1
|
Zhou YQ, Puliyadi V, Chen X, Lee JL, Zhang LY, Knierim JJ. Vector coding and place coding in hippocampus share a common directional signal. Nat Commun 2024; 15:10630. [PMID: 39638805 PMCID: PMC11621709 DOI: 10.1038/s41467-024-54935-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Vector coding is a major mechanism by which neural systems represent an animal's location in both global and local, item-based reference frames. Landmark vector cells (LVCs) in the hippocampus complement classic place cells by encoding vector relationships between the organism and specific landmarks. How these place- and vector-coding properties interact is not known. We recorded place cells and LVCs using calcium imaging of the CA1 region of freely moving rats during cue-card rotation studies. Place fields rotated around the center of the platform to follow the cue rotation, whereas the fields of simultaneously recorded LVCs rotated by the same amount around the nearby landmarks. Some neurons demonstrated conjunctive coding of both classic place field properties and LVC properties. These results demonstrate that CA1 neurons employ a common directional input, presumably provided by the head direction cell system, to encode animals' locations in both world-centered and landmark-centered reference frames.
Collapse
Affiliation(s)
- Yue-Qing Zhou
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA.
| | - Vyash Puliyadi
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Xiaojing Chen
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Joonhee Leo Lee
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Lan-Yuan Zhang
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James J Knierim
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA.
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Donahue MM, Robson E, Colgin LL. Hippocampal place cell sequences are impaired in a rat model of Fragile X Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619112. [PMID: 39553951 PMCID: PMC11566021 DOI: 10.1101/2024.10.18.619112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Fragile X Syndrome (FXS) is a neurodevelopmental disorder that can cause impairments in spatial cognition and memory. The hippocampus is thought to support spatial cognition through the activity of place cells, neurons with spatial receptive fields. Coordinated firing of place cell populations is organized by different oscillatory patterns in the hippocampus during specific behavioral states. Theta rhythms organize place cell populations during awake exploration. Sharp wave-ripples organize place cell population reactivation during waking rest. Here, we examined the coordination of CA1 place cell populations during active behavior and subsequent rest in a rat model of FXS ( Fmr1 knockout rats). While the organization of individual place cells by the theta rhythm was normal, the coordinated activation of sequences of place cells during individual theta cycles was impaired in Fmr1 knockout rats. Further, the subsequent replay of place cell sequences was impaired during waking rest following active exploration. Together, these results expand our understanding of how genetic modifications that model those observed in FXS affect hippocampal physiology and suggest a potential mechanism underlying impaired spatial cognition in FXS. Significance Statement Fragile X Syndrome (FXS) is a neurodevelopmental disorder that can cause impaired memory and atypical spatial behaviors such as "elopement" (i.e., wandering off and becoming lost). Activity in the CA1 subregion of the hippocampus supports spatial memory and spatial cognition, making it an important candidate to study in the context of FXS; however, how neuronal population activity in CA1 is affected by FXS is poorly understood. In this study, we found that the coordination of populations of CA1 neurons during active behavior and waking rest was impaired in a rat model of FXS. These results reveal hippocampal physiological deficits that may contribute to cognitive impairments in FXS.
Collapse
|
3
|
Singer RA, Rajchin V, Park K, Heintz N, Darnell RB. Opto-CLIP reveals dynamic FMRP regulation of mRNAs upon CA1 neuronal activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607210. [PMID: 39185177 PMCID: PMC11343148 DOI: 10.1101/2024.08.13.607210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Neuronal diversity and function are intricately linked to the dynamic regulation of RNA metabolism, including splicing, localization, and translation. Electrophysiologic studies of synaptic plasticity, models for learning and memory, are disrupted in Fragile X Syndrome (FXS). FXS is characterized by the loss of FMRP, an RNA-binding protein (RBP) known to bind and suppress translation of specific neuronal RNAs. Since molecular studies have demonstrated that synaptic plasticity in CA1 excitatory hippocampal neurons is protein-synthesis dependent, together these observations have suggested a potential role for FMRP in synaptic plasticity in FXS. To explore this model, we developed a new experimental platform, Opto-CLIP, to integrate optogenetics with cell-type specific FMRP CLIP and RiboTag in CA1 hippocampal neurons, allowing investigation of FMRP-regulated dynamics after neuronal activation. We tracked changes in FMRP binding and ribosome-associated RNA profiles 30 minutes after neuronal activation. Our findings reveal a significant reduction in FMRP-RNA binding to transcripts encoding nuclear proteins, suggesting FMRP translational inhibition may be de-repressed to allow rapid translational responses required for neuronal homeostasis. In contrast, FMRP binding to transcripts encoding synaptic targets were generally stable after activation, but all categories of targets demonstrated variability in FMRP translational control. Opto-CLIP revealed differential regulation of subsets of transcripts within CA1 neurons rapidly after depolarization, and offers promise as a generally useful platform to uncover mechanisms of RBP-mediated RNA regulation in the context of synaptic plasticity.
Collapse
Affiliation(s)
- Ruth A. Singer
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
| | - Veronika Rajchin
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
| | - Kwanghoon Park
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Nathaniel Heintz
- Laboratory of Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Robert B. Darnell
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| |
Collapse
|
4
|
Fenton AA. Remapping revisited: how the hippocampus represents different spaces. Nat Rev Neurosci 2024; 25:428-448. [PMID: 38714834 DOI: 10.1038/s41583-024-00817-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 05/25/2024]
Abstract
The representation of distinct spaces by hippocampal place cells has been linked to changes in their place fields (the locations in the environment where the place cells discharge strongly), a phenomenon that has been termed 'remapping'. Remapping has been assumed to be accompanied by the reorganization of subsecond cofiring relationships among the place cells, potentially maximizing hippocampal information coding capacity. However, several observations challenge this standard view. For example, place cells exhibit mixed selectivity, encode non-positional variables, can have multiple place fields and exhibit unreliable discharge in fixed environments. Furthermore, recent evidence suggests that, when measured at subsecond timescales, the moment-to-moment cofiring of a pair of cells in one environment is remarkably similar in another environment, despite remapping. Here, I propose that remapping is a misnomer for the changes in place fields across environments and suggest instead that internally organized manifold representations of hippocampal activity are actively registered to different environments to enable navigation, promote memory and organize knowledge.
Collapse
Affiliation(s)
- André A Fenton
- Center for Neural Science, New York University, New York, NY, USA.
- Neuroscience Institute at the NYU Langone Medical Center, New York, NY, USA.
| |
Collapse
|
5
|
Severino L, Kim J, Nam MH, McHugh TJ. From synapses to circuits: What mouse models have taught us about how autism spectrum disorder impacts hippocampal function. Neurosci Biobehav Rev 2024; 158:105559. [PMID: 38246230 DOI: 10.1016/j.neubiorev.2024.105559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that impacts a variety of cognitive and behavioral domains. While a genetic component of ASD has been well-established, none of the numerous syndromic genes identified in humans accounts for more than 1% of the clinical patients. Due to this large number of target genes, numerous mouse models of the disorder have been generated. However, the focus on distinct brain circuits, behavioral phenotypes and diverse experimental approaches has made it difficult to synthesize the overwhelming number of model animal studies into concrete throughlines that connect the data across levels of investigation. Here we chose to focus on one circuit, the hippocampus, and one hypothesis, a shift in excitatory/inhibitory balance, to examine, from the level of the tripartite synapse up to the level of in vivo circuit activity, the key commonalities across disparate models that can illustrate a path towards a better mechanistic understanding of ASD's impact on hippocampal circuit function.
Collapse
Affiliation(s)
- Leandra Severino
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea
| | - Jinhyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea.
| | - Thomas J McHugh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi Saitama, Japan.
| |
Collapse
|
6
|
Subramanian M, Mills WT, Paranjpe MD, Onuchukwu US, Inamdar M, Maytin AR, Li X, Pomerantz JL, Meffert MK. Growth-suppressor microRNAs mediate synaptic overgrowth and behavioral deficits in Fragile X mental retardation protein deficiency. iScience 2024; 27:108676. [PMID: 38235335 PMCID: PMC10792201 DOI: 10.1016/j.isci.2023.108676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/20/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024] Open
Abstract
Abnormal neuronal and synapse growth is a core pathology resulting from deficiency of the Fragile X mental retardation protein (FMRP), but molecular links underlying the excessive synthesis of key synaptic proteins remain incompletely defined. We find that basal brain levels of the growth suppressor let-7 microRNA (miRNA) family are selectively lowered in FMRP-deficient mice and activity-dependent let-7 downregulation is abrogated. Primary let-7 miRNA transcripts are not altered in FMRP-deficiency and posttranscriptional misregulation occurs downstream of MAPK pathway induction and elevation of Lin28a, a let-7 biogenesis inhibitor. Neonatal restoration of brain let-7 miRNAs corrects hallmarks of FMRP-deficiency, including dendritic spine overgrowth and social and cognitive behavioral deficits, in adult mice. Blockade of MAPK hyperactivation normalizes let-7 miRNA levels in both brain and peripheral blood plasma from Fmr1 KO mice. These results implicate dysregulated let-7 miRNA biogenesis in the pathogenesis of FMRP-deficiency, and highlight let-7 miRNA-based strategies for future biomarker and therapeutic development.
Collapse
Affiliation(s)
- Megha Subramanian
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - William T. Mills
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Manish D. Paranjpe
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Uche S. Onuchukwu
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Manasi Inamdar
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amanda R. Maytin
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xinbei Li
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joel L. Pomerantz
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mollie K. Meffert
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Viana da Silva S, Haberl MG, Gaur K, Patel R, Narayan G, Ledakis M, Fu ML, de Castro Vieira M, Koo EH, Leutgeb JK, Leutgeb S. Localized APP expression results in progressive network dysfunction by disorganizing spike timing. Neuron 2024; 112:124-140.e6. [PMID: 37909036 PMCID: PMC10877582 DOI: 10.1016/j.neuron.2023.10.001] [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: 12/26/2022] [Revised: 06/16/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023]
Abstract
Progressive cognitive decline in Alzheimer's disease could either be caused by a spreading molecular pathology or by an initially focal pathology that causes aberrant neuronal activity in a larger network. To distinguish between these possibilities, we generated a mouse model with expression of mutant human amyloid precursor protein (APP) in only hippocampal CA3 cells. We found that performance in a hippocampus-dependent memory task was impaired in young adult and aged mutant mice. In both age groups, we then recorded from the CA1 region, which receives inputs from APP-expressing CA3 cells. We observed that theta oscillation frequency in CA1 was reduced along with disrupted relative timing of principal cells. Highly localized pathology limited to the presynaptic CA3 cells is thus sufficient to cause aberrant firing patterns in postsynaptic neuronal networks, which indicates that disease progression is not only from spreading pathology but also mediated by progressively advancing physiological dysfunction.
Collapse
Affiliation(s)
- Silvia Viana da Silva
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA; NeuroCure Excellence Cluster and German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Matthias G Haberl
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, Charitéplatz 1, 10117 Berlin, Germany
| | - Kshitij Gaur
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Rina Patel
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Gautam Narayan
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Max Ledakis
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Maylin L Fu
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Miguel de Castro Vieira
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, Charitéplatz 1, 10117 Berlin, Germany
| | - Edward H Koo
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jill K Leutgeb
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - Stefan Leutgeb
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA; Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Park EH, Kao HY, Jourdi H, van Dijk MT, Carrillo-Segura S, Tunnell KW, Gutierrez J, Wallace EJ, Troy-Regier M, Radwan B, Lesburguères E, Alarcon JM, Fenton AA. Phencyclidine Disrupts Neural Coordination and Cognitive Control by Dysregulating Translation. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:252-263. [PMID: 38298788 PMCID: PMC10829677 DOI: 10.1016/j.bpsgos.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 02/02/2024] Open
Abstract
Background Phencyclidine (PCP) causes psychosis, is abused with increasing frequency, and was extensively used in antipsychotic drug discovery. PCP discoordinates hippocampal ensemble action potential discharge and impairs cognitive control in rats, but how this uncompetitive NMDA receptor (NMDAR) antagonist impairs cognition remains unknown. Methods The effects of PCP were investigated on hippocampal CA1 ensemble action potential discharge in vivo in urethane-anesthetized rats and during awake behavior in mice, on synaptic responses in ex vivo mouse hippocampus slices, in mice on a hippocampus-dependent active place avoidance task that requires cognitive control, and on activating the molecular machinery of translation in acute hippocampus slices. Mechanistic causality was assessed by comparing the PCP effects with the effects of inhibitors of protein synthesis, group I metabotropic glutamate receptors (mGluR1/5), and subunit-selective NMDARs. Results Consistent with ionotropic actions, PCP discoordinated CA1 ensemble action potential discharge. PCP caused hyperactivity and impaired active place avoidance, despite the rodents having learned the task before PCP administration. Consistent with metabotropic actions, PCP exaggerated protein synthesis-dependent DHPG-induced mGluR1/5-stimulated long-term synaptic depression. Pretreatment with anisomycin or the mGluR1/5 antagonist MPEP, both of which repress translation, prevented PCP-induced discoordination and the cognitive and sensorimotor impairments. PCP as well as the NR2A-containing NMDAR antagonist NVP-AAM077 unbalanced translation that engages the Akt, mTOR (mechanistic target of rapamycin), and 4EBP1 translation machinery and increased protein synthesis, whereas the NR2B-containing antagonist Ro25-6981 did not. Conclusions PCP dysregulates translation, acting through NR2A-containing NMDAR subtypes, recruiting mGluR1/5 signaling pathways, and leading to neural discoordination that is central to the cognitive and sensorimotor impairments.
Collapse
Affiliation(s)
- Eun Hye Park
- Center for Neural Science, New York University, New York, New York
| | - Hsin-Yi Kao
- Center for Neural Science, New York University, New York, New York
| | - Hussam Jourdi
- Center for Neural Science, New York University, New York, New York
| | - Milenna T. van Dijk
- Center for Neural Science, New York University, New York, New York
- Graduate Program in Neuroscience and Physiology, New York University Langone Medical Center, New York, New York
| | - Simón Carrillo-Segura
- Center for Neural Science, New York University, New York, New York
- Graduate Program in Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, New York, New York
| | - Kayla W. Tunnell
- Center for Neural Science, New York University, New York, New York
| | | | - Emma J. Wallace
- Graduate Program in Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Matthew Troy-Regier
- Graduate Program in Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Basma Radwan
- Graduate Program in Neural Science, Center for Neural Science, New York University, New York, New York
| | | | - Juan Marcos Alarcon
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - André A. Fenton
- Center for Neural Science, New York University, New York, New York
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Neuroscience Institute, NYU Langone Health, New York, New York
| |
Collapse
|
9
|
Levy ERJ, Carrillo-Segura S, Park EH, Redman WT, Hurtado JR, Chung S, Fenton AA. A manifold neural population code for space in hippocampal coactivity dynamics independent of place fields. Cell Rep 2023; 42:113142. [PMID: 37742193 PMCID: PMC10842170 DOI: 10.1016/j.celrep.2023.113142] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/14/2023] [Accepted: 08/30/2023] [Indexed: 09/26/2023] Open
Abstract
Hippocampus place cell discharge is temporally unreliable across seconds and days, and place fields are multimodal, suggesting an "ensemble cofiring" spatial coding hypothesis with manifold dynamics that does not require reliable spatial tuning, in contrast to hypotheses based on place field (spatial tuning) stability. We imaged mouse CA1 (cornu ammonis 1) ensembles in two environments across three weeks to evaluate these coding hypotheses. While place fields "remap," being more distinct between than within environments, coactivity relationships generally change less. Decoding location and environment from 1-s ensemble location-specific activity is effective and improves with experience. Decoding environment from cell-pair coactivity relationships is also effective and improves with experience, even after removing place tuning. Discriminating environments from 1-s ensemble coactivity relies crucially on the cells with the most anti-coactive cell-pair relationships because activity is internally organized on a low-dimensional manifold of non-linear coactivity relationships that intermittently reregisters to environments according to the anti-cofiring subpopulation activity.
Collapse
Affiliation(s)
| | - Simón Carrillo-Segura
- Center for Neural Science, New York University, New York, NY 10003, USA; Graduate Program in Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, NY 11201, USA
| | - Eun Hye Park
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - William Thomas Redman
- Interdepartmental Graduate Program in Dynamical Neuroscience, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | | | - SueYeon Chung
- Center for Neural Science, New York University, New York, NY 10003, USA; Flatiron Institute Center for Computational Neuroscience, New York, NY 10010, USA
| | - André Antonio Fenton
- Center for Neural Science, New York University, New York, NY 10003, USA; Neuroscience Institute at the NYU Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
10
|
Fenton AA, Hurtado JR, Broek JAC, Park E, Mishra B. Do Place Cells Dream of Deceptive Moves in a Signaling Game? Neuroscience 2023; 529:129-147. [PMID: 37591330 PMCID: PMC10592151 DOI: 10.1016/j.neuroscience.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 07/27/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Abstract
We consider the possibility of applying game theory to analysis and modeling of neurobiological systems. Specifically, the basic properties and features of information asymmetric signaling games are considered and discussed as having potential to explain diverse neurobiological phenomena; we focus on neuronal action potential discharge that can represent cognitive variables in memory and purposeful behavior. We begin by arguing that there is a pressing need for conceptual frameworks that can permit analysis and integration of information and explanations across many scales of biological function including gene regulation, molecular and biochemical signaling, cellular and metabolic function, neuronal population, and systems level organization to generate plausible hypotheses across these scales. Developing such integrative frameworks is crucial if we are to understand cognitive functions like learning, memory, and perception. The present work focuses on systems neuroscience organized around the connected brain regions of the entorhinal cortex and hippocampus. These areas are intensely studied in rodent subjects as model neuronal systems that undergo activity-dependent synaptic plasticity to form neuronal circuits and represent memories and spatial knowledge used for purposeful navigation. Examples of cognition-related spatial information in the observed neuronal discharge of hippocampal place cell populations and medial entorhinal head-direction cell populations are used to illustrate possible challenges to information maximization concepts. It may be natural to explain these observations using the ideas and features of information asymmetric signaling games.
Collapse
Affiliation(s)
- André A Fenton
- Neurobiology of Cognition Laboratory, Center for Neural Science, New York University, New York, NY, USA; Neuroscience Institute at the NYU Langone Medical Center, New York, NY, USA.
| | - José R Hurtado
- Neurobiology of Cognition Laboratory, Center for Neural Science, New York University, New York, NY, USA
| | - Jantine A C Broek
- Departments of Computer Science and Mathematics, Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
| | - EunHye Park
- Neurobiology of Cognition Laboratory, Center for Neural Science, New York University, New York, NY, USA
| | - Bud Mishra
- Departments of Computer Science and Mathematics, Courant Institute of Mathematical Sciences, New York University, New York, NY, USA; Department of Cell Biology, NYU Langone Medical Center, New York, NY, USA; Simon Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| |
Collapse
|
11
|
Munn RGK, Wolff A, Speers LJ, Bilkey DK. Disrupted hippocampal synchrony following maternal immune activation in a rat model. Hippocampus 2023; 33:995-1008. [PMID: 37129454 DOI: 10.1002/hipo.23545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/14/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Maternal immune activation (MIA) is a risk factor for schizophrenia and other neurodevelopmental disorders. MIA in rats models a number of the brain and behavioral changes that are observed in schizophrenia, including impaired memory. Recent studies in the MIA model have shown that the firing of the hippocampal place cells that are involved in memory processes appear relatively normal, but with abnormalities in the temporal ordering of firing. In this study, we re-analyzed data from prior hippocampal electrophysiological recordings of MIA and control animals to determine whether temporal dysfunction was evident. We find that there is a decreased ratio of slow to fast gamma power, resulting from an increase in fast gamma power and a tendency toward reduced slow gamma power in MIA rats. Moreover, we observe a robust reduction in spectral coherence between hippocampal theta and both fast and slow gamma rhythms, as well as changes in the phase of theta at which fast gamma occurs. We also find the phasic organization of place cell phase precession on the theta wave to be abnormal in MIA rats. Lastly, we observe that the local field potential of MIA rats contains more frequent sharp-wave ripple events, and that place cells were more likely to fire spikes during ripples in these animals than control. These findings provide further evidence of desynchrony in MIA animals and may point to circuit-level changes that underlie failures to integrate and encode information in schizophrenia.
Collapse
Affiliation(s)
- Robert G K Munn
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Amy Wolff
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lucinda J Speers
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
12
|
Parra-Barrero E, Vijayabaskaran S, Seabrook E, Wiskott L, Cheng S. A map of spatial navigation for neuroscience. Neurosci Biobehav Rev 2023; 152:105200. [PMID: 37178943 DOI: 10.1016/j.neubiorev.2023.105200] [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: 01/25/2023] [Revised: 04/13/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023]
Abstract
Spatial navigation has received much attention from neuroscientists, leading to the identification of key brain areas and the discovery of numerous spatially selective cells. Despite this progress, our understanding of how the pieces fit together to drive behavior is generally lacking. We argue that this is partly caused by insufficient communication between behavioral and neuroscientific researchers. This has led the latter to under-appreciate the relevance and complexity of spatial behavior, and to focus too narrowly on characterizing neural representations of space-disconnected from the computations these representations are meant to enable. We therefore propose a taxonomy of navigation processes in mammals that can serve as a common framework for structuring and facilitating interdisciplinary research in the field. Using the taxonomy as a guide, we review behavioral and neural studies of spatial navigation. In doing so, we validate the taxonomy and showcase its usefulness in identifying potential issues with common experimental approaches, designing experiments that adequately target particular behaviors, correctly interpreting neural activity, and pointing to new avenues of research.
Collapse
Affiliation(s)
- Eloy Parra-Barrero
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, Bochum, Germany; International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Sandhiya Vijayabaskaran
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, Bochum, Germany
| | - Eddie Seabrook
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, Bochum, Germany
| | - Laurenz Wiskott
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, Bochum, Germany; International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Sen Cheng
- Institute for Neural Computation, Faculty of Computer Science, Ruhr University Bochum, Bochum, Germany; International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
13
|
Fmr1-KO mice failure to detect object novelty associates with a post-test decrease of structural and synaptic plasticity upstream of the hippocampus. Sci Rep 2023; 13:755. [PMID: 36641518 PMCID: PMC9840621 DOI: 10.1038/s41598-023-27991-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Mice with deletion of the FMR1 gene show episodic memory impairments and exhibit dendritic spines and synaptic plasticity defects prevalently identified in non-training conditions. Based on evidence that synaptic changes associated with normal or abnormal memory emerge when mice are cognitively challenged, here we examine whether, and how, fragile entorhinal and hippocampal synapses are remodeled when mice succeed or fail to learn. We trained Fmr1 knockout (KO) and wild-type C57BL/6J (WT) mice in the novel object recognition (NOR) paradigm with 1 h or 24 h training-to-test intervals and then assessed whether varying the time between the presentation of similar and different objects modulates NOR performance and plasticity along the entorhinal cortex-hippocampus axis. At the 1 h-interval, KO mice failed to discriminate the novel object, showed a collapse of spines in the lateral entorhinal cortex (LEC), and of long-term potentiation (LTP) in the lateral perforant path (LPP), but a normal increase in hippocampal spines. At the 24 h, they exhibited intact NOR performance, typical LEC and hippocampal spines, and exaggerated LPP-LTP. Our findings reveal that the inability of mice to detect object novelty primarily stands in their impediment to elaborate, and convey to the hippocampus, sensory/perceptive object representations.
Collapse
|
14
|
Asiminas A, Booker SA, Dando OR, Kozic Z, Arkell D, Inkpen FH, Sumera A, Akyel I, Kind PC, Wood ER. Experience-dependent changes in hippocampal spatial activity and hippocampal circuit function are disrupted in a rat model of Fragile X Syndrome. Mol Autism 2022; 13:49. [PMID: 36536454 PMCID: PMC9764562 DOI: 10.1186/s13229-022-00528-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a common single gene cause of intellectual disability and autism spectrum disorder. Cognitive inflexibility is one of the hallmarks of FXS with affected individuals showing extreme difficulty adapting to novel or complex situations. To explore the neural correlates of this cognitive inflexibility, we used a rat model of FXS (Fmr1-/y). METHODS We recorded from the CA1 in Fmr1-/y and WT littermates over six 10-min exploration sessions in a novel environment-three sessions per day (ITI 10 min). Our recordings yielded 288 and 246 putative pyramidal cells from 7 WT and 7 Fmr1-/y rats, respectively. RESULTS On the first day of exploration of a novel environment, the firing rate and spatial tuning of CA1 pyramidal neurons was similar between wild-type (WT) and Fmr1-/y rats. However, while CA1 pyramidal neurons from WT rats showed experience-dependent changes in firing and spatial tuning between the first and second day of exposure to the environment, these changes were decreased or absent in CA1 neurons of Fmr1-/y rats. These findings were consistent with increased excitability of Fmr1-/y CA1 neurons in ex vivo hippocampal slices, which correlated with reduced synaptic inputs from the medial entorhinal cortex. Lastly, activity patterns of CA1 pyramidal neurons were dis-coordinated with respect to hippocampal oscillatory activity in Fmr1-/y rats. LIMITATIONS It is still unclear how the observed circuit function abnormalities give rise to behavioural deficits in Fmr1-/y rats. Future experiments will focus on this connection as well as the contribution of other neuronal cell types in the hippocampal circuit pathophysiology associated with the loss of FMRP. It would also be interesting to see if hippocampal circuit deficits converge with those seen in other rodent models of intellectual disability. CONCLUSIONS In conclusion, we found that hippocampal place cells from Fmr1-/y rats show similar spatial firing properties as those from WT rats but do not show the same experience-dependent increase in spatial specificity or the experience-dependent changes in network coordination. Our findings offer support to a network-level origin of cognitive deficits in FXS.
Collapse
Affiliation(s)
- Antonis Asiminas
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.5254.60000 0001 0674 042XPresent Address: Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sam A. Booker
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Owen R. Dando
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988UK Dementia Research Institute at the Edinburgh Medical School, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Zrinko Kozic
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Daisy Arkell
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Felicity H. Inkpen
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Anna Sumera
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Irem Akyel
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Peter C. Kind
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,Centre for Brain Development and Repair, Bangalore, 560065 India
| | - Emma R. Wood
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,Centre for Brain Development and Repair, Bangalore, 560065 India
| |
Collapse
|
15
|
Gobbo F, Mitchell-Heggs R, Tse D, Al Omrani M, Spooner PA, Schultz SR, Morris RGM. Neuronal signature of spatial decision-making during navigation by freely moving rats by using calcium imaging. Proc Natl Acad Sci U S A 2022; 119:e2212152119. [PMID: 36279456 PMCID: PMC9636941 DOI: 10.1073/pnas.2212152119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
A challenge in spatial memory is understanding how place cell firing contributes to decision-making in navigation. A spatial recency task was created in which freely moving rats first became familiar with a spatial context over several days and thereafter were required to encode and then selectively recall one of three specific locations within it that was chosen to be rewarded that day. Calcium imaging was used to record from more than 1,000 cells in area CA1 of the hippocampus of five rats during the exploration, sample, and choice phases of the daily task. The key finding was that neural activity in the startbox rose steadily in the short period prior to entry to the arena and that this selective population cell firing was predictive of the daily changing goal on correct trials but not on trials in which the animals made errors. Single-cell and population activity measures converged on the idea that prospective coding of neural activity can be involved in navigational decision-making.
Collapse
Affiliation(s)
- Francesco Gobbo
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Rufus Mitchell-Heggs
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Department of Bioengineering and Centre for Neurotechnology, Imperial College London, London, SW7 2AZ, UK
| | - Dorothy Tse
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Department of Psychology, Edge Hill University, Ormskirk, L39 4QP, UK
| | - Meera Al Omrani
- MSc Program in Integrative Neuroscience, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Patrick A. Spooner
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Simon R. Schultz
- Department of Bioengineering and Centre for Neurotechnology, Imperial College London, London, SW7 2AZ, UK
| | - Richard G. M. Morris
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| |
Collapse
|
16
|
Wirtshafter HS, Disterhoft JF. In Vivo Multi-Day Calcium Imaging of CA1 Hippocampus in Freely Moving Rats Reveals a High Preponderance of Place Cells with Consistent Place Fields. J Neurosci 2022; 42:4538-4554. [PMID: 35501152 PMCID: PMC9172072 DOI: 10.1523/jneurosci.1750-21.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Calcium imaging using GCaMP indicators and miniature microscopes has been used to image cellular populations during long timescales and in different task phases, as well as to determine neuronal circuit topology and organization. Because the hippocampus (HPC) is essential for tasks of memory, spatial navigation, and learning, calcium imaging of large populations of HPC neurons can provide new insight on cell changes over time during these tasks. All reported HPC in vivo calcium imaging experiments have been done in mouse. However, rats have many behavioral and physiological experimental advantages over mice. In this paper, we present the first (to our knowledge) in vivo calcium imaging from CA1 HPC in freely moving male rats. Using the UCLA Miniscope, we demonstrate that, in rat, hundreds of cells can be visualized and held across weeks. We show that calcium events in these cells are highly correlated with periods of movement, with few calcium events occurring during periods without movement. We additionally show that an extremely large percent of cells recorded during a navigational task are place cells (77.3 ± 5.0%, surpassing the percent seen during mouse calcium imaging), and that these cells enable accurate decoding of animal position and can be held over days with consistent place fields in a consistent spatial map. A detailed protocol is included, and implications of these advancements on in vivo imaging and place field literature are discussed.SIGNIFICANCE STATEMENT In vivo calcium imaging in freely moving animals allows the visualization of cellular activity across days. In this paper, we present the first in vivo Ca2+ recording from CA1 hippocampus (HPC) in freely moving rats. We demonstrate that hundreds of cells can be visualized and held across weeks, and that calcium activity corresponds to periods of movement. We show that a high percentage (77.3 ± 5.0%) of imaged cells are place cells, and that these place cells enable accurate decoding and can be held stably over days with little change in field location. Because the HPC is essential for many tasks involving memory, navigation, and learning, imaging of large populations of HPC neurons can shed new insight on cellular activity changes and organization.
Collapse
Affiliation(s)
- Hannah S Wirtshafter
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - John F Disterhoft
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| |
Collapse
|
17
|
Homeostatic plasticity and excitation-inhibition balance: The good, the bad, and the ugly. Curr Opin Neurobiol 2022; 75:102553. [PMID: 35594578 DOI: 10.1016/j.conb.2022.102553] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/15/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022]
Abstract
In this review, we discuss the significance of the synaptic excitation/inhibition (E/I) balance in the context of homeostatic plasticity, whose primary goal is thought to maintain neuronal firing rates at a set point. We first provide an overview of the processes through which patterned input activity drives synaptic E/I tuning and maturation of circuits during development. Next, we emphasize the importance of the E/I balance at the synaptic level (homeostatic control of message reception) as a means to achieve the goal (homeostatic control of information transmission) at the network level and consider how compromised homeostatic plasticity associated with neurological diseases leads to hyperactivity, network instability, and ultimately improper information processing. Lastly, we highlight several pathological conditions related to sensory deafferentation and describe how, in some cases, homeostatic compensation without appropriate sensory inputs can result in phantom perceptions.
Collapse
|
18
|
Marquez-Legorreta E, Constantin L, Piber M, Favre-Bulle IA, Taylor MA, Blevins AS, Giacomotto J, Bassett DS, Vanwalleghem GC, Scott EK. Brain-wide visual habituation networks in wild type and fmr1 zebrafish. Nat Commun 2022; 13:895. [PMID: 35173170 PMCID: PMC8850451 DOI: 10.1038/s41467-022-28299-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
Habituation is a form of learning during which animals stop responding to repetitive stimuli, and deficits in habituation are characteristic of several psychiatric disorders. Due to technical challenges, the brain-wide networks mediating habituation are poorly understood. Here we report brain-wide calcium imaging during larval zebrafish habituation to repeated visual looming stimuli. We show that different functional categories of loom-sensitive neurons are located in characteristic locations throughout the brain, and that both the functional properties of their networks and the resulting behavior can be modulated by stimulus saliency and timing. Using graph theory, we identify a visual circuit that habituates minimally, a moderately habituating midbrain population proposed to mediate the sensorimotor transformation, and downstream circuit elements responsible for higher order representations and the delivery of behavior. Zebrafish larvae carrying a mutation in the fmr1 gene have a systematic shift toward sustained premotor activity in this network, and show slower behavioral habituation.
Collapse
Affiliation(s)
- Emmanuel Marquez-Legorreta
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Lena Constantin
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Marielle Piber
- School of Medicine, Medical Sciences, and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Itia A Favre-Bulle
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,School of Mathematics and Physics, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Michael A Taylor
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ann S Blevins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean Giacomotto
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Queensland Centre for Mental Health Research, West Moreton Hospital and Health Service, Wacol, QLD, 4076, Australia.,Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.,Discovery Biology, Griffith University, Brisbane, QLD, 4111, Australia
| | - Dani S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Departments of Electrical & Systems Engineering, Physics & Astronomy, Neurology, Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Santa Fe Institute, Santa Fe, NM, 87501, USA
| | - Gilles C Vanwalleghem
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| | - Ethan K Scott
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
19
|
Chung A, Jou C, Grau-Perales A, Levy E, Dvorak D, Hussain N, Fenton AA. Cognitive control persistently enhances hippocampal information processing. Nature 2021; 600:484-488. [PMID: 34759316 PMCID: PMC8872635 DOI: 10.1038/s41586-021-04070-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 09/29/2021] [Indexed: 01/30/2023]
Abstract
Could learning that uses cognitive control to judiciously use relevant information while ignoring distractions generally improve brain function, beyond forming explicit memories? According to a neuroplasticity hypothesis for how some cognitive behavioural therapies are effective, cognitive control training (CCT) changes neural circuit information processing1-3. Here we investigated whether CCT persistently alters hippocampal neural circuit function. We show that mice learned and remembered a conditioned place avoidance during CCT that required ignoring irrelevant locations of shock. CCT facilitated learning new tasks in novel environments for several weeks, relative to unconditioned controls and control mice that avoided the same place during reduced distraction. CCT rapidly changes entorhinal cortex-to-dentate gyrus synaptic circuit function, resulting in an excitatory-inhibitory subcircuit change that persists for months. CCT increases inhibition that attenuates the dentate response to medial entorhinal cortical input, and through disinhibition, potentiates the response to strong inputs, pointing to overall signal-to-noise enhancement. These neurobiological findings support the neuroplasticity hypothesis that, as well as storing item-event associations, CCT persistently optimizes neural circuit information processing.
Collapse
Affiliation(s)
- Ain Chung
- Center for Neural Science, New York University
| | - Claudia Jou
- Department of Psychology, Hunter College, City University of New York
| | | | - Eliott Levy
- Center for Neural Science, New York University
| | - Dino Dvorak
- Center for Neural Science, New York University
| | | | - André A. Fenton
- Center for Neural Science, New York University,Neuroscience Institute at the NYU Langone Medical Center
| |
Collapse
|
20
|
Contractor A, Ethell IM, Portera-Cailliau C. Cortical interneurons in autism. Nat Neurosci 2021; 24:1648-1659. [PMID: 34848882 PMCID: PMC9798607 DOI: 10.1038/s41593-021-00967-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023]
Abstract
The mechanistic underpinnings of autism remain a subject of debate and controversy. Why do individuals with autism share an overlapping set of atypical behaviors and symptoms, despite having different genetic and environmental risk factors? A major challenge in developing new therapies for autism has been the inability to identify convergent neural phenotypes that could explain the common set of symptoms that result in the diagnosis. Although no striking macroscopic neuropathological changes have been identified in autism, there is growing evidence that inhibitory interneurons (INs) play an important role in its neural basis. In this Review, we evaluate and interpret this evidence, focusing on recent findings showing reduced density and activity of the parvalbumin class of INs. We discuss the need for additional studies that investigate how genes and the environment interact to change the developmental trajectory of INs, permanently altering their numbers, connectivity and circuit engagement.
Collapse
Affiliation(s)
- Anis Contractor
- Department of Neuroscience Feinberg School of Medicine, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, UC Riverside School of Medicine, Riverside, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
21
|
Dvorak D, Chung A, Park EH, Fenton AA. Dentate spikes and external control of hippocampal function. Cell Rep 2021; 36:109497. [PMID: 34348165 PMCID: PMC8369486 DOI: 10.1016/j.celrep.2021.109497] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 11/11/2022] Open
Abstract
Mouse hippocampus CA1 place-cell discharge typically encodes current location, but during slow gamma dominance (SGdom), when SG oscillations (30-50 Hz) dominate mid-frequency gamma oscillations (70-90 Hz) in CA1 local field potentials, CA1 discharge switches to represent distant recollected locations. We report that dentate spike type 2 (DSM) events initiated by medial entorhinal cortex II (MECII)→ dentate gyrus (DG) inputs promote SGdom and change excitation-inhibition coordinated discharge in DG, CA3, and CA1, whereas type 1 (DSL) events initiated by lateral entorhinal cortex II (LECII)→DG inputs do not. Just before SGdom, LECII-originating SG oscillations in DG and CA3-originating SG oscillations in CA1 phase and frequency synchronize at the DSM peak when discharge within DG and CA3 increases to promote excitation-inhibition cofiring within and across the DG→CA3→CA1 pathway. This optimizes discharge for the 5-10 ms DG-to-CA1 neuro-transmission that SGdom initiates. DSM properties identify extrahippocampal control of SGdom and a cortico-hippocampal mechanism that switches between memory-related modes of information processing.
Collapse
Affiliation(s)
- Dino Dvorak
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Ain Chung
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Eun Hye Park
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - André Antonio Fenton
- Center for Neural Science, New York University, New York, NY 10003, USA; Neuroscience Institute at the NYU Langone Medical Center, New York, NY 10003, USA.
| |
Collapse
|
22
|
Booker SA, Kind PC. Mechanisms regulating input-output function and plasticity of neurons in the absence of FMRP. Brain Res Bull 2021; 175:69-80. [PMID: 34245842 DOI: 10.1016/j.brainresbull.2021.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/13/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
The function of brain circuits relies on high-fidelity information transfer within neurons. Synaptic inputs arrive primarily at dendrites, where they undergo integration and summation throughout the somatodendritic domain, ultimately leading to the generation of precise patterns of action potentials. Emerging evidence suggests that the ability of neurons to transfer synaptic information and modulate their output is impaired in a number of neurodevelopmental disorders including Fragile X Syndrome. In this review we summarise recent findings that have revealed the pathophysiological and plasticity mechanisms that alter the ability of neurons in sensory and limbic circuits to reliably code information in the absence of FMRP. We examine which aspects of this transform may result directly from the loss of FMRP and those that a result from compensatory or homeostatic alterations to neuronal function. Dissection of the mechanisms leading to altered input-output function of neurons in the absence of FMRP and their effects on regulating neuronal plasticity throughout development could have important implications for potential therapies for Fragile X Syndrome, including directing the timing and duration of different treatment options.
Collapse
Affiliation(s)
- Sam A Booker
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK; Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Peter C Kind
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK; Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK; National Centre for Biological Sciences (NCBS), Bangalore, India.
| |
Collapse
|
23
|
Coding of social novelty in the hippocampal CA2 region and its disruption and rescue in a 22q11.2 microdeletion mouse model. Nat Neurosci 2020; 23:1365-1375. [PMID: 33077947 PMCID: PMC8861630 DOI: 10.1038/s41593-020-00720-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/04/2020] [Indexed: 12/25/2022]
Abstract
The hippocampal CA2 region is essential for social memory. To determine whether CA2 activity encodes social interactions, we recorded from CA2 pyramidal neurons in male mice during social behavior. While CA2 neuronal firing showed only weak spatial selectivity, it accurately encoded contextual changes and distinguished between a novel and familiar mouse. In the Df(16)A+/− mouse model of the human 22q11.2 microdeletion, which confers a 30-fold increased risk of schizophrenia, CA2 social coding was impaired, consistent with the social memory deficit observed in these mice; in contrast, spatial coding accuracy was greatly enhanced. CA2 pyramidal neurons were previously found to be hyperpolarized in Df(16)A+/− mice, likely due to upregulation of TREK-1 K+ current. We found that TREK-1 blockade rescued social memory and CA2 social coding in Df(16)A+/− mice, supporting a crucial role for CA2 in the normal encoding of social stimuli and in social behavioral dysfunction in disease.
Collapse
|
24
|
Mansur F, Alarcon JM, Stackpole EE, Wang R, Richter JD. Noncanonical cytoplasmic poly(A) polymerases regulate RNA levels, alternative RNA processing, and synaptic plasticity but not hippocampal-dependent behaviours. RNA Biol 2020; 18:962-971. [PMID: 32954964 DOI: 10.1080/15476286.2020.1824061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Noncanonical poly(A) polymerases are frequently tethered to mRNA 3' untranslated regions and regulate poly(A) tail length and resulting translation. In the brain, one such poly(A) polymerase is Gld2, which is anchored to mRNA by the RNA-binding protein CPEB1 to control local translation at postsynaptic regions. Depletion of CPEB1 or Gld2 from the mouse hippocampus results in a deficit in long-term potentiation (LTP), but only depletion of CPEB1 alters animal behaviour. To test whether a related enzyme, Gld4, compensates for the lack of Gld2, we separately or simultaneously depleted both proteins from hippocampal area CA1 and again found little change in animal behaviour, but observed a deficit in LTP as well as an increase in long-term depression (LTD), two forms of protein synthesis-dependent synaptic plasticity. RNA-seq data from Gld2, Gld4, and Gld2/Gld4-depleted hippocampus show widespread changes in steady state RNA levels, alternative splicing, and alternative poly(A) site selection. Many of the RNAs subject to these alterations encode proteins that mediate synaptic function, suggesting a molecular foundation for impaired synaptic plasticity.
Collapse
Affiliation(s)
- Fernanda Mansur
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Juan Marcos Alarcon
- Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Emily E Stackpole
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ruijia Wang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Joel D Richter
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
25
|
Yang J, Han W, Liu J, Yang C, Zhao WJ, Sun H, Pan YN, Chen HS, Cheng L, Jiang L. [Effect of advanced maternal age on development of hippocampal neural stem cells in offspring rats]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:1017-1026. [PMID: 32933637 PMCID: PMC7499440 DOI: 10.7499/j.issn.1008-8830.2003213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/05/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To study the effect of advanced maternal age (AMA) on the development of hippocampal neural stem cells in offspring rats. METHODS Ten 3-month-old and ten 12-month-old female Sprague-Dawley rats were housed individually with 3-month-old male rats (1:1, n=20), whose offspring rats were assigned to a control group and an AMA group. A total of 40 rats were randomly selected from each group. Immunofluorescence assay and Western blot were used to localize and determine the levels of protein expression of Nestin and doublecortin (DCX) on day 7 as well as neuronal nuclear antigen (NeuN) and glial fibrillary acidic protein (GFAP) on day 28 (n=8 for each marker). Immunofluorescence assay was also used to localize the hippocampal expression of polysialylated isoforms of neural cell adhesion molecule (PSA-NCAM) on day 14 (n=8 for each marker). RESULTS According to the Western blot results, the AMA group had significantly lower protein expression of DCX than the control group (P<0.05), while there were no significant differences in the protein expression of Nestin, NeuN, and GFAP between the two groups (P>0.05). According to the results of immunofluorescence assay, the AMA group had significantly lower protein expression of Nestin, DCX, and PSA-NCAM in the hippocampal dentate gyrus (DG) region than the control group (P<0.05), while there were no significant differences in the above indices in the hippocampal CA1 and CA3 regions between the two groups (P>0.05). The AMA group had significantly higher expression of NeuN in the hippocampal CA1 region than the control group (P<0.01), while there were no significant differences in the expression of NeuN in the hippocampal DG and CA3 regions between the two groups (P>0.05). The AMA group had significantly lower expression of GFAP in the hippocampal CA1, CA3, and DG regions than the control group (P<0.05). CONCLUSIONS AMA may cause inhibition of proliferation, survival, and migration of hippocampal neural stem cells. AMA may also affect their differentiation into neurons and astrocytes, which will eventually lead to developmental disorders of hippocampal neural stem cells in offspring rats.
Collapse
Affiliation(s)
- Jing Yang
- Department of Neurology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Stefanini F, Kushnir L, Jimenez JC, Jennings JH, Woods NI, Stuber GD, Kheirbek MA, Hen R, Fusi S. A Distributed Neural Code in the Dentate Gyrus and in CA1. Neuron 2020; 107:703-716.e4. [PMID: 32521223 PMCID: PMC7442694 DOI: 10.1016/j.neuron.2020.05.022] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/01/2020] [Accepted: 05/15/2020] [Indexed: 01/28/2023]
Abstract
Neurons are often considered specialized functional units that encode a single variable. However, many neurons are observed to respond to a mix of disparate sensory, cognitive, and behavioral variables. For such representations, information is distributed across multiple neurons. Here we find this distributed code in the dentate gyrus and CA1 subregions of the hippocampus. Using calcium imaging in freely moving mice, we decoded an animal's position, direction of motion, and speed from the activity of hundreds of cells. The response properties of individual neurons were only partially predictive of their importance for encoding position. Non-place cells encoded position and contributed to position encoding when combined with other cells. Indeed, disrupting the correlations between neural activities decreased decoding performance, mostly in CA1. Our analysis indicates that population methods rather than classical analyses based on single-cell response properties may more accurately characterize the neural code in the hippocampus.
Collapse
Affiliation(s)
- Fabio Stefanini
- Center for Theoretical Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Lyudmila Kushnir
- GNT-LNC, Départment d'Études Cognitives, École Normale Supérieure, INSERM, PSL Research University, 75005 Paris, France
| | - Jessica C Jimenez
- Departments of Neuroscience, Psychiatry, & Pharmacology, Columbia University, New York, NY, USA; Division of Integrative Neuroscience, Department of Psychiatry, New York State Psychiatric Institute, New York, NY, USA
| | - Joshua H Jennings
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Nicholas I Woods
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Mazen A Kheirbek
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| | - René Hen
- Departments of Neuroscience, Psychiatry, & Pharmacology, Columbia University, New York, NY, USA; Division of Integrative Neuroscience, Department of Psychiatry, New York State Psychiatric Institute, New York, NY, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA.
| | - Stefano Fusi
- Center for Theoretical Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA.
| |
Collapse
|
27
|
Booker SA, Simões de Oliveira L, Anstey NJ, Kozic Z, Dando OR, Jackson AD, Baxter PS, Isom LL, Sherman DL, Hardingham GE, Brophy PJ, Wyllie DJ, Kind PC. Input-Output Relationship of CA1 Pyramidal Neurons Reveals Intact Homeostatic Mechanisms in a Mouse Model of Fragile X Syndrome. Cell Rep 2020; 32:107988. [PMID: 32783927 PMCID: PMC7435362 DOI: 10.1016/j.celrep.2020.107988] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 04/01/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
Cellular hyperexcitability is a salient feature of fragile X syndrome animal models. The cellular basis of hyperexcitability and how it responds to changing activity states is not fully understood. Here, we show increased axon initial segment length in CA1 of the Fmr1-/y mouse hippocampus, with increased cellular excitability. This change in length does not result from reduced AIS plasticity, as prolonged depolarization induces changes in AIS length independent of genotype. However, depolarization does reduce cellular excitability, the magnitude of which is greater in Fmr1-/y neurons. Finally, we observe reduced functional inputs from the entorhinal cortex, with no genotypic difference in the firing rates of CA1 pyramidal neurons. This suggests that AIS-dependent hyperexcitability in Fmr1-/y mice may result from adaptive or homeostatic regulation induced by reduced functional synaptic connectivity. Thus, while AIS length and intrinsic excitability contribute to cellular hyperexcitability, they may reflect a homeostatic mechanism for reduced synaptic input onto CA1 neurons.
Collapse
Affiliation(s)
- Sam A. Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Corresponding author
| | - Laura Simões de Oliveira
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK
| | - Natasha J. Anstey
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Zrinko Kozic
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Owen R. Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Dementia Research Institute, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Adam D. Jackson
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Paul S. Baxter
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Dementia Research Institute, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-5632, USA
| | - Diane L. Sherman
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Giles E. Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Dementia Research Institute, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Peter J. Brophy
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - David J.A. Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Peter C. Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India,Corresponding author
| |
Collapse
|
28
|
Barry JM, Mahoney JM, Holmes GL. Coordination of hippocampal theta and gamma oscillations relative to spatial active avoidance reflects cognitive outcome after febrile status epilepticus. Behav Neurosci 2020; 134:562-576. [PMID: 32628031 DOI: 10.1037/bne0000388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cognitive deficits may arise from a variety of genetic alterations and neurological insults that impair neural coding mechanisms and the routing of neural information underpinning learning and memory. Slow and medium gamma oscillations underpin memory recall and sensorimotor processing and represent dynamic inputs at CA1 synapses. Febrile status epilepticus (FSE) can lead to increased risk for temporal lobe epilepsy and enduring cognitive impairments. In a rodent model, we assessed how FSE alters hippocampal CA1 signals relative to spatial task performance and serve as a readout of synaptic input efficacy. The power of theta (5-12 Hz), slow gamma (30-50 Hz), and medium gamma (70-90 Hz) differentially interact with respect to cognitive demands during active avoidance behavior on a rotating arena. Successful avoidance was characterized by slow gamma that was largest several seconds before or after peak acceleration. Peak acceleration coincides with peak theta oscillations, followed within approximately 1 s by peak medium gamma. FSE animals showing impairment in the task maintained the profiles of theta and medium gamma associated with increased sensorimotor processing following peak acceleration but did not exhibit the same slow gamma profile associated with epochs of memory retrieval. While CA1 synapses from entorhinal cortex were functionally unaffected by FSE, communication via synapses from CA3 may have been impaired, leading to both temporal discoordination and poor memory retrieval. These findings demonstrate theta/gamma profiles can serve as both physiological biomarkers for memory retrieval or encoding deficits and synapse level treatment targets that could attenuate cognitive comorbidities associated with early life seizures. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
|
29
|
Jonak CR, Lovelace JW, Ethell IM, Razak KA, Binder DK. Multielectrode array analysis of EEG biomarkers in a mouse model of Fragile X Syndrome. Neurobiol Dis 2020; 138:104794. [PMID: 32036032 DOI: 10.1016/j.nbd.2020.104794] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/27/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety and social and sensory processing deficits. Recent EEG studies in humans with FXS have identified neural oscillation deficits that include increased resting state gamma power, increased amplitude of auditory evoked potentials, and reduced inter-trial phase coherence of sound-evoked gamma oscillations. Identification of comparable EEG biomarkers in mouse models of FXS could facilitate the pre-clinical to clinical therapeutic pipeline. However, while human EEG studies have involved 128-channel scalp EEG acquisition, no mouse studies have been performed with more than three EEG channels. In the current study, we employed a recently developed 30-channel mouse multielectrode array (MEA) system to record and analyze resting and stimulus-evoked EEG signals in WT vs. Fmr1 KO mice. Using this system, we now report robust MEA-derived phenotypes including higher resting EEG power, altered event-related potentials (ERPs) and reduced inter-trial phase coherence to auditory chirp stimuli in Fmr1 KO mice that are remarkably similar to those reported in humans with FXS. We propose that the MEA system can be used for: (i) derivation of higher-level EEG parameters; (ii) EEG biomarkers for drug testing; and (ii) mechanistic studies of FXS pathophysiology.
Collapse
Affiliation(s)
- Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America
| | - Jonathan W Lovelace
- Department of Psychology, University of California, Riverside, United States of America
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America; Neuroscience Graduate Program, University of California, Riverside, United States of America
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, United States of America; Department of Psychology, University of California, Riverside, United States of America
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America; Neuroscience Graduate Program, University of California, Riverside, United States of America.
| |
Collapse
|
30
|
Seshadri S, Hoeppner DJ, Tajinda K. Calcium Imaging in Drug Discovery for Psychiatric Disorders. Front Psychiatry 2020; 11:713. [PMID: 32793004 PMCID: PMC7390878 DOI: 10.3389/fpsyt.2020.00713] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/06/2020] [Indexed: 12/31/2022] Open
Abstract
The past 5 years have seen a sharp increase in the number of studies using calcium imaging in behaving rodents. These studies have helped identify important roles for individual cells, brain regions, and circuits in some of the core behavioral phenotypes of psychiatric disorders, such as schizophrenia and autism, and have characterized network dysfunction in well-established models of these disorders. Since rescuing clinically relevant behavioral deficits in disease model mice remains a foundation of preclinical CNS research, these studies have the potential to inform new therapeutic approaches targeting specific cell types or projections, or perhaps most importantly, the network-level context in which neurons function. In this mini-review, we will provide a brief overview of recent insights into psychiatric disease-associated mouse models and behavior paradigms, focusing on those achieved by cellular resolution imaging of calcium dynamics in neural populations. We will then discuss how these experiments can support efforts within the pharmaceutical industry, such as target identification, assay development, and candidate screening and validation. Calcium imaging is uniquely capable of bridging the gap between two of the key resources that currently enable CNS drug discovery: genomic and transcriptomic data from human patients, and translatable, population-resolution measures of brain activity (such as fMRI and EEG). Applying this knowledge could yield real value to patients in the near future.
Collapse
Affiliation(s)
- Saurav Seshadri
- Neuroscience, La Jolla Laboratory, Astellas Research Institute of America LLC, San Diego, CA, United States
| | - Daniel J Hoeppner
- Neuroscience, La Jolla Laboratory, Astellas Research Institute of America LLC, San Diego, CA, United States
| | - Katsunori Tajinda
- Neuroscience, La Jolla Laboratory, Astellas Research Institute of America LLC, San Diego, CA, United States
| |
Collapse
|
31
|
Sawicka K, Hale CR, Park CY, Fak JJ, Gresack JE, Van Driesche SJ, Kang JJ, Darnell JC, Darnell RB. FMRP has a cell-type-specific role in CA1 pyramidal neurons to regulate autism-related transcripts and circadian memory. eLife 2019; 8:e46919. [PMID: 31860442 PMCID: PMC6924960 DOI: 10.7554/elife.46919] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Loss of the RNA binding protein FMRP causes Fragile X Syndrome (FXS), the most common cause of inherited intellectual disability, yet it is unknown how FMRP function varies across brain regions and cell types and how this contributes to disease pathophysiology. Here we use conditional tagging of FMRP and CLIP (FMRP cTag CLIP) to examine FMRP mRNA targets in hippocampal CA1 pyramidal neurons, a critical cell type for learning and memory relevant to FXS phenotypes. Integrating these data with analysis of ribosome-bound transcripts in these neurons revealed CA1-enriched binding of autism-relevant mRNAs, and CA1-specific regulation of transcripts encoding circadian proteins. This contrasted with different targets in cerebellar granule neurons, and was consistent with circadian defects in hippocampus-dependent memory in Fmr1 knockout mice. These findings demonstrate differential FMRP-dependent regulation of mRNAs across neuronal cell types that may contribute to phenotypes such as memory defects and sleep disturbance associated with FXS.
Collapse
Affiliation(s)
- Kirsty Sawicka
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Caryn R Hale
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Christopher Y Park
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - John J Fak
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Jodi E Gresack
- Laboratory of Molecular and Cellular NeuroscienceThe Rockefeller UniversityNew YorkUnited States
| | - Sarah J Van Driesche
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Jin Joo Kang
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Jennifer C Darnell
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Robert B Darnell
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| |
Collapse
|
32
|
Impaired Reliability and Precision of Spiking in Adults But Not Juveniles in a Mouse Model of Fragile X Syndrome. eNeuro 2019; 6:ENEURO.0217-19.2019. [PMID: 31685673 PMCID: PMC6917895 DOI: 10.1523/eneuro.0217-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/26/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common source of intellectual disability and autism. Extensive studies have been performed on the network and behavioral correlates of the syndrome, but our knowledge about intrinsic conductance changes is still limited. In this study, we show a differential effect of FMRP knockout in different subsections of hippocampus using whole-cell patch clamp in mouse hippocampal slices. We observed no significant change in spike numbers in the CA1 region of hippocampus, but a significant increase in CA3, in juvenile mice. However, in adult mice we see a reduction in spike number in the CA1 with no significant difference in CA3. In addition, we see increased variability in spike numbers in CA1 cells following a variety of steady and modulated current step protocols. This effect emerges in adult mice (8 weeks) but not juvenile mice (4 weeks). This increased spiking variability was correlated with reduced spike number and with elevated AHP. The increased AHP arose from elevated SK currents (small conductance calcium-activated potassium channels), but other currents involved in medium AHP, such as Ih and M, were not significantly different. We obtained a partial rescue of the cellular variability phenotype when we blocked SK current using the specific blocker apamin. Our observations provide a single-cell correlate of the network observations of response variability and loss of synchronization, and suggest that the elevation of SK currents in FXS may provide a partial mechanistic explanation for this difference.
Collapse
|
33
|
Monsalve‐Mercado MM, Roudi Y. Hippocampal spike‐time correlations and place field overlaps during open field foraging. Hippocampus 2019; 30:354-366. [DOI: 10.1002/hipo.23173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/05/2019] [Accepted: 10/08/2019] [Indexed: 11/05/2022]
Affiliation(s)
- Mauro M. Monsalve‐Mercado
- Physik‐Department Technische Universitat Munchen Munich Germany
- Center for Theoretical Neuroscience Zuckerman Institute, Columbia University New York New York
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU Trondheim Norway
| | - Yasser Roudi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU Trondheim Norway
| |
Collapse
|
34
|
Long-Lasting Input-Specific Experience-Dependent Changes of Hippocampus Synaptic Function Measured in the Anesthetized Rat. eNeuro 2019; 6:ENEURO.0506-18.2019. [PMID: 31434661 PMCID: PMC6731537 DOI: 10.1523/eneuro.0506-18.2019] [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: 12/24/2018] [Revised: 08/05/2019] [Accepted: 08/10/2019] [Indexed: 11/21/2022] Open
Abstract
How experience causes long-lasting changes in the brain is a central question in neuroscience. The common view is that synaptic function is altered by experience to change brain circuit functions that underlie conditioned behavior. We examined hippocampus synaptic circuit function in vivo, in three groups of animals, to assess the impact of experience on hippocampus function in rats. The “conditioned” group acquired a shock-conditioned place response during a cognitively-challenging, hippocampus synaptic plasticity-dependent task. The no-shock group had similar exposure to the environmental conditions but no conditioning. The home-cage group was experimentally naive. After the one-week retention test, under anesthesia, we stimulated the perforant path inputs to CA1, which terminate in stratum lacunosum moleculare (slm), and to the dentate gyrus (DG), which terminate in the molecular layer. We find synaptic compartment specific changes that differ amongst the groups. The evoked field EPSP (fEPSP) and pre-spike field response are enhanced only at the DG input layer and only in conditioned animals. The DG responses, measured by the population spiking activity and post-spike responses, are enhanced in both the conditioned and no-shock groups compared to home-cage animals. These changes are pathway specific because no differences are observed in slm of CA1. These findings demonstrate long-term, experience-dependent, pathway-specific alterations to synaptic circuit function of the hippocampus.
Collapse
|
35
|
O'Reilly KC, Perica MI, Fenton AA. Synaptic plasticity/dysplasticity, process memory and item memory in rodent models of mental dysfunction. Schizophr Res 2019; 207:22-36. [PMID: 30174252 PMCID: PMC6395534 DOI: 10.1016/j.schres.2018.08.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 08/14/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022]
Abstract
Activity-dependent changes in the effective connection strength of synapses are a fundamental feature of a nervous system. This so-called synaptic plasticity is thought to underlie storage of information in memory and has been hypothesized to be crucial for the effects of cognitive behavioral therapy. Synaptic plasticity stores information in a neural network, creating a trace of neural activity from past experience. The plasticity can also change the behavior of the network so the network can differentially transform/compute information in future activations. We discuss these two related but separable functions of synaptic plasticity; one we call "item memory" as it represents and stores items of information in memory, the other we call "process memory" as it encodes and stores functions such as computations to modify network information processing capabilities. We review evidence of item and process memory operations in behavior and evidence that experience modifies the brain's functional networks. We discuss neurodevelopmental rodent models relevant for understanding mental illness and compare two models in which one model, neonatal ventral hippocampal lesion (NVHL) has beneficial adult outcomes after being exposed to an adolescent cognitive experience that is potentially similar to cognitive behavioral therapy. The other model, gestational day 17 methylazoxymethanol acetate (GD17-MAM), does not benefit from the same adolescent cognitive experience. We propose that process memory is altered by early cognitive experience in NVHL rats but not in GD17-MAM rats, and discuss how dysplasticity factors may contribute to the differential adult outcomes after early cognitive experience in the NVHL and MAM models.
Collapse
Affiliation(s)
- Kally C O'Reilly
- Center for Neural Science, New York University, New York, NY 10003, USA.
| | - Maria I Perica
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - André A Fenton
- Center for Neural Science, New York University, New York, NY 10003, USA; Neuroscience Institute at the New York University Langone Medical Center, New York, NY 10016, USA; Department of Physiology & Pharmacology, Robert F. Furchgott Center for Neuroscience, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA.
| |
Collapse
|
36
|
O'Reilly KC, Levy ERJ, Patino AV, Perica MI, Fenton AA. Sub-circuit alterations in dorsal hippocampus structure and function after global neurodevelopmental insult. Brain Struct Funct 2018; 223:3543-3556. [PMID: 29951917 PMCID: PMC6278823 DOI: 10.1007/s00429-018-1704-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 06/20/2018] [Indexed: 11/30/2022]
Abstract
Patients with neuropsychiatric and neurological disorders often express limbic circuit abnormalities and deficits in information processing. While these disorders appear to have diverse etiologies, their common features suggest neurodevelopmental origins. Neurodevelopment is a prolonged process of diverse events including neurogenesis/apoptosis, axon pathfinding, synaptogenesis, and pruning, to name a few. The precise timing of the neurodevelopmental insult to these processes likely determines the resulting functional outcome. We used the epilepsy and schizophrenia-related gestational day 17 methylazoxymethanol acetate model to examine the impact of this timed neurodevelopmental insult on principal cell morphology and synaptic network function of the dorsal hippocampus (dHPC) circuit. Our observed structural and functional alterations in dHPC are compartment specific, indicating that adverse global exposure during gestation can produce specific alterations and distort information processing in neural circuits that underlie cognitive abilities.
Collapse
Affiliation(s)
- Kally C O'Reilly
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, 10003, USA.
- Child and Adolescent Psychiatry, New York State Psychiatric Institute, 1051 Riverside Dr, New York, NY, 10032, USA.
| | - Eliott R J Levy
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, 10003, USA
| | - Alejandra V Patino
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, 10003, USA
| | - Maria I Perica
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, 10003, USA
| | - André A Fenton
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, 10003, USA.
- Neuroscience Institute at the New York University Langone Medical Center, New York, NY, 10016, USA.
- Department of Physiology and Pharmacology, Robert F. Furchgott Center for Neuroscience, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
| |
Collapse
|
37
|
Mably AJ, Colgin LL. Gamma oscillations in cognitive disorders. Curr Opin Neurobiol 2018; 52:182-187. [PMID: 30121451 DOI: 10.1016/j.conb.2018.07.009] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/18/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023]
Abstract
Gamma oscillations (∼25-100 Hz) are believed to play a role in cognition. Accordingly, aberrant gamma oscillations have been observed in several cognitive disorders, including Alzheimer's disease and Fragile X syndrome. Here, we review how recent results showing abnormal gamma rhythms in Alzheimer's disease and Fragile X syndrome help reveal links between cellular disturbances and cognitive impairments. We also discuss how gamma results from rodent models of Alzheimer's disease and Fragile X syndrome may provide insights about unique functions of distinct slow (∼25-50 Hz) and fast gamma (∼55-100 Hz) subtypes. Finally, we consider studies employing brain stimulation paradigms in Alzheimer's disease and discuss how such studies may reveal causal relationships between gamma impairments and memory disturbances.
Collapse
Affiliation(s)
- Alexandra J Mably
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, 1 University Station Stop C7000, Austin, TX 78712, USA
| | - Laura Lee Colgin
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, 1 University Station Stop C7000, Austin, TX 78712, USA.
| |
Collapse
|
38
|
The cognitive nuances of surprising events: exposure to unexpected stimuli elicits firing variations in neurons of the dorsal CA1 hippocampus. Brain Struct Funct 2018; 223:3183-3211. [PMID: 29789932 PMCID: PMC6132666 DOI: 10.1007/s00429-018-1681-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 05/08/2018] [Indexed: 12/02/2022]
Abstract
The ability to recognize novel situations is among the most fascinating and vital of the brain functions. A hypothesis posits that encoding of novelty is prompted by failures in expectancy, according to computation matching incoming information with stored events. Thus, unexpected changes in context are detected within the hippocampus and transferred to downstream structures, eliciting the arousal of the dopamine system. Nevertheless, the precise locus of detection is a matter of debate. The dorsal CA1 hippocampus (dCA1) appears as an ideal candidate for operating a mismatch computation and discriminating the occurrence of diverse stimuli within the same environment. In this study, we sought to determine dCA1 neuronal firing during the experience of novel stimuli embedded in familiar contexts. We performed population recordings while head-fixed mice navigated virtual environments. Three stimuli were employed, namely a novel pattern of visual cues, an odor, and a reward with enhanced valence. The encounter of unexpected events elicited profound variations in dCA1 that were assessed both as opposite rate directions and altered network connectivity. When experienced in sequence, novel stimuli elicited specific responses that often exhibited cross-sensitization. Short-latency, event-triggered responses were in accordance with the detection of novelty being computed within dCA1. We postulate that firing variations trigger neuronal disinhibition, and constitute a fundamental mechanism in the processing of unexpected events and in learning. Elucidating the mechanisms underlying detection and computation of novelty might help in understanding hippocampal-dependent cognitive dysfunctions associated with neuropathologies and psychiatric conditions.
Collapse
|
39
|
On How the Dentate Gyrus Contributes to Memory Discrimination. Neuron 2018; 98:832-845.e5. [PMID: 29731252 DOI: 10.1016/j.neuron.2018.04.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 02/13/2018] [Accepted: 04/16/2018] [Indexed: 11/20/2022]
Abstract
The dentate gyrus (DG) is crucial for behaviorally discriminating similar spatial memories, predicting that DG place cells change ("remap") their relative spatial tuning ("place fields") for memory discrimination. This prediction was never tested, although DG place cells remap across similar environments without memory tasks. We confirm this prior finding but find that DG place fields do not remap across spatial tasks that require DG-dependent memory discrimination. Instead of remapping, place-discriminating discharge is observed transiently among DG place cells, particularly when memory discrimination is most necessary. The DG network may signal memory discrimination by expressing distinctive sub-second network patterns of co-firing at memory discrimination sites. This involves increased coupling of discharge from place cells and interneurons, as was observed during successful, but not failed, behavioral expression of memory discrimination. Instead of remapping, these findings indicate that memory discrimination is signaled by sub-second patterns of correlated discharge within the dentate network.
Collapse
|
40
|
Dvorak D, Radwan B, Sparks FT, Talbot ZN, Fenton AA. Control of recollection by slow gamma dominating mid-frequency gamma in hippocampus CA1. PLoS Biol 2018; 16:e2003354. [PMID: 29346381 PMCID: PMC5790293 DOI: 10.1371/journal.pbio.2003354] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/30/2018] [Accepted: 12/28/2017] [Indexed: 11/19/2022] Open
Abstract
Behavior is used to assess memory and cognitive deficits in animals like Fmr1-null mice that model Fragile X Syndrome, but behavior is a proxy for unknown neural events that define cognitive variables like recollection. We identified an electrophysiological signature of recollection in mouse dorsal Cornu Ammonis 1 (CA1) hippocampus. During a shocked-place avoidance task, slow gamma (SG) (30–50 Hz) dominates mid-frequency gamma (MG) (70–90 Hz) oscillations 2–3 s before successful avoidance, but not failures. Wild-type (WT) but not Fmr1-null mice rapidly adapt to relocating the shock; concurrently, SG/MG maxima (SGdom) decrease in WT but not in cognitively inflexible Fmr1-null mice. During SGdom, putative pyramidal cell ensembles represent distant locations; during place avoidance, these are avoided places. During shock relocation, WT ensembles represent distant locations near the currently correct shock zone, but Fmr1-null ensembles represent the formerly correct zone. These findings indicate that recollection occurs when CA1 SG dominates MG and that accurate recollection of inappropriate memories explains Fmr1-null cognitive inflexibility. Behavior is often used as proxy to study memory and cognitive deficits in animals like Fmr1-KO mice that model Fragile X Syndrome, the most prevalent single-gene cause of intellectual disability and autism. However, it is unclear what neural events define cognitive variables like recollection of memory and cognitive inflexibility. We identified a signature of recollection in the local field potentials of mouse dorsal CA1 hippocampus. When mice on a rotating platform avoided an invisible, fixed shock zone, slow gamma (30–50 Hz) oscillations dominated mid-frequency gamma (70–90 Hz) oscillations (SGdom) 2–3 s before mice successfully avoided the shock zone. Wild-type but not Fmr1-KO mice adapt to relocating the shock zone; concurrently, SGdom decreases in wild-type but not in cognitively inflexible Fmr1-KO mice. During SGdom, principal cell ensembles represent distant locations; during place avoidance, these are avoided places in the shock zone vicinity. During shock relocation, wild-type ensembles encode distant locations near the currently correct shock zone, but Fmr1-KO ensembles manifest representational inflexibility, encoding the formerly correct zone. These findings suggest evidence for competition amongst CA1 inputs for CA1 information-processing modes and indicate that recollection occurs when CA1 slow gamma dominates mid-frequency gamma and that accurate recollection of inappropriate memories explains Fmr1-KO cognitive inflexibility.
Collapse
Affiliation(s)
- Dino Dvorak
- Center for Neural Science, New York University, New York, New York, United States of America
| | - Basma Radwan
- Center for Neural Science, New York University, New York, New York, United States of America
| | - Fraser T. Sparks
- Center for Neural Science, New York University, New York, New York, United States of America
| | - Zoe Nicole Talbot
- School of Medicine, New York University, New York, New York, United States of America
| | - André A. Fenton
- Center for Neural Science, New York University, New York, New York, United States of America
- Neuroscience Institute at the New York University Langone Medical Center, New York, New York, United States of America
- Department of Physiology and Pharmacology, The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
- * E-mail:
| |
Collapse
|