frankland PW. Neurogenesis-dependent remodeling of hippocampal circuits reduces PTSD-like behaviors in adult mice. Mol Psychiatry 2024; 29:3316-3329. [PMID: 38719894 DOI: 10.1038/s41380-024-02585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 11/08/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a hypermnesic condition that develops in a subset of individuals following exposure to severe trauma. PTSD symptoms are debilitating, and include increased anxiety, abnormal threat generalization, and impaired extinction. In developing treatment strategies for PTSD, preclinical studies in rodents have largely focused on interventions that target post-encoding memory processes such as reconsolidation and extinction. Instead, here we focus on forgetting, another post-encoding process that regulates memory expression. Using a double trauma murine model for PTSD, we asked whether promoting neurogenesis-mediated forgetting can weaken trauma memories and associated PTSD-relevant behavioral phenotypes. In the double trauma paradigm, consecutive aversive experiences lead to a constellation of behavioral phenotypes associated with PTSD including increases in anxiety-like behavior, abnormal threat generalization, and deficient extinction. We found that post-training interventions that elevate hippocampal neurogenesis weakened the original trauma memory and decreased these PTSD-relevant phenotypes. These effects were observed using multiple methods to manipulate hippocampal neurogenesis, including interventions restricted to neural progenitor cells that selectively promoted integration of adult-generated granule cells into hippocampal circuits. The same interventions also weakened cocaine place preference memories, suggesting that promoting hippocampal neurogenesis may represent a broadly useful approach in hypermnesic conditions such as PTSD and substance abuse disorders.
Collapse
Affiliation(s)
- Risako Fujikawa
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Adam I Ramsaran
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Axel Guskjolen
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Juan de la Parra
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Yi Zou
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Andrew J Mocle
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Child & Brain Development Program, Canadian Institute for Advanced Research, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
6
|
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:S0896-6273(24)00660-3. [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] [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
|
7
|
Howe ML. Early Childhood Memories Are not Repressed: Either They Were Never Formed or Were Quickly Forgotten. Top Cogn Sci 2024; 16:707-717. [PMID: 36458626 DOI: 10.1111/tops.12636] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/11/2022] [Accepted: 07/11/2022] [Indexed: 12/03/2022]
Abstract
Early childhood events are rarely remembered in adulthood. In fact, memory for these early experiences declines during childhood itself. This holds regardless of whether these memories of autobiographical experiences are traumatic or mundane, everyday experiences. Indeed, what people tend to remember from their childhoods involves relatively innocuous experiences, ones often devoid of emotion. In this article, I provide an overview of the types of memories adults recall from their childhoods and the ages at which these memories are believed to have been formed. Along the way, I provide a brief exegesis of the neurobiological and cognitive underpinnings of early memory development. I will show that changes and growth in neural interconnectivity as well as the development of various cognitive structures (e.g., the inception of the cognitive self) help propel the emergence of a mature autobiographical memory system, one that can and does serve as a reconstructive base for remembering events that occur in later childhood and adulthood. During the course of this review, I detail the nature of early memories, their fragility, and the adaptive consequences of forgetting and supplanting these memories with newer, more age-appropriate experiences throughout childhood.
Collapse
Affiliation(s)
- Mark L Howe
- Department of Psychology, School of Health and Psychological Sciences, City, University of London
| |
Collapse
|
8
|
Sanguino-Gómez J, Huijgens S, den Hartog M, Schenk IJM, Kluck W, Versluis TD, Krugers HJ. Neural correlates of learning and memory are altered by early-life stress. Neurobiol Learn Mem 2024; 213:107952. [PMID: 38906243 DOI: 10.1016/j.nlm.2024.107952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024]
Abstract
The ability to learn and remember, which is fundamental for behavioral adaptation, is susceptible to stressful experiences during the early postnatal period, such as abnormal levels of maternal care. The exact mechanisms underlying these effects still remain elusive. This study examined whether early life stress (ELS) alters memory and brain activation patterns in male mice. Therefore, we examined the expression of the immediate early genes (IEGs) c-Fos and Arc in the dentate gyrus (DG) and basolateral amygdala (BLA) after training and memory retrieval in a fear conditioning task. Furthermore, we examined the potential of RU38486 (RU486), a glucocorticoid receptor antagonist, to mitigate ELS-induced memory deficits by blocking stress signalling during adolescence. Arc::dVenus reporter mice, which allow investigating experience-dependent expression of the immediate early gene Arc also at more remote time points, were exposed to ELS by housing dams and offspring with limited bedding and nesting material (LBN) between postnatal days (PND) 2-9 and trained in a fear conditioning task at adult age. We found that ELS reduced both fear acquisition and contextual memory retrieval. RU486 did not prevent these effects. ELS reduced the number of Arc::dVenus+ cells in DG and BLA after training, while the number of c-Fos+ cells were left unaffected. After memory retrieval, ELS decreased c-Fos+ cells in the ventral DG and BLA. ELS also altered the colocalization of c-Fos+ cells with Arc::dVenus+ cells in the ventral DG, possibly indicating impaired engram allocation in the ventral DG after memory retrieval. In conclusion, this study shows that ELS alters neuronal activation patterns after fear acquisition and retrieval, which may provide mechanistic insights into enduring impact of ELS on the processing of fear memories, possibly via changes in cell (co-) activation and engram cell allocation.
Collapse
Affiliation(s)
| | - Stefan Huijgens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Maxine den Hartog
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Inim J M Schenk
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Wenya Kluck
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamara D Versluis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Chen R, Wang Z, Lin Q, Hou X, Jiang Y, Le Q, Liu X, Ma L, Wang F. Destabilization of fear memory by Rac1-driven engram-microglia communication in hippocampus. Brain Behav Immun 2024; 119:621-636. [PMID: 38670239 DOI: 10.1016/j.bbi.2024.04.024] [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: 09/21/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024] Open
Abstract
Rac1 is a key regulator of the cytoskeleton and neuronal plasticity, and is known to play a critical role in psychological and cognitive brain disorders. To elucidate the engram specific Rac1 signaling in fear memory, a doxycycline (Dox)-dependent robust activity marking (RAM) system was used to label dorsal dentate gyrus (DG) engram cells in mice during contextual fear conditioning. Rac1 mRNA and protein levels in DG engram cells were peaked at 24 h (day 1) after fear conditioning and were more abundant in the fear engram cells than in the non-engram cells. Optogenetic activation of Rac1 in a temporal manner in DG engram cells before memory retrieval decreased the freezing level in the fear context. Optogenetic activation of Rac1 increased autophagy protein 7 (ATG7) expression in the DG engram cells and activated DG microglia. Microglia-specific transcriptomics and fluorescence in situ hybridization revealed that overexpression of ATG7 in the fear engram cells upregulated the mRNA of Toll-like receptor TLR2/4 in DG microglia. Knockdown of microglial TLR2/4 rescued fear memory destabilization induced by ATG7 overexpression or Rac1 activation in DG engram cells. These results indicate that Rac1-driven communications between engram cells and microglia contributes to contextual fear memory destabilization, and is mediated by ATG7 and TLR2/4, and suggest a novel mechanistic framework for the cytoskeletal regulator in fear memory interference.
Collapse
Affiliation(s)
- Ruyan Chen
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Zhilin Wang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qing Lin
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Xutian Hou
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Yan Jiang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qiumin Le
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Xing Liu
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Lan Ma
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Feifei Wang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China.
| |
Collapse
|
10
|
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
|
11
|
Lazarov O, Gupta M, Kumar P, Morrissey Z, Phan T. Memory circuits in dementia: The engram, hippocampal neurogenesis and Alzheimer's disease. Prog Neurobiol 2024; 236:102601. [PMID: 38570083 PMCID: PMC11221328 DOI: 10.1016/j.pneurobio.2024.102601] [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: 08/10/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Here, we provide an in-depth consideration of our current understanding of engrams, spanning from molecular to network levels, and hippocampal neurogenesis, in health and Alzheimer's disease (AD). This review highlights novel findings in these emerging research fields and future research directions for novel therapeutic avenues for memory failure in dementia. Engrams, memory in AD, and hippocampal neurogenesis have each been extensively studied. The integration of these topics, however, has been relatively less deliberated, and is the focus of this review. We primarily focus on the dentate gyrus (DG) of the hippocampus, which is a key area of episodic memory formation. Episodic memory is significantly impaired in AD, and is also the site of adult hippocampal neurogenesis. Advancements in technology, especially opto- and chemogenetics, have made sophisticated manipulations of engram cells possible. Furthermore, innovative methods have emerged for monitoring neurons, even specific neuronal populations, in vivo while animals engage in tasks, such as calcium imaging. In vivo calcium imaging contributes to a more comprehensive understanding of engram cells. Critically, studies of the engram in the DG using these technologies have shown the important contribution of hippocampal neurogenesis for memory in both health and AD. Together, the discussion of these topics provides a holistic perspective that motivates questions for future research.
Collapse
Affiliation(s)
- Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Pavan Kumar
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zachery Morrissey
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Trongha Phan
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
12
|
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
|
13
|
Calin-Jageman RJ, Gonzalez Delgadillo B, Gamino E, Juarez Z, Kurkowski A, Musajeva N, Valdez L, Wittrock D, Wilsterman T, Zarate Torres J, Calin-Jageman IE. Evidence of Active-Forgetting Mechanisms? Blocking Arachidonic Acid Release May Slow Forgetting of Sensitization in Aplysia. eNeuro 2024; 11:ENEURO.0516-23.2024. [PMID: 38538086 PMCID: PMC10999730 DOI: 10.1523/eneuro.0516-23.2024] [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: 12/07/2023] [Revised: 01/14/2024] [Accepted: 02/02/2024] [Indexed: 04/07/2024] Open
Abstract
Long-term sensitization in Aplysia is accompanied by a persistent up-regulation of mRNA encoding the peptide neurotransmitter Phe-Met-Arg-Phe-amide (FMRFa), a neuromodulator that opposes the expression of sensitization through activation of the arachidonic acid second-messenger pathway. We completed a preregistered test of the hypothesis that FMRFa plays a critical role in the forgetting of sensitization. Aplysia received long-term sensitization training and were then given whole-body injections of vehicle (N = 27), FMRFa (N = 26), or 4-bromophenacylbromide (4-BPB; N = 31), a phospholipase inhibitor that prevents the release of arachidonic acid. FMRFa produced no changes in forgetting. 4-BPB decreased forgetting measured 6 d after training [d s = 0.55 95% CI(0.01, 1.09)], though the estimated effect size is uncertain. Our results provide preliminary evidence that forgetting of sensitization may be a regulated, active process in Aplysia, but could also indicate a role for arachidonic acid in stabilizing the induction of sensitization.
Collapse
Affiliation(s)
| | | | - Elise Gamino
- Neuroscience Program, Dominican University, River Forest, Illinois 60305
| | - Zayra Juarez
- Neuroscience Program, Dominican University, River Forest, Illinois 60305
| | - Anna Kurkowski
- Neuroscience Program, Dominican University, River Forest, Illinois 60305
| | - Nelly Musajeva
- Neuroscience Program, Dominican University, River Forest, Illinois 60305
| | - Leslie Valdez
- Neuroscience Program, Dominican University, River Forest, Illinois 60305
| | - Diana Wittrock
- Neuroscience Program, Dominican University, River Forest, Illinois 60305
| | - Theresa Wilsterman
- Neuroscience Program, Dominican University, River Forest, Illinois 60305
| | | | | |
Collapse
|
14
|
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
|
15
|
Liu X, Lu T, Chen X, Huang S, Zheng W, Zhang W, Meng S, Yan W, Shi L, Bao Y, Xue Y, Shi J, Yuan K, Han Y, Lu L. Memory consolidation drives the enhancement of remote cocaine memory via prefrontal circuit. Mol Psychiatry 2024; 29:730-741. [PMID: 38221548 DOI: 10.1038/s41380-023-02364-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024]
Abstract
Remote memory usually decreases over time, whereas remote drug-cue associated memory exhibits enhancement, increasing the risk of relapse during abstinence. Memory system consolidation is a prerequisite for remote memory formation, but neurobiological underpinnings of the role of consolidation in the enhancement of remote drug memory are unclear. Here, we found that remote cocaine-cue associated memory was enhanced in rats that underwent self-administration training, together with a progressive increase in the response of prelimbic cortex (PrL) CaMKII neurons to cues. System consolidation was required for the enhancement of remote cocaine memory through PrL CaMKII neurons during the early period post-training. Furthermore, dendritic spine maturation in the PrL relied on the basolateral amygdala (BLA) input during the early period of consolidation, contributing to remote memory enhancement. These findings indicate that memory consolidation drives the enhancement of remote cocaine memory through a time-dependent increase in activity and maturation of PrL CaMKII neurons receiving a sustained BLA input.
Collapse
Affiliation(s)
- Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Xuan Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
- Xinxiang Medical University, Xinxiang, 453003, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, 100191, Beijing, China
| | - Wei Zheng
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Wei Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Le Shi
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191, Beijing, China.
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No. 2018RU006), Dongcheng, Beijing, China.
| |
Collapse
|
16
|
Li H, Tamura R, Hayashi D, Asai H, Koga J, Ando S, Yokota S, Kaneko J, Sakurai K, Sumiyoshi A, Yamamoto T, Hikishima K, Tanaka KZ, McHugh TJ, Hisatsune T. Silencing dentate newborn neurons alters excitatory/inhibitory balance and impairs behavioral inhibition and flexibility. SCIENCE ADVANCES 2024; 10:eadk4741. [PMID: 38198539 PMCID: PMC10780870 DOI: 10.1126/sciadv.adk4741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
Adult neurogenesis confers the hippocampus with unparalleled neural plasticity, essential for intricate cognitive functions. The specific influence of sparse newborn neurons (NBNs) in modulating neural activities and subsequently steering behavior, however, remains obscure. Using an engineered NBN-tetanus toxin mouse model (NBN-TeTX), we noninvasively silenced NBNs, elucidating their crucial role in impulse inhibition and cognitive flexibility as evidenced through Morris water maze reversal learning and Go/Nogo task in operant learning. Task-based functional MRI (tb-fMRI) paired with operant learning revealed dorsal hippocampal hyperactivation during the Nogo task in male NBN-TeTX mice, suggesting that hippocampal hyperexcitability might underlie the observed behavioral deficits. Additionally, resting-state fMRI (rs-fMRI) exhibited enhanced functional connectivity between the dorsal and ventral dentate gyrus following NBN silencing. Further investigations into the activities of PV+ interneurons and mossy cells highlighted the indispensability of NBNs in maintaining the hippocampal excitation/inhibition balance. Our findings emphasize that the neural plasticity driven by NBNs extensively modulates the hippocampus, sculpting inhibitory control and cognitive flexibility.
Collapse
Affiliation(s)
- Haowei Li
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Risako Tamura
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Daiki Hayashi
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Hirotaka Asai
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Junya Koga
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Shota Ando
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Sayumi Yokota
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Jun Kaneko
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Keisuke Sakurai
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Akira Sumiyoshi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tadashi Yamamoto
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Keigo Hikishima
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kazumasa Z. Tanaka
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Thomas J. McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Tatsuhiro Hisatsune
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
17
|
Vasudevan K, Hassell JE, Maren S. Hippocampal Engrams and Contextual Memory. ADVANCES IN NEUROBIOLOGY 2024; 38:45-66. [PMID: 39008010 DOI: 10.1007/978-3-031-62983-9_4] [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
Memories are not formed in a vacuum and often include rich details about the time and place in which events occur. Contextual stimuli promote the retrieval of events that have previously occurred in the encoding context and limit the retrieval of context-inappropriate information. Contexts that are associated with traumatic or harmful events both directly elicit fear and serve as reminders of aversive events associated with trauma. It has long been appreciated that the hippocampus is involved in contextual learning and memory and is central to contextual fear conditioning. However, little is known about the underlying neuronal mechanisms underlying the encoding and retrieval of contextual fear memories. Recent advancements in neuronal labeling methods, including activity-dependent tagging of cellular ensembles encoding memory ("engrams"), provide unique insight into the neural substrates of memory in the hippocampus. Moreover, these methods allow for the selective manipulation of memory ensembles. Attenuating or erasing fear memories may have considerable therapeutic value for patients with post-traumatic stress disorder or other trauma- or stressor-related conditions. In this chapter, we review the role of the hippocampus in contextual fear conditioning in rodents and explore recent work implicating hippocampal ensembles in the encoding and retrieval of aversive memories.
Collapse
Affiliation(s)
- Krithika Vasudevan
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - James E Hassell
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
18
|
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
|
19
|
Ohno M. A Strategy for Allowing Earlier Diagnosis and Rigorous Evaluation of BACE1 Inhibitors in Preclinical Alzheimer's Disease. J Alzheimers Dis 2024; 99:431-445. [PMID: 38701146 DOI: 10.3233/jad-231451] [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] [Indexed: 05/05/2024]
Abstract
Given continued failure of BACE1 inhibitor programs at symptomatic and prodromal stages of Alzheimer's disease (AD), clinical trials need to target the earlier preclinical stage. However, trial design is complex in this population with negative diagnosis of classical hippocampal amnesia on standard memory tests. Besides recent advances in brain imaging, electroencephalogram, and fluid-based biomarkers, new cognitive markers should be established for earlier diagnosis that can optimize recruitment to BACE1 inhibitor trials in presymptomatic AD. Notably, accelerated long-term forgetting (ALF) is emerging as a sensitive cognitive measure that can discriminate between asymptomatic individuals with high risks for developing AD and healthy controls. ALF is a form of declarative memory impairment characterized by increased forgetting rates over longer delays (days to months) despite normal storage within the standard delays of testing (20-60 min). Therefore, ALF may represent a harbinger of preclinical dementia and the impairment of systems memory consolidation, during which memory traces temporarily stored in the hippocampus become gradually integrated into cortical networks. This review provides an overview of the utility of ALF in a rational design of next-generation BACE1 inhibitor trials in preclinical AD. I explore potential mechanisms underlying ALF and relevant early-stage biomarkers useful for BACE1 inhibitor evaluation, including synaptic protein alterations, astrocytic dysregulation and neuron hyperactivity in the hippocampal-cortical network. Furthermore, given the physiological role of the isoform BACE2 as an AD-suppressor gene, I also discuss the possible association between the poor selectivity of BACE1 inhibitors and their side effects (e.g., cognitive worsening) in prior clinical trials.
Collapse
Affiliation(s)
- Masuo Ohno
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Ortega-de San Luis C, Pezzoli M, Urrieta E, Ryan TJ. Engram cell connectivity as a mechanism for information encoding and memory function. Curr Biol 2023; 33:5368-5380.e5. [PMID: 37992719 DOI: 10.1016/j.cub.2023.10.074] [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: 06/19/2023] [Revised: 09/18/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023]
Abstract
Information derived from experiences is incorporated into the brain as changes to ensembles of cells, termed engram cells, which allow memory storage and recall. The mechanism by which those changes hold specific information is unclear. Here, we test the hypothesis that the specific synaptic wiring between engram cells is the substrate of information storage. First, we monitor how learning modifies the connectivity pattern between engram cells at a monosynaptic connection involving the hippocampal ventral CA1 (vCA1) region and the amygdala. Then, we assess the functional significance of these connectivity changes by artificially activating or inhibiting its presynaptic and postsynaptic components, respectively. Finally, we identify a synaptic plasticity mechanism mediated by postsynaptic density protein 95 (PSD-95), which impacts the connectivity pattern among engram cells and contributes to the long-term stability of the memory. These findings impact our theory of learning and memory by helping us explain the translation of specific information into engram cells and how these connections shape brain function.
Collapse
Affiliation(s)
- Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin D02 PN40, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Maurizio Pezzoli
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin D02 PN40, Ireland; Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Esteban Urrieta
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin D02 PN40, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin D02 PN40, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin D02 PN40, Ireland; Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC 3052, Australia; Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
23
|
Brosens N, Lesuis SL, Rao-Ruiz P, van den Oever MC, Krugers HJ. Shaping Memories Via Stress: A Synaptic Engram Perspective. Biol Psychiatry 2023:S0006-3223(23)01720-1. [PMID: 37977215 DOI: 10.1016/j.biopsych.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/09/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Stress modulates the activity of various memory systems and can thereby guide behavioral interaction with the environment in an adaptive or maladaptive manner. At the cellular level, a large body of evidence indicates that (nor)adrenaline and glucocorticoid release induced by acute stress exposure affects synapse function and synaptic plasticity, which are critical substrates for learning and memory. Recent evidence suggests that memories are supported in the brain by sparsely distributed neurons within networks, termed engram cell ensembles. While the physiological and molecular effects of stress on the synapse are increasingly well characterized, how these synaptic modifications shape the multiscale dynamics of engram cell ensembles is still poorly understood. In this review, we discuss and integrate recent information on how acute stress affects synapse function and how this may alter engram cell ensembles and their synaptic connectivity to shape memory strength and memory precision. We provide a mechanistic framework of a synaptic engram under stress and put forward outstanding questions that address knowledge gaps in our understanding of the mechanisms that underlie stress-induced memory modulation.
Collapse
Affiliation(s)
- Niek Brosens
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| | - Sylvie L Lesuis
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Cellular and Cognitive Neuroscience group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Priyanka Rao-Ruiz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 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, Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences-Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
24
|
Power SD, Stewart E, Zielke LG, Byrne EP, Douglas A, Ortega-de San Luis C, Lynch L, Ryan TJ. Immune activation state modulates infant engram expression across development. SCIENCE ADVANCES 2023; 9:eadg9921. [PMID: 37939176 PMCID: PMC10631722 DOI: 10.1126/sciadv.adg9921] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Infantile amnesia is possibly the most ubiquitous form of memory loss in mammals. We investigated how memories are stored in the brain throughout development by integrating engram labeling technology with mouse models of infantile amnesia. Here, we found a phenomenon in which male offspring in maternal immune activation models of autism spectrum disorder do not experience infantile amnesia. Maternal immune activation altered engram ensemble size and dendritic spine plasticity. We rescued the same apparently forgotten infantile memories in neurotypical mice by optogenetically reactivating dentate gyrus engram cells labeled during complex experiences in infancy. Furthermore, we permanently reinstated lost infantile memories by artificially updating the memory engram, demonstrating that infantile amnesia is a reversible process. Our findings suggest not only that infantile amnesia is due to a reversible retrieval deficit in engram expression but also that immune activation during development modulates innate, and reversible, forgetting switches that determine whether infantile amnesia will occur.
Collapse
Affiliation(s)
- Sarah D. Power
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Erika Stewart
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Louisa G. Zielke
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Eric P. Byrne
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Aaron Douglas
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomás J. Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for 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
|
25
|
Bianco R, Hall ET, Pearce MT, Chait M. Implicit auditory memory in older listeners: From encoding to 6-month retention. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100115. [PMID: 38020808 PMCID: PMC10663129 DOI: 10.1016/j.crneur.2023.100115] [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: 04/04/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Any listening task, from sound recognition to sound-based communication, rests on auditory memory which is known to decline in healthy ageing. However, how this decline maps onto multiple components and stages of auditory memory remains poorly characterised. In an online unsupervised longitudinal study, we tested ageing effects on implicit auditory memory for rapid tone patterns. The test required participants (younger, aged 20-30, and older adults aged 60-70) to quickly respond to rapid regularly repeating patterns emerging from random sequences. Patterns were novel in most trials (REGn), but unbeknownst to the participants, a few distinct patterns reoccurred identically throughout the sessions (REGr). After correcting for processing speed, the response times (RT) to REGn should reflect the information held in echoic and short-term memory before detecting the pattern; long-term memory formation and retention should be reflected by the RT advantage (RTA) to REGr vs REGn which is expected to grow with exposure. Older participants were slower than younger adults in detecting REGn and exhibited a smaller RTA to REGr. Computational simulations using a model of auditory sequence memory indicated that these effects reflect age-related limitations both in early and long-term memory stages. In contrast to ageing-related accelerated forgetting of verbal material, here older adults maintained stable memory traces for REGr patterns up to 6 months after the first exposure. The results demonstrate that ageing is associated with reduced short-term memory and long-term memory formation for tone patterns, but not with forgetting, even over surprisingly long timescales.
Collapse
Affiliation(s)
- Roberta Bianco
- Ear Institute, University College London, WC1X 8EE, London, United Kingdom
- Neuroscience of Perception and Action Laboratory, Italian Institute of Technology, 00161, Rome, Italy
| | - Edward T.R. Hall
- School of Electronic Engineering and Computer Science, Queen Mary University of London, E1 4NS, London, United Kingdom
| | - Marcus T. Pearce
- School of Electronic Engineering and Computer Science, Queen Mary University of London, E1 4NS, London, United Kingdom
- Department of Clinical Medicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Maria Chait
- Ear Institute, University College London, WC1X 8EE, London, United Kingdom
| |
Collapse
|
26
|
Wang CM, Wu CY, Lin CE, Hsu MC, Lin JC, Huang CC, Lien TY, Lin HK, Chang TW, Chiang HC. Forgotten memory storage and retrieval in Drosophila. Nat Commun 2023; 14:7153. [PMID: 37935667 PMCID: PMC10630420 DOI: 10.1038/s41467-023-42753-x] [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: 09/28/2022] [Accepted: 10/20/2023] [Indexed: 11/09/2023] Open
Abstract
Inaccessibility of stored memory in ensemble cells through the forgetting process causes animals to be unable to respond to natural recalling cues. While accumulating evidence has demonstrated that reactivating memory-stored cells can switch cells from an inaccessible state to an accessible form and lead to recall of previously learned information, the underlying cellular and molecular mechanisms remain elusive. The current study used Drosophila as a model to demonstrate that the memory of one-trial aversive olfactory conditioning, although inaccessible within a few hours after learning, is stored in KCαβ and retrievable after mild retraining. One-trial aversive conditioning triggers protein synthesis to form a long-lasting cellular memory trace, approximately 20 days, via creb in KCαβ, and a transient cellular memory trace, approximately one day, via orb in MBON-α3. PPL1-α3 negatively regulates forgotten one-trial conditioning memory retrieval. The current study demonstrated that KCαβ, PPL1-α3, and MBON-α3 collaboratively regulate the formation of forgotten one-cycle aversive conditioning memory formation and retrieval.
Collapse
Affiliation(s)
- Chih-Ming Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Chun-Yuan Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Chen-En Lin
- Department of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Ming-Chi Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Jing-Chun Lin
- Department of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Chuan-Chin Huang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Ting-Yu Lien
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Hsin-Kai Lin
- Department of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC
| | - Ting-Wei Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Hsueh-Cheng Chiang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC.
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan, Taiwan, ROC.
| |
Collapse
|
27
|
Luis CODS, Pezzoli M, Urrieta E, Ryan TJ. Engram cell connectivity as a mechanism for information encoding and memory function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558774. [PMID: 37790352 PMCID: PMC10542553 DOI: 10.1101/2023.09.21.558774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Information derived from experiences is incorporated into the brain as changes to ensembles of cells, termed engram cells, that allow memory storage and recall. The mechanism by which those changes hold specific information is unclear. Here we test the hypothesis that the specific synaptic wiring between engram cells is the substrate of information storage. First, we monitor how learning modifies the connectivity pattern between engram cells at a monosynaptic connection involving the hippocampal vCA1 region and the amygdala. Then, we assess the functional significance of these connectivity changes by artificially activating or inhibiting its presynaptic and postsynaptic components respectively. Finally, we identify a synaptic plasticity mechanism mediated by PSD-95, which impacts the connectivity pattern among engram cells and contributes to the long-term stability of the memory. These findings impact our theory of learning and memory by helping us explain the translation of specific information into engram cells and how these connections shape brain function.
Collapse
Affiliation(s)
- Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Maurizio Pezzoli
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin, Ireland
- Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Switzerland
| | - Esteban Urrieta
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College of 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, Victoria, Australia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
| |
Collapse
|
28
|
Robins S. The 21st century engram. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2023; 14:e1653. [PMID: 37177850 DOI: 10.1002/wcs.1653] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/10/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023]
Abstract
The search for the engram-the neural mechanism of memory-has been a guiding research project for neuroscience since its emergence as a distinct scientific field. Recent developments in the tools and techniques available for investigating the mechanisms of memory have allowed researchers to proclaimed the search is over. While there is ongoing debate about the justification for that claim, renewed interest in the engram is clear. This attention highlights the impoverished status of the engram concept. As research accelerates, the simple characterization of the engram as an enduring physical change is stretched thin. Now that the engram commitment has been made more explicit, it must also be made more precise. If the project of 20th century neurobiology was finding the engram, the project of the 21st must be supplying a richer account of what's been found. This paper sketches a history of the engram, and a way forward. This article is categorized under: Philosophy > Foundations of Cognitive Science.
Collapse
Affiliation(s)
- Sarah Robins
- Department of Philosophy, University of Kansas, Lawrence, Kansas, USA
| |
Collapse
|
29
|
Autore L, O'Leary JD, Ortega-de San Luis C, Ryan TJ. Adaptive expression of engrams by retroactive interference. Cell Rep 2023; 42:112999. [PMID: 37590145 DOI: 10.1016/j.celrep.2023.112999] [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: 03/03/2023] [Revised: 06/17/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
Long-term memories are stored as configurations of neuronal ensembles, termed engrams. Although investigation of engram cell properties and functionality in memory recall has been extensive, less is known about how engram cells are affected by forgetting. We describe a form of interference-based forgetting using an object memory behavioral paradigm. By using activity-dependent cell labeling, we show that although retroactive interference results in decreased engram cell reactivation during recall trials, optogenetic stimulation of the labeled engram cells is sufficient to induce memory retrieval. Forgotten engrams may be reinstated via the presentation of similar or related environmental information. Furthermore, we demonstrate that engram activity is necessary for interference to occur. Taken together, these findings indicate that retroactive interference modules engram expression in a manner that is both reversible and updatable. Inference may constitute a form of adaptive forgetting where, in everyday life, new perceptual and environmental inputs modulate the natural forgetting process.
Collapse
Affiliation(s)
- Livia Autore
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - James D O'Leary
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College of Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College of 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
|
30
|
Dalto JF, Medina JH. Time-dependent inhibition of Rac1 in the VTA enhances long-term aversive memory: implications in active forgetting mechanisms. Sci Rep 2023; 13:13507. [PMID: 37598223 PMCID: PMC10439914 DOI: 10.1038/s41598-023-40434-9] [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: 05/19/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
The fate of memories depends mainly on two opposing forces: the mechanisms required for the storage and maintenance of memory and the mechanisms underlying forgetting, being the latter much less understood. Here, we show the effect of inhibiting the small Rho GTPase Rac1 on the fate of inhibitory avoidance memory in male rats. The immediate post-training micro-infusion of the specific Rac1 inhibitor NSC23766 (150 ng/0.5 µl/ side) into the ventral tegmental area (VTA) enhanced long-term memory at 1, 7, and 14 days after a single training. Additionally, an opposed effect occurred when the inhibitor was infused at 12 h after training while no effect was observed immediately after testing animals at 1 day. Control experiments ruled out the possibility that post-training memory enhancement was due to facilitation of memory formation since no effect was found when animals were tested at 1 h after acquisition and no memory enhancement was observed after the formation of a weak memory. Immediate post-training micro-infusion of Rac1 inhibitor into the dorsal hippocampus, or the amygdala did not affect memory. Our findings support the idea of a Rac1-dependent time-specific active forgetting mechanism in the VTA controlling the strength of a long-term aversive memory.
Collapse
Affiliation(s)
- Juliana F Dalto
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo de Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155, 3rd Floor, C1121ABG, Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto de Biología Celular y Neurociencias "Prof. Eduardo de Robertis", Facultad de Medicina, Universidad de Buenos Aires-CONICET, Paraguay 2155, 3rd Floor, C1121ABG, Buenos Aires, Argentina.
- Instituto Tecnológico de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
31
|
Kozyrev SA, Solntseva SV, Storozheva ZI, Nikitin VP. Epigenetic Processes of DNA Methylation Are Selectively Involved in the Mechanisms of Retrograde and Anteograde Amnesia. Bull Exp Biol Med 2023; 175:427-432. [PMID: 37768459 DOI: 10.1007/s10517-023-05879-y] [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: 01/27/2023] [Indexed: 09/29/2023]
Abstract
The participation of DNA methylation processes in the mechanisms of anterograde and retrograde amnesia caused by impaired reconsolidation of conditioned food aversion memory by NMDA glutamate receptor antagonists or serotonin receptor antagonists, respectively, were studied on grape snails. Anterograde amnesia was characterized by impaired formation of long-term memory during repeated learning. Administration of a DNA methyltransferase (DNMT) inhibitor to amnestic animals resulted in accelerated formation of long-term memory during 1 day of repetitive training vs 3 days during initial training. In serotonin-dependent retrograde amnesia, repeated learning without DNMT inhibitor administration or after inhibitor injections led to the formation of long-term memory. The dynamics of memory formation was similar in both cases and did not differ from that during the initial training: the memory was formed within 3 days of training. Thus, epigenetic processes of DNA methylation are selectively involved in the mechanisms of anterograde amnesia, but do not participate in the mechanisms of retrograde amnesia.
Collapse
Affiliation(s)
- S A Kozyrev
- P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - S V Solntseva
- P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - Z I Storozheva
- P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - V P Nikitin
- P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.
| |
Collapse
|
32
|
Guskjolen A, Cembrowski MS. Engram neurons: Encoding, consolidation, retrieval, and forgetting of memory. Mol Psychiatry 2023; 28:3207-3219. [PMID: 37369721 PMCID: PMC10618102 DOI: 10.1038/s41380-023-02137-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
Tremendous strides have been made in our understanding of the neurobiological substrates of memory - the so-called memory "engram". Here, we integrate recent progress in the engram field to illustrate how engram neurons transform across the "lifespan" of a memory - from initial memory encoding, to consolidation and retrieval, and ultimately to forgetting. To do so, we first describe how cell-intrinsic properties shape the initial emergence of the engram at memory encoding. Second, we highlight how these encoding neurons preferentially participate in synaptic- and systems-level consolidation of memory. Third, we describe how these changes during encoding and consolidation guide neural reactivation during retrieval, and facilitate memory recall. Fourth, we describe neurobiological mechanisms of forgetting, and how these mechanisms can counteract engram properties established during memory encoding, consolidation, and retrieval. Motivated by recent experimental results across these four sections, we conclude by proposing some conceptual extensions to the traditional view of the engram, including broadening the view of cell-type participation within engrams and across memory stages. In collection, our review synthesizes general principles of the engram across memory stages, and describes future avenues to further understand the dynamic engram.
Collapse
Affiliation(s)
- Axel Guskjolen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Mark S Cembrowski
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
33
|
Borzello M, Ramirez S, Treves A, Lee I, Scharfman H, Stark C, Knierim JJ, Rangel LM. Assessments of dentate gyrus function: discoveries and debates. Nat Rev Neurosci 2023; 24:502-517. [PMID: 37316588 PMCID: PMC10529488 DOI: 10.1038/s41583-023-00710-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/16/2023]
Abstract
There has been considerable speculation regarding the function of the dentate gyrus (DG) - a subregion of the mammalian hippocampus - in learning and memory. In this Perspective article, we compare leading theories of DG function. We note that these theories all critically rely on the generation of distinct patterns of activity in the region to signal differences between experiences and to reduce interference between memories. However, these theories are divided by the roles they attribute to the DG during learning and recall and by the contributions they ascribe to specific inputs or cell types within the DG. These differences influence the information that the DG is thought to impart to downstream structures. We work towards a holistic view of the role of DG in learning and memory by first developing three critical questions to foster a dialogue between the leading theories. We then evaluate the extent to which previous studies address our questions, highlight remaining areas of conflict, and suggest future experiments to bridge these theories.
Collapse
Affiliation(s)
- Mia Borzello
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | | | - Inah Lee
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, South Korea
| | - Helen Scharfman
- Departments of Child and Adolescent Psychiatry, Neuroscience and Physiology and Psychiatry and the Neuroscience Institute, New York University Langone Health, New York, NY, USA
- The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Craig Stark
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, CA, USA
| | - James J Knierim
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Lara M Rangel
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
34
|
Nakamura NH, Furue H, Kobayashi K, Oku Y. Hippocampal ensemble dynamics and memory performance are modulated by respiration during encoding. Nat Commun 2023; 14:4391. [PMID: 37500646 PMCID: PMC10374532 DOI: 10.1038/s41467-023-40139-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/13/2023] [Indexed: 07/29/2023] Open
Abstract
During offline brain states, such as sleep and memory consolidation, respiration coordinates hippocampal activity. However, the role of breathing during online memory traces remains unclear. Here, we show that respiration can be recruited during online memory encoding. Optogenetic manipulation was used to control activation of the primary inspiratory rhythm generator PreBötzinger complex (PreBötC) in transgenic mice. When intermittent PreBötC-induced apnea covered the object exploration time during encoding, novel object detection was impaired. Moreover, the mice did not exhibit freezing behavior during presentation of fear-conditioned stimuli (CS+) when PreBötC-induced apnea occurred at the exact time of encoding. This apnea did not evoke changes in CA3 cell ensembles between presentations of CS+ and conditioned inhibition (CS-), whereas in normal breathing, CS+ presentations produced dynamic changes. Our findings demonstrate that components of central respiratory activity (e.g., frequency) during online encoding strongly contribute to shaping hippocampal ensemble dynamics and memory performance.
Collapse
Affiliation(s)
- Nozomu H Nakamura
- Division of Physiome, Department of Physiology, Hyogo Medical University, 1-1, Mukogawa cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Hidemasa Furue
- Division of Neurophysiology, Department of Physiology, Hyogo Medical University, 1-1, Mukogawa cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, 444-8585, Japan
| | - Yoshitaka Oku
- Division of Physiome, Department of Physiology, Hyogo Medical University, 1-1, Mukogawa cho, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
35
|
Ireton KE, Xing X, Kim K, Weiner JC, Jacobi AA, Grover A, Foote M, Ota Y, Berman R, Hanks T, Hell JW. Regulation of the Ca 2+ Channel Ca V1.2 Supports Spatial Memory and Its Flexibility and LTD. J Neurosci 2023; 43:5559-5573. [PMID: 37419689 PMCID: PMC10376936 DOI: 10.1523/jneurosci.1521-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 04/30/2023] [Accepted: 05/15/2023] [Indexed: 07/09/2023] Open
Abstract
Widespread release of norepinephrine (NE) throughout the forebrain fosters learning and memory via adrenergic receptor (AR) signaling, but the molecular mechanisms are largely unknown. The β2 AR and its downstream effectors, the trimeric stimulatory Gs-protein, adenylyl cyclase (AC), and the cAMP-dependent protein kinase A (PKA), form a unique signaling complex with the L-type Ca2+ channel (LTCC) CaV1.2. Phosphorylation of CaV1.2 by PKA on Ser1928 is required for the upregulation of Ca2+ influx on β2 AR stimulation and long-term potentiation induced by prolonged theta-tetanus (PTT-LTP) but not LTP induced by two 1-s-long 100-Hz tetani. However, the function of Ser1928 phosphorylation in vivo is unknown. Here, we show that S1928A knock-in (KI) mice of both sexes, which lack PTT-LTP, express deficiencies during initial consolidation of spatial memory. Especially striking is the effect of this mutation on cognitive flexibility as tested by reversal learning. Mechanistically, long-term depression (LTD) has been implicated in reversal learning. It is abrogated in male and female S1928A knock-in mice and by β2 AR antagonists and peptides that displace β2 AR from CaV1.2. This work identifies CaV1.2 as a critical molecular locus that regulates synaptic plasticity, spatial memory and its reversal, and LTD.SIGNIFICANCE STATEMENT We show that phosphorylation of the Ca2+ channel CaV1.2 on Ser1928 is important for consolidation of spatial memory and especially its reversal, and long-term depression (LTD). Identification of Ser1928 as critical for LTD and reversal learning supports the model that LTD underlies flexibility of reference memory.
Collapse
Affiliation(s)
- Kyle E Ireton
- Department of Pharmacology, University of California, Davis, California 95616-8636
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Xiaoming Xing
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Karam Kim
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Justin C Weiner
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Ariel A Jacobi
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Aarushi Grover
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Molly Foote
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Yusuke Ota
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Robert Berman
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Timothy Hanks
- Center for Neuroscience, University of California, Davis, California 95616-8636
- Department of Neurology, University of California, Davis, California 95616-8636
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, California 95616-8636
- Center for Neuroscience, University of California, Davis, California 95616-8636
| |
Collapse
|
36
|
Brosens N, Lesuis SL, Bassie I, Reyes L, Gajadien P, Lucassen PJ, Krugers HJ. Elevated corticosterone after fear learning impairs remote auditory memory retrieval and alters brain network connectivity. Learn Mem 2023; 30:125-132. [PMID: 37487708 PMCID: PMC10519398 DOI: 10.1101/lm.053836.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023]
Abstract
Glucocorticoids are potent memory modulators that can modify behavior in an adaptive or maladaptive manner. Elevated glucocorticoid levels after learning promote memory consolidation at recent time points, but their effects on remote time points are not well established. Here we set out to assess whether corticosterone (CORT) given after learning modifies remote fear memory. To that end, mice were exposed to a mild auditory fear conditioning paradigm followed by a single 2 mg/kg CORT injection, and after 28 d, auditory memory was assessed. Neuronal activation was investigated using immunohistochemistry for the immediate early gene c-Fos, and coactivation of brain regions was determined using a correlation matrix analysis. CORT-treated mice displayed significantly less remote auditory memory retrieval. While the net activity of studied brain regions was similar compared with the control condition, CORT-induced remote memory impairment was associated with altered correlated activity between brain regions. Specifically, connectivity of the lateral amygdala with the basal amygdala and the dorsal dentate gyrus was significantly reduced in CORT-treated mice, suggesting disrupted network connectivity that may underlie diminished remote memory retrieval. Elucidating the pathways underlying these effects could help provide mechanistic insight into the effects of stress on memory and possibly provide therapeutic targets for psychopathology.
Collapse
Affiliation(s)
- Niek Brosens
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Sylvie L Lesuis
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Ilse Bassie
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Lara Reyes
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Priya Gajadien
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences (SILS)-Cognitive and Systems Neuroscience (CNS), University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| |
Collapse
|
37
|
Ohno M. Accelerated long-term forgetting: A sensitive paradigm for detecting subtle cognitive impairment and evaluating BACE1 inhibitor efficacy in preclinical Alzheimer's disease. FRONTIERS IN DEMENTIA 2023; 2:1161875. [PMID: 39081986 PMCID: PMC11285641 DOI: 10.3389/frdem.2023.1161875] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/27/2023] [Indexed: 08/02/2024]
Abstract
Given a long preclinical stage of Alzheimer's disease (AD) continuum before the onset of dementia, there is a growing demand for tools capable of detecting the earliest feature of subtle cognitive impairment and optimizing recruitment to clinical trials for potentially disease-modifying therapeutic interventions such as BACE1 inhibitors. Now that all BACE1 inhibitor programs in symptomatic and prodromal AD populations have ended in failure, trials need to shift to target the earlier preclinical stage. However, evaluating cognitive efficacy (if any) in asymptomatic AD individuals is a great challenge. In this context, accelerated long-term forgetting (ALF) is emerging as a sensitive cognitive measure that can discriminate between presymptomatic individuals with high risks for developing AD and healthy controls. ALF is characterized by increased forgetting rates over extended delays (e.g., days, weeks, months) despite normal learning and short-term retention on standard memory assessments that typically use around 30-min delays. This review provides an overview of recent progress in animal model and clinical studies on this topic, focusing on the utility and underlying mechanism of ALF that may be applicable to earlier diagnosis and BACE1 inhibitor efficacy evaluation at a preclinical stage of AD.
Collapse
Affiliation(s)
- Masuo Ohno
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, United States
| |
Collapse
|
38
|
Kenna M, Marek R, Sah P. Insights into the encoding of memories through the circuitry of fear. Curr Opin Neurobiol 2023; 80:102712. [PMID: 37003106 DOI: 10.1016/j.conb.2023.102712] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 04/03/2023]
Abstract
Associative learning induces physical changes to a network of cells, known as the memory engram. Fear is widely used as a model to understand the circuit motifs that underpin associative memories. Recent advances suggest that the distinct circuitry engaged by different conditioned stimuli (e.g. tone vs. context) can provide insights into what information is being encoded in the fear engram. Moreover, as the fear memory matures, the circuitry engaged indicates how information is remodelled after learning and hints at potential mechanisms for consolidation. Finally, we propose that the consolidation of fear memories involves plasticity of engram cells through coordinated activity between brain regions, and the inherent characteristics of the circuitry may mediate this process.
Collapse
Affiliation(s)
- Matthew Kenna
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Roger Marek
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Pankaj Sah
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
39
|
Yang Q, Zhou J, Wang L, Hu W, Zhong Y, Li Q. Spontaneous recovery of reward memory through active forgetting of extinction memory. Curr Biol 2023; 33:838-848.e3. [PMID: 36731465 DOI: 10.1016/j.cub.2023.01.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 12/16/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023]
Abstract
Learned behavior can be suppressed by the extinction procedure. Such extinguished memory often returns spontaneously over time, making it difficult to treat diseases such as addiction. However, the biological mechanisms underlying such spontaneous recovery remain unclear. Here, we report that the extinguished reward memory in Drosophila recovers spontaneously because extinction training forms an aversive memory that can be actively forgotten via the Rac1/Dia pathway. Manipulating Rac1 activity does not affect sugar-reward memory and its immediate extinction effect but bidirectionally regulates spontaneous recovery-the decay process of extinction. Experiments using thermogenetic inhibition and functional imaging support that such extinction appears to be coded as an aversive experience. Genetic and pharmacological inhibition of formin Dia, a downstream effector of Rac1, specifically prevents spontaneous recovery after extinction in both behavioral performance and corresponding physiological traces. Together, our data suggest that spontaneous recovery is caused by active forgetting of the opposing extinction memory.
Collapse
Affiliation(s)
- Qi Yang
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jun Zhou
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Lingling Wang
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Wantong Hu
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yi Zhong
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| | - Qian Li
- School of Life Sciences, IDG/McGovern Institute for Brain Research, MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
40
|
Mollon JD, Danilova MV, Zhuravlev AV. A possible mechanism of neural read-out from a molecular engram. Neurobiol Learn Mem 2023; 200:107748. [PMID: 36907505 DOI: 10.1016/j.nlm.2023.107748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023]
Abstract
What is the physical basis of declarative memory? The predominant view holds that stored information is embedded in the structure of a neural net, that is, in the signs and weights of its synaptic connections. An alternative possibility is that storage and processing are separated, and that the engram is encoded chemically, most probably in the sequence of a nucleic acid. One deterrent to adoption of the latter hypothesis has been the difficulty of envisaging how neural actively could be converted to and from a molecular code. Our purpose here is limited to suggesting how a molecular sequence could be read out from nucleic acid to neural activity by means of nanopores.
Collapse
Affiliation(s)
- J D Mollon
- Department of Psychology, University of Cambridge, Downing St., Cambridge CB2 3EB, United Kingdom.
| | - M V Danilova
- Department of Psychology, University of Cambridge, Downing St., Cambridge CB2 3EB, United Kingdom; I.P. Pavlov Institute of Physiology, nab Makarova 6, 199034 St Petersburg, Russian Federation
| | - A V Zhuravlev
- I.P. Pavlov Institute of Physiology, nab Makarova 6, 199034 St Petersburg, Russian Federation
| |
Collapse
|
41
|
Enhanced Learning and Forgetting Behavior for Contextual Knowledge Tracing. INFORMATION 2023. [DOI: 10.3390/info14030168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Knowledge tracing (KT) is based on modeling students’ behavior sequences to obtain students’ knowledge state and predict students’ future performance. The KT task aims to model students’ knowledge state in real-time according to their historical learning behavior, so as to predict their future learning performance. Online education has become more critical in recent years due to the impact of COVID-19, and KT has also attracted much attention due to its importance in the education field. However, previous KT models generally have the following three problems. Firstly, students’ learning and forgetting behaviors affect their knowledge state, and past KT models have yet to exploit this fully. Secondly, the input of traditional KT models is mainly limited to students’ exercise sequence and answers. In the learning process, students’ answering performance can reflect their knowledge level. Finally, the context of students’ learning sequence also affects their judgment of the knowledge state. In this paper, we combined educational psychology theories to propose enhanced learning and forgetting behavior for contextual knowledge tracing (LFEKT). LFEKT enriches the features of exercises by introducing difficulty information and considers the influence of students’ answering behavior on the knowledge state. In order to model students’ learning and forgetting behavior, LFEKT integrates multiple influencing factors to build a knowledge acquisition module and a knowledge retention module. Furthermore, LFEKT introduces a long short-term memory (LSTM) network to capture the contextual relations of learned sequences. From the experimental results, it can be seen that LFEKT had better prediction performance than existing models on four public datasets, which indicates that LFEKT can better trace students’ knowledge state and has better prediction performance.
Collapse
|
42
|
Bolsius YG, Heckman PRA, Paraciani C, Wilhelm S, Raven F, Meijer EL, Kas MJH, Ramirez S, Meerlo P, Havekes R. Recovering object-location memories after sleep deprivation-induced amnesia. Curr Biol 2023; 33:298-308.e5. [PMID: 36577400 DOI: 10.1016/j.cub.2022.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/19/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022]
Abstract
It is well established that sleep deprivation after learning impairs hippocampal memory processes and can cause amnesia. It is unknown, however, whether sleep deprivation leads to the loss of information or merely the suboptimal storage of information that is difficult to retrieve. Here, we show that hippocampal object-location memories formed under sleep deprivation conditions can be successfully retrieved multiple days following training, using optogenetic dentate gyrus (DG) memory engram activation or treatment with the clinically approved phosphodiesterase 4 (PDE4) inhibitor roflumilast. Moreover, the combination of optogenetic DG memory engram activation and roflumilast treatment, 2 days following training and sleep deprivation, made the memory more persistently accessible for retrieval even several days later (i.e., without further optogenetic or pharmacological manipulation). Altogether, our studies in mice demonstrate that sleep deprivation does not necessarily cause memory loss but instead leads to the suboptimal storage of information that cannot be retrieved without drug treatment or optogenetic stimulation. Furthermore, our findings suggest that object-location memories, consolidated under sleep deprivation conditions and thought to be lost, can be made accessible again several days after the learning and sleep deprivation episode, using the clinically approved PDE4 inhibitor roflumilast.
Collapse
Affiliation(s)
- Youri G Bolsius
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands; Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, the Netherlands
| | - Camilla Paraciani
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Sophia Wilhelm
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Frank Raven
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Elroy L Meijer
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Martien J H Kas
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, The Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA
| | - Peter Meerlo
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
43
|
Roüast NM, Schönauer M. Continuously changing memories: a framework for proactive and non-linear consolidation. Trends Neurosci 2023; 46:8-19. [PMID: 36428193 DOI: 10.1016/j.tins.2022.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/10/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022]
Abstract
The traditional view of long-term memory is that memory traces mature in a predetermined 'linear' process: their neural substrate shifts from rapidly plastic medial temporal regions towards stable neocortical networks. We propose that memories remain malleable, not by repeated reinstantiations of this linear process but instead via dynamic routes of proactive and non-linear consolidation: memories change, their trajectory is flexible and reversible, and their physical basis develops continuously according to anticipated demands. Studies demonstrating memory updating, increasing hippocampal dependence to support adaptive use, and rapid neocortical plasticity provide evidence for continued non-linear consolidation. Although anticipated demand can affect all stages of memory formation, the extent to which it shapes the physical memory trace repeatedly and proactively will require further dedicated research.
Collapse
Affiliation(s)
- Nora Malika Roüast
- Institute for Psychology, Neuropsychology, University of Freiburg, Freiburg, Germany.
| | - Monika Schönauer
- Institute for Psychology, Neuropsychology, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
44
|
(-)- Gossypol Inhibition of Musashi-Mediated Forgetting Improves Memory and Age-Dependent Memory Decline in Caenorhabditis elegans. Mol Neurobiol 2023; 60:820-835. [PMID: 36378468 PMCID: PMC9849318 DOI: 10.1007/s12035-022-03116-7] [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: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Musashi RNA-binding proteins (MSIs) retain a pivotal role in stem cell maintenance, tumorigenesis, and nervous system development. Recently, we showed in C. elegans that Musashi (MSI-1) actively promotes forgetting upon associative learning via a 3'UTR-dependent translational expression of the Arp2/3 actin branching complex. Here, we investigated the evolutionary conserved role of MSI proteins and the effect of their pharmacological inhibition on memory. Expression of human Musashi 1 (MSI1) and Musashi 2 (MSI2) under the endogenous Musashi promoter fully rescued the phenotype of msi-1(lf) worms. Furthermore, pharmacological inhibition of human MSI1 and MSI2 activity using (-)- gossypol resulted in improved memory retention, without causing locomotor, chemotactic, or learning deficits. No drug effect was observed in msi-1(lf) treated worms. Using Western blotting and confocal microscopy, we found no changes in MSI-1 protein abundance following (-)- gossypol treatment, suggesting that Musashi gene expression remains unaltered and that the compound exerts its inhibitory effect post-translationally. Additionally, (-)- gossypol suppressed the previously seen rescue of the msi-1(lf) phenotype in worms expressing human MSI1 specifically in the AVA neuron, indicating that (-)- gossypol can regulate the Musashi pathway in a memory-related neuronal circuit in worms. Finally, treating aged worms with (-)- gossypol reversed physiological age-dependent memory decline. Taken together, our findings indicate that pharmacological inhibition of Musashi might represent a promising approach for memory modulation.
Collapse
|
45
|
Park S, Jung JH, Karimi SA, Jacob AD, Josselyn SA. Opto-extinction of a threat memory in mice. Brain Res Bull 2022; 191:61-68. [DOI: 10.1016/j.brainresbull.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/02/2022]
|
46
|
Ernst E. The AI trilemma: Saving the planet without ruining our jobs. Front Artif Intell 2022; 5:886561. [PMID: 36337142 PMCID: PMC9626962 DOI: 10.3389/frai.2022.886561] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/12/2022] [Indexed: 01/24/2023] Open
Abstract
Digitalization and artificial intelligence increasingly affect the world of work. Rising risk of massive job losses have sparked technological fears. Limited income and productivity gains concentrated among a few tech companies are fueling inequalities. In addition, the increasing ecological footprint of digital technologies has become the focus of much discussion. This creates a trilemma of rising inequality, low productivity growth and high ecological costs brought by technological progress. How can this trilemma be resolved? Which digital applications should be promoted specifically? And what should policymakers do to address this trilemma? This contribution shows that policymakers should create suitable conditions to fully exploit the potential in the area of network applications (transport, information exchange, supply, provisioning) in order to reap maximum societal benefits that can be widely shared. This requires shifting incentives away from current uses toward those that can, at least partially, address the trilemma. The contribution analyses the scope and limits of current policy instruments in this regard and discusses alternative approaches that are more aligned with the properties of the emerging technological paradigm underlying the digital economy. In particular, it discusses the possibility of institutional innovations required to address the socio-economic challenges resulting from the technological innovations brought about by artificial intelligence.
Collapse
Affiliation(s)
- Ekkehard Ernst
- International Labour Organization, Department of Research, Geneva, Switzerland
| |
Collapse
|
47
|
Dynamic tripartite construct of interregional engram circuits underlies forgetting of extinction memory. Mol Psychiatry 2022; 27:4077-4091. [PMID: 35804093 DOI: 10.1038/s41380-022-01684-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
Fear extinction allows for adaptive control of learned fear responses but often fails, resulting in a renewal or spontaneous recovery of the extinguished fear, i.e., forgetting of the extinction memory readily occurs. Using an activity-dependent neuronal labeling strategy, we demonstrate that engram neurons for fear extinction memory are dynamically positioned in the medial prefrontal cortex (mPFC), basolateral amygdala (BLA), and ventral hippocampus (vHPC), which constitute an engram construct in the term of directional engram synaptic connectivity from the BLA or vHPC to mPFC, but not that in the opposite direction, for retrieval of extinction memory. Fear renewal or spontaneous recovery switches the extinction engram construct from an accessible to inaccessible state, whereas additional extinction learning or optogenetic induction of long-term potentiation restores the directional engram connectivity and prevents the return of fear. Thus, the plasticity of engram construct underlies forgetting of extinction memory.
Collapse
|
48
|
Active forgetting requires Sickie function in a dedicated dopamine circuit in Drosophila. Proc Natl Acad Sci U S A 2022; 119:e2204229119. [PMID: 36095217 PMCID: PMC9499536 DOI: 10.1073/pnas.2204229119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Forgetting is an essential component of the brain's memory management system, providing a balance to memory formation processes by removing unused or unwanted memories, or by suppressing their expression. However, the molecular, cellular, and circuit mechanisms underlying forgetting are poorly understood. Here we show that the memory suppressor gene, sickie, functions in a single dopamine neuron (DAn) by supporting the process of active forgetting in Drosophila. RNAi knockdown (KD) of sickie impairs forgetting by reducing the Ca2+ influx and DA release from the DAn that promotes forgetting. Coimmunoprecipitation/mass spectrometry analyses identified cytoskeletal and presynaptic active zone (AZ) proteins as candidates that physically interact with Sickie, and a focused RNAi screen of the candidates showed that Bruchpilot (Brp)-a presynaptic AZ protein that regulates calcium channel clustering and neurotransmitter release-impairs active forgetting like sickie KD. In addition, overexpression of brp rescued the impaired forgetting of sickie KD, providing evidence that they function in the same process. Moreover, we show that sickie KD in the DAn reduces the abundance and size of AZ markers but increases their number, suggesting that Sickie controls DAn activity for forgetting by modulating the presynaptic AZ structure. Our results identify a molecular and circuit mechanism for normal levels of active forgetting and reveal a surprising role of Sickie in maintaining presynaptic AZ structure for neurotransmitter release.
Collapse
|
49
|
Ben-Simon Y, Kaefer K, Velicky P, Csicsvari J, Danzl JG, Jonas P. A direct excitatory projection from entorhinal layer 6b neurons to the hippocampus contributes to spatial coding and memory. Nat Commun 2022; 13:4826. [PMID: 35974109 PMCID: PMC9381769 DOI: 10.1038/s41467-022-32559-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/03/2022] [Indexed: 11/08/2022] Open
Abstract
The mammalian hippocampal formation (HF) plays a key role in several higher brain functions, such as spatial coding, learning and memory. Its simple circuit architecture is often viewed as a trisynaptic loop, processing input originating from the superficial layers of the entorhinal cortex (EC) and sending it back to its deeper layers. Here, we show that excitatory neurons in layer 6b of the mouse EC project to all sub-regions comprising the HF and receive input from the CA1, thalamus and claustrum. Furthermore, their output is characterized by unique slow-decaying excitatory postsynaptic currents capable of driving plateau-like potentials in their postsynaptic targets. Optogenetic inhibition of the EC-6b pathway affects spatial coding in CA1 pyramidal neurons, while cell ablation impairs not only acquisition of new spatial memories, but also degradation of previously acquired ones. Our results provide evidence of a functional role for cortical layer 6b neurons in the adult brain.
Collapse
Affiliation(s)
- Yoav Ben-Simon
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
- Department of Neurophysiology and Pharmacology, Vienna Medical University, Vienna, Austria.
| | - Karola Kaefer
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
- Department of Neuroinformatics, Radboud University, Nijmegen, The Netherlands
| | - Philipp Velicky
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Jozsef Csicsvari
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Johann G Danzl
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Peter Jonas
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| |
Collapse
|
50
|
Gisquet-Verrier P. Can Forgetting Be Due to Changes in Engram Cell Plasticity? Front Behav Neurosci 2022; 16:945985. [PMID: 35874646 PMCID: PMC9302596 DOI: 10.3389/fnbeh.2022.945985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
|