1
|
Paulson AL, Zhang L, Prichard AM, Singer AC. 40 Hz sensory stimulation enhances CA3-CA1 coordination and prospective coding during navigation in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619408. [PMID: 39484571 PMCID: PMC11526945 DOI: 10.1101/2024.10.23.619408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
40 Hz sensory stimulation ("flicker") has emerged as a new technique to potentially mitigate pathology and improve cognition in mouse models of Alzheimer's disease (AD) pathology. However, it remains unknown how 40 Hz flicker affects neural codes essential for memory. Accordingly, we investigate the effects of 40 Hz flicker on neural representations of experience in the hippocampus of the 5XFAD mouse model of AD by recording 1000s of neurons during a goal-directed spatial navigation task. We find that an hour of daily exposure to 40 Hz audio-visual stimulation over 8 days leads to higher coordination between hippocampal subregions CA3 and CA1 during navigation. Consistent with CA3's role in generating sequential activity that represents future positions, 40 Hz flicker exposure increased prospective coding of future positions. In turn, prospective coding was more prominent during efficient navigation behavior. Our findings show how 40 Hz flicker enhances key hippocampal activity during behavior that is important for memory. Significance Statement Brain stimulation has emerged as a new potential therapeutic approach to potentially correct or improve altered neural activity in Alzheimer's disease. One such approach, 40 Hz sensory stimulation, or flicker, has been shown to improve cognition in disease models. However, it is not clear how 40 Hz flicker affects neural activity underlying memory processes. Here, we investigate how 40 Hz flicker exposure affects neural activity patterns that are crucial for memory. We find 40Hz flicker increases neural coordination in memory circuits, indicating better communication. Furthermore, 40Hz flicker increased neural representations of future positions, patterns theorized to support memory-based planning. These results indicate that 40 Hz flicker increases key neural activity that is important for memory.
Collapse
|
2
|
Li M, Jiang YQ, Lee DK, Wang H, Lu MC, Sun Q. Dorsoventral Heterogeneity of Synaptic Connectivity in Hippocampal CA3 Pyramidal Neurons. J Neurosci 2024; 44:e0370242024. [PMID: 39025678 PMCID: PMC11326861 DOI: 10.1523/jneurosci.0370-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/11/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024] Open
Abstract
The hippocampal CA3 region plays an important role in learning and memory. CA3 pyramidal neurons (PNs) receive two prominent excitatory inputs-mossy fibers (MFs) from dentate gyrus (DG) and recurrent collaterals (RCs) from CA3 PNs-that play opposing roles in pattern separation and pattern completion, respectively. Although the dorsoventral heterogeneity of the hippocampal anatomy, physiology, and behavior has been well established, nothing is known about the dorsoventral heterogeneity of synaptic connectivity in CA3 PNs. In this study, we performed Timm's sulfide silver staining, dendritic and spine morphological analyses, and ex vivo electrophysiology in mice of both sexes to investigate the heterogeneity of MF and RC pathways along the CA3 dorsoventral axis. Our morphological analyses demonstrate that ventral CA3 (vCA3) PNs possess greater dendritic lengths and more complex dendritic arborization, compared with dorsal CA3 (dCA3) PNs. Moreover, using ChannelRhodopsin2 (ChR2)-assisted patch-clamp recording, we found that the ratio of the RC-to-MF excitatory drive onto CA3 PNs increases substantially from dCA3 to vCA3, with vCA3 PNs receiving significantly weaker MFs, but stronger RCs, excitation than dCA3 PNs. Given the distinct roles of MF versus RC inputs in pattern separation versus completion, our findings of the significant dorsoventral variations of MF and RC excitation in CA3 PNs may have important functional implications for the contribution of CA3 circuit to the dorsoventral difference in hippocampal function.
Collapse
Affiliation(s)
- Minghua Li
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yu-Qiu Jiang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Daniel K Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Haoran Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Melissa C Lu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Qian Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
3
|
Miranda M, Silva A, Morici JF, Coletti MA, Belluscio M, Bekinschtein P. Retrieval of contextual memory can be predicted by CA3 remapping and is differentially influenced by NMDAR activity in rat hippocampus subregions. PLoS Biol 2024; 22:e3002706. [PMID: 38950066 PMCID: PMC11244845 DOI: 10.1371/journal.pbio.3002706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 07/12/2024] [Accepted: 06/12/2024] [Indexed: 07/03/2024] Open
Abstract
Episodic memory is essential to navigate in a changing environment by recalling past events, creating new memories, and updating stored information from experience. Although the mechanisms for acquisition and consolidation have been profoundly studied, much less is known about memory retrieval. Hippocampal spatial representations are key for retrieval of contextually guided episodic memories. Indeed, hippocampal place cells exhibit stable location-specific activity which is thought to support contextual memory, but can also undergo remapping in response to environmental changes. It is unclear if remapping is directly related to the expression of different episodic memories. Here, using an incidental memory recognition task in rats, we showed that retrieval of a contextually guided memory is reflected by the levels of CA3 remapping, demonstrating a clear link between external cues, hippocampal remapping, and episodic memory retrieval that guides behavior. Furthermore, we describe NMDARs as key players in regulating the balance between retrieval and memory differentiation processes by controlling the reactivation of specific memory traces. While an increase in CA3 NMDAR activity boosts memory retrieval, dentate gyrus NMDAR activity enhances memory differentiation. Our results contribute to understanding how the hippocampal circuit sustains a flexible balance between memory formation and retrieval depending on the environmental cues and the internal representations of the individual. They also provide new insights into the molecular mechanisms underlying the contributions of hippocampal subregions to generate this balance.
Collapse
Affiliation(s)
- Magdalena Miranda
- Laboratorio de Memoria y Cognición Molecular, Instituto de Neurociencia Cognitiva y Traslacional, CONICET-Fundación INECO-Universidad Favaloro, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Azul Silva
- Laboratorio Bases neuronales del comportamiento, Departamento de Ciencias Fisiológicas, Facultad de Ciencias Médicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- CONICET—Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Juan Facundo Morici
- Laboratorio de Memoria y Cognición Molecular, Instituto de Neurociencia Cognitiva y Traslacional, CONICET-Fundación INECO-Universidad Favaloro, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Marcos Antonio Coletti
- Laboratorio Bases neuronales del comportamiento, Departamento de Ciencias Fisiológicas, Facultad de Ciencias Médicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- CONICET—Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Mariano Belluscio
- Laboratorio Bases neuronales del comportamiento, Departamento de Ciencias Fisiológicas, Facultad de Ciencias Médicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
- CONICET—Universidad de Buenos Aires, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Pedro Bekinschtein
- Laboratorio de Memoria y Cognición Molecular, Instituto de Neurociencia Cognitiva y Traslacional, CONICET-Fundación INECO-Universidad Favaloro, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
4
|
Tamboli S, Singh S, Topolnik D, El Amine Barkat M, Radhakrishnan R, Guet-McCreight A, Topolnik L. Mouse hippocampal CA1 VIP interneurons detect novelty in the environment and support recognition memory. Cell Rep 2024; 43:114115. [PMID: 38607918 DOI: 10.1016/j.celrep.2024.114115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/17/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
In the CA1 hippocampus, vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) play a prominent role in disinhibitory circuit motifs. However, the specific behavioral conditions that lead to circuit disinhibition remain uncertain. To investigate the behavioral relevance of VIP-IN activity, we employed wireless technologies allowing us to monitor and manipulate their function in freely behaving mice. Our findings reveal that, during spatial exploration in new environments, VIP-INs in the CA1 hippocampal region become highly active, facilitating the rapid encoding of novel spatial information. Remarkably, both VIP-INs and pyramidal neurons (PNs) exhibit increased activity when encountering novel changes in the environment, including context- and object-related alterations. Concurrently, somatostatin- and parvalbumin-expressing inhibitory populations show an inverse relationship with VIP-IN and PN activity, revealing circuit disinhibition that occurs on a timescale of seconds. Thus, VIP-IN-mediated disinhibition may constitute a crucial element in the rapid encoding of novelty and the acquisition of recognition memory.
Collapse
Affiliation(s)
- Suhel Tamboli
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Sanjay Singh
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Dimitry Topolnik
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Mohamed El Amine Barkat
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Risna Radhakrishnan
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | | | - Lisa Topolnik
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada.
| |
Collapse
|
5
|
Barnstedt O, Mocellin P, Remy S. A hippocampus-accumbens code guides goal-directed appetitive behavior. Nat Commun 2024; 15:3196. [PMID: 38609363 PMCID: PMC11015045 DOI: 10.1038/s41467-024-47361-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
The dorsal hippocampus (dHPC) is a key brain region for the expression of spatial memories, such as navigating towards a learned reward location. The nucleus accumbens (NAc) is a prominent projection target of dHPC and implicated in value-based action selection. Yet, the contents of the dHPC→NAc information stream and their acute role in behavior remain largely unknown. Here, we found that optogenetic stimulation of the dHPC→NAc pathway while mice navigated towards a learned reward location was both necessary and sufficient for spatial memory-related appetitive behaviors. To understand the task-relevant coding properties of individual NAc-projecting hippocampal neurons (dHPC→NAc), we used in vivo dual-color two-photon imaging. In contrast to other dHPC neurons, the dHPC→NAc subpopulation contained more place cells, with enriched spatial tuning properties. This subpopulation also showed enhanced coding of non-spatial task-relevant behaviors such as deceleration and appetitive licking. A generalized linear model revealed enhanced conjunctive coding in dHPC→NAc neurons which improved the identification of the reward zone. We propose that dHPC routes specific reward-related spatial and behavioral state information to guide NAc action selection.
Collapse
Affiliation(s)
- Oliver Barnstedt
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany.
- Institute for Biology, Otto-von-Guericke University, 39120, Magdeburg, Germany.
| | - Petra Mocellin
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
- International Max Planck Research, School for Brain & Behavior (IMPRS), 53175, Bonn, Germany
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720-3370, USA
| | - Stefan Remy
- Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), 39106, Magdeburg, Germany.
- German Center for Mental Health (DZGP), partner site Halle-Jena-Magdeburg, 39118, Magdeburg, Germany.
| |
Collapse
|
6
|
Chettih SN, Mackevicius EL, Hale S, Aronov D. Barcoding of episodic memories in the hippocampus of a food-caching bird. Cell 2024; 187:1922-1935.e20. [PMID: 38554707 PMCID: PMC11015962 DOI: 10.1016/j.cell.2024.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/28/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024]
Abstract
The hippocampus is critical for episodic memory. Although hippocampal activity represents place and other behaviorally relevant variables, it is unclear how it encodes numerous memories of specific events in life. To study episodic coding, we leveraged the specialized behavior of chickadees-food-caching birds that form memories at well-defined moments in time whenever they cache food for subsequent retrieval. Our recordings during caching revealed very sparse, transient barcode-like patterns of firing across hippocampal neurons. Each "barcode" uniquely represented a caching event and transiently reactivated during the retrieval of that specific cache. Barcodes co-occurred with the conventional activity of place cells but were uncorrelated even for nearby cache locations that had similar place codes. We propose that animals recall episodic memories by reactivating hippocampal barcodes. Similarly to computer hash codes, these patterns assign unique identifiers to different events and could be a mechanism for rapid formation and storage of many non-interfering memories.
Collapse
Affiliation(s)
- Selmaan N Chettih
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Emily L Mackevicius
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Basis Research Institute, New York, NY 10027, USA
| | - Stephanie Hale
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Dmitriy Aronov
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
7
|
Hainmueller T, Cazala A, Huang LW, Bartos M. Subfield-specific interneuron circuits govern the hippocampal response to novelty in male mice. Nat Commun 2024; 15:714. [PMID: 38267409 PMCID: PMC10808551 DOI: 10.1038/s41467-024-44882-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/04/2024] [Indexed: 01/26/2024] Open
Abstract
The hippocampus is the brain's center for episodic memories. Its subregions, the dentate gyrus and CA1-3, are differentially involved in memory encoding and recall. Hippocampal principal cells represent episodic features like movement, space, and context, but less is known about GABAergic interneurons. Here, we performed two-photon calcium imaging of parvalbumin- and somatostatin-expressing interneurons in the dentate gyrus and CA1-3 of male mice exploring virtual environments. Parvalbumin-interneurons increased activity with running-speed and reduced it in novel environments. Somatostatin-interneurons in CA1-3 behaved similar to parvalbumin-expressing cells, but their dentate gyrus counterparts increased activity during rest and in novel environments. Congruently, chemogenetic silencing of dentate parvalbumin-interneurons had prominent effects in familiar contexts, while silencing somatostatin-expressing cells increased similarity of granule cell representations between novel and familiar environments. Our data indicate unique roles for parvalbumin- and somatostatin-positive interneurons in the dentate gyrus that are distinct from those in CA1-3 and may support routing of novel information.
Collapse
Affiliation(s)
- Thomas Hainmueller
- Institute for Physiology I, University of Freiburg, Medical Faculty, 79104, Freiburg, Germany.
- NYU Neuroscience Institute, 435 East 30th Street, New York, NY, 10016, USA.
- Department of Psychiatry, New York University Langone Medical Center, New York, NY, 10016, USA.
| | - Aurore Cazala
- Institute for Physiology I, University of Freiburg, Medical Faculty, 79104, Freiburg, Germany
| | - Li-Wen Huang
- Institute for Physiology I, University of Freiburg, Medical Faculty, 79104, Freiburg, Germany
| | - Marlene Bartos
- Institute for Physiology I, University of Freiburg, Medical Faculty, 79104, Freiburg, Germany.
| |
Collapse
|
8
|
Bannerman DM, Barkus C, Eltokhi A. Behavioral Analysis of NMDAR Function in Rodents: Tests of Long-Term Spatial Memory. Methods Mol Biol 2024; 2799:107-138. [PMID: 38727905 DOI: 10.1007/978-1-0716-3830-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
NMDAR-dependent forms of synaptic plasticity in brain regions like the hippocampus are widely believed to provide the neural substrate for long-term associative memory formation. However, the experimental data are equivocal at best and may suggest a more nuanced role for NMDARs and synaptic plasticity in memory. Much of the experimental data available comes from studies in genetically modified mice in which NMDAR subunits have been deleted or mutated in order to disrupt NMDAR function. Behavioral assessment of long-term memory in these mice has involved tests like the Morris watermaze and the radial arm maze. Here we describe these behavioral tests and some of the different testing protocols that can be used to assess memory performance. We discuss the importance of distinguishing selective effects on learning and memory processes from nonspecific effects on sensorimotor or motivational aspects of performance.
Collapse
Affiliation(s)
- David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK.
| | - Chris Barkus
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Ahmed Eltokhi
- Department of Biomedical Sciences, School of Medicine, Mercer University, Columbus, GA, USA
| |
Collapse
|
9
|
Dragoi G. The generative grammar of the brain: a critique of internally generated representations. Nat Rev Neurosci 2024; 25:60-75. [PMID: 38036709 DOI: 10.1038/s41583-023-00763-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 12/02/2023]
Abstract
The past decade of progress in neurobiology has uncovered important organizational principles for network preconfiguration and neuronal selection that suggest a generative grammar exists in the brain. In this Perspective, I discuss the competence of the hippocampal neural network to generically express temporally compressed sequences of neuronal firing that represent novel experiences, which is envisioned as a form of generative neural syntax supporting a neurobiological perspective on brain function. I compare this neural competence with the hippocampal network performance that represents specific experiences with higher fidelity after new learning during replay, which is envisioned as a form of neural semantic that supports a complementary neuropsychological perspective. I also demonstrate how the syntax of network competence emerges a priori during early postnatal life and is followed by the later development of network performance that enables rapid encoding and memory consolidation. Thus, I propose that this generative grammar of the brain is essential for internally generated representations, which are crucial for the cognitive processes underlying learning and memory, prospection, and inference, which ultimately underlie our reason and representation of the world.
Collapse
Affiliation(s)
- George Dragoi
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
10
|
Pinke D, Issa JB, Dara GA, Dobos G, Dombeck DA. Full field-of-view virtual reality goggles for mice. Neuron 2023; 111:3941-3952.e6. [PMID: 38070501 PMCID: PMC10841834 DOI: 10.1016/j.neuron.2023.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/03/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023]
Abstract
Visual virtual reality (VR) systems for head-fixed mice offer advantages over real-world studies for investigating the neural circuitry underlying behavior. However, current VR approaches do not fully cover the visual field of view of mice, do not stereoscopically illuminate the binocular zone, and leave the lab frame visible. To overcome these limitations, we developed iMRSIV (Miniature Rodent Stereo Illumination VR)-VR goggles for mice. Our system is compact, separately illuminates each eye for stereo vision, and provides each eye with an ∼180° field of view, thus excluding the lab frame while accommodating saccades. Mice using iMRSIV while navigating engaged in virtual behaviors more quickly than in a current monitor-based system and displayed freezing and fleeing reactions to overhead looming stimulation. Using iMRSIV with two-photon functional imaging, we found large populations of hippocampal place cells during virtual navigation, global remapping during environment changes, and unique responses of place cell ensembles to overhead looming stimulation.
Collapse
Affiliation(s)
- Domonkos Pinke
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - John B Issa
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Gabriel A Dara
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Gergely Dobos
- 360world Ltd, Sümegvár köz 9, 1118 Budapest, Hungary
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
11
|
Yu Y, Wu K, Yang X, Long J, Chang C. Terahertz Photons Improve Cognitive Functions in Posttraumatic Stress Disorder. RESEARCH (WASHINGTON, D.C.) 2023; 6:0278. [PMID: 38111677 PMCID: PMC10726292 DOI: 10.34133/research.0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/12/2023] [Indexed: 12/20/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a serious psychosis leading to cognitive impairment. To restore cognitive functions for patients, the main treatments are based on medication or rehabilitation training but with limited effectiveness and strong side effects. Here, we demonstrate a new treatment approach for PTSD by using terahertz (THz) photons stimulating the hippocampal CA3 subregion. We verified that this method can nonthermally restore cognitive function in PTSD rats in vivo. After THz photon irradiation, the PTSD rats' recognitive index improved by about 10% in a novel object recognition test, the PTSD rats' accuracy improved by about 100% in a shuttler box test, the PTSD rats' numbers to identify target box was about 5 times lower in a Barnes maze test, and the rate of staying in new arm increased by approximately 40% in a Y-maze test. Further experimental studies found that THz photon (34.5 THz) irradiation could improve the expression of NR2B (increased by nearly 40%) and phosphorylated NR2B (increased by about 50%). In addition, molecular dynamics simulations showed that THz photons at a frequency of 34.5 THz are mainly absorbed by the pocket of glutamate receptors rather than by glutamate molecules. Moreover, the binding between glutamate receptors and glutamate molecules was increased by THz photons. This study offers a nondrug, nonthermal approach to regulate the binding between the excitatory neurotransmitter (glutamate) and NR2B. By increasing synaptic plasticity, it effectively improves the cognitive function of animals with PTSD, providing a promising treatment strategy for NR2B-related cognitive disorders.
Collapse
Affiliation(s)
- Yun Yu
- School of Life Science and Technology,
Xi’an Jiaotong University, Xi’an 710049, China
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Kaijie Wu
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Xiao Yang
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Jiangang Long
- School of Life Science and Technology,
Xi’an Jiaotong University, Xi’an 710049, China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
- School of Physics,
Peking University, Beijing 100871, China
| |
Collapse
|
12
|
Atucha E, Ku SP, Lippert MT, Sauvage MM. Recalling gist memory depends on CA1 hippocampal neurons for lifetime retention and CA3 neurons for memory precision. Cell Rep 2023; 42:113317. [PMID: 37897725 DOI: 10.1016/j.celrep.2023.113317] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/05/2023] [Accepted: 10/05/2023] [Indexed: 10/30/2023] Open
Abstract
Why some of us remember events more clearly than others and why memory loses precision over time is a major focus in memory research. Here, we show that the recruitment of specific neuroanatomical pathways within the medial temporal lobe (MTL) of the brain defines the precision of the memory recalled over the lifespan. Using optogenetics, neuronal activity mapping, and studying recent to very remote memories, we report that the hippocampal subfield CA1 is necessary for retrieving the gist of events and receives maximal support from MTL cortical areas (MEC, LEC, PER, and POR) for recalling the most remote memories. In contrast, reduction of CA3's activity alone coincides with the loss of memory precision over time. We propose that a shift between specific MTL subnetworks over time might be a fundamental mechanism of memory consolidation.
Collapse
Affiliation(s)
- Erika Atucha
- Functional Architecture of Memory Department, Leibniz Institute for Neurobiology, Magdeburg, Germany.
| | - Shih-Pi Ku
- Functional Architecture of Memory Department, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Michael T Lippert
- Systems Physiology of Learning Department, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Magdalena M Sauvage
- Functional Architecture of Memory Department, Leibniz Institute for Neurobiology, Magdeburg, Germany; Otto von Guericke University, Medical Faculty, Functional Neuroplasticity Department, Magdeburg, Germany; Otto von Guericke University, Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
13
|
Chettih SN, Mackevicius EL, Hale S, Aronov D. Barcoding of episodic memories in the hippocampus of a food-caching bird. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.27.542597. [PMID: 37461442 PMCID: PMC10349996 DOI: 10.1101/2023.05.27.542597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Episodic memory, or memory of experienced events, is a critical function of the hippocampus1-3. It is therefore important to understand how hippocampal activity represents specific events in an animal's life. We addressed this question in chickadees - specialist food-caching birds that hide food at scattered locations and use memory to find their caches later in time4,5. We performed high-density neural recordings in the hippocampus of chickadees as they cached and retrieved seeds in a laboratory arena. We found that each caching event was represented by a burst of firing in a unique set of hippocampal neurons. These 'barcode-like' patterns of activity were sparse (<10% of neurons active), uncorrelated even for immediately adjacent caches, and different even for separate caches at the same location. The barcode representing a specific caching event was transiently reactivated whenever a bird later interacted with the same cache - for example, to retrieve food. Barcodes co-occurred with conventional place cell activity6,7, as well as location-independent responses to cached seeds. We propose that barcodes are signatures of episodic memories evoked during memory recall. These patterns assign a unique identifier to each event and may be a mechanism for rapid formation and storage of many non-interfering memories.
Collapse
Affiliation(s)
| | | | - Stephanie Hale
- Zuckerman Mind Brain Behavior Institute, Columbia University
| | - Dmitriy Aronov
- Zuckerman Mind Brain Behavior Institute, Columbia University
| |
Collapse
|
14
|
Li M, Kinney JL, Jiang YQ, Lee DK, Wu Q, Lee D, Xiong WC, Sun Q. Hypothalamic Supramammillary Nucleus Selectively Excites Hippocampal CA3 Interneurons to Suppress CA3 Pyramidal Neuron Activity. J Neurosci 2023; 43:4612-4624. [PMID: 37117012 PMCID: PMC10286942 DOI: 10.1523/jneurosci.1910-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
A key mode of neuronal communication between distant brain regions is through excitatory synaptic transmission mediated by long-range glutamatergic projections emitted from principal neurons. The long-range glutamatergic projection normally forms numerous en passant excitatory synapses onto both principal neurons and interneurons along its path. Under physiological conditions, the monosynaptic excitatory drive onto postsynaptic principal neurons outweighs disynaptic feedforward inhibition, with the net effect of depolarizing principal neurons. In contrast with this conventional doctrine, here we report that a glutamatergic projection from the hypothalamic supramammillary nucleus (SuM) largely evades postsynaptic pyramidal neurons (PNs), but preferentially target interneurons in the hippocampal CA3 region to predominantly provide feedforward inhibition. Using viral-based retrograde and anterograde tracing and ChannelRhodopsin2 (ChR2)-assisted patch-clamp recording in mice of either sex, we show that SuM projects sparsely to CA3 and provides minimal excitation onto CA3 PNs. Surprisingly, despite its sparse innervation, the SuM input inhibits all CA3 PNs along the transverse axis. Further, we find that SuM provides strong monosynaptic excitation onto CA3 parvalbumin-expressing interneurons evenly along the transverse axis, which likely mediates the SuM-driven feedforward inhibition. Together, our results demonstrate that a novel long-range glutamatergic pathway largely evades principal neurons, but rather preferentially innervates interneurons in a distant brain region to suppress principal neuron activity. Moreover, our findings reveal a new means by which SuM regulates hippocampal activity through SuM-to-CA3 circuit, independent of the previously focused projections from SuM to CA2 or dentate gyrus.SIGNIFICANCE STATEMENT The dominant mode of neuronal communication between brain regions is the excitatory synaptic transmission mediated by long-range glutamatergic projections, which form en passant excitatory synapses onto both pyramidal neurons and interneurons along its path. Under normal conditions, the excitation onto postsynaptic neurons outweighs feedforward inhibition, with the net effect of depolarization. In contrast with this conventional doctrine, here we report that a glutamatergic input from hypothalamic supramammillary nucleus (SuM) largely evades PNs but selectively targets interneurons to almost exclusively provide disynaptic feedforward inhibition onto hippocampal CA3 PNs. Thus, our findings reveal a novel subcortical-hippocampal circuit that enables SuM to regulate hippocampal activity via SuM-CA3 circuit, independent of its projections to CA2 or dentate gyrus.
Collapse
Affiliation(s)
- Minghua Li
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jessica L Kinney
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yu-Qiu Jiang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Daniel K Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Qiwen Wu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Qian Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| |
Collapse
|
15
|
Lutzu S, Alviña K, Puente N, Grandes P, Castillo PE. Target cell-specific plasticity rules of NMDA receptor-mediated synaptic transmission in the hippocampus. Front Cell Neurosci 2023; 17:1068472. [PMID: 37091922 PMCID: PMC10113460 DOI: 10.3389/fncel.2023.1068472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Long-term potentiation and depression of NMDA receptor-mediated synaptic transmission (NMDAR LTP/LTD) can significantly impact synapse function and information transfer in several brain areas. However, the mechanisms that determine the direction of NMDAR plasticity are poorly understood. Here, using physiologically relevant patterns of presynaptic and postsynaptic burst activities, whole-cell patch clamp recordings, 2-photon laser calcium imaging in acute rat hippocampal slices and immunoelectron microscopy, we tested whether distinct calcium dynamics and group I metabotropic glutamate receptor (I-mGluR) subtypes control the sign of NMDAR plasticity. We found that postsynaptic calcium transients (CaTs) in response to hippocampal MF stimulation were significantly larger during the induction of NMDAR-LTP compared to NMDAR-LTD at the MF-to-CA3 pyramidal cell (MF-CA3) synapse. This difference was abolished by pharmacological blockade of mGluR5 and was significantly reduced by depletion of intracellular calcium stores, whereas blocking mGluR1 had no effect on these CaTs. In addition, we discovered that MF to hilar mossy cell (MF-MC) synapses, which share several structural and functional commonalities with MF-CA3 synapses, also undergoes NMDAR plasticity. To our surprise, however, we found that the postsynaptic distribution of I-mGluR subtypes at these two synapses differ, and the same induction protocol that induces NMDAR-LTD at MF-CA3 synapses, only triggered NMDAR-LTP at MF-MC synapses, despite a comparable calcium dynamics. Thus, postsynaptic calcium dynamics alone cannot predict the sign of NMDAR plasticity, indicating that both postsynaptic calcium rise and the relative contribution of I-mGluR subtypes likely determine the learning rules of NMDAR plasticity.
Collapse
Affiliation(s)
- Stefano Lutzu
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Karina Alviña
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Nagore Puente
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, Leioa, Spain
| | - Pedro Grandes
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Science Park of the University of the Basque Country UPV/EHU, Leioa, Spain
| | - Pablo E. Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
- *Correspondence: Pablo E. Castillo,
| |
Collapse
|
16
|
Traunmüller L, Schulz J, Ortiz R, Feng H, Furlanis E, Gomez AM, Schreiner D, Bischofberger J, Zhang C, Scheiffele P. A cell-type-specific alternative splicing regulator shapes synapse properties in a trans-synaptic manner. Cell Rep 2023; 42:112173. [PMID: 36862556 PMCID: PMC10066595 DOI: 10.1016/j.celrep.2023.112173] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/07/2022] [Accepted: 02/12/2023] [Indexed: 03/03/2023] Open
Abstract
The specification of synaptic properties is fundamental for the function of neuronal circuits. "Terminal selector" transcription factors coordinate terminal gene batteries that specify cell-type-specific properties. Moreover, pan-neuronal splicing regulators have been implicated in directing neuronal differentiation. However, the cellular logic of how splicing regulators instruct specific synaptic properties remains poorly understood. Here, we combine genome-wide mapping of mRNA targets and cell-type-specific loss-of-function studies to uncover the contribution of the RNA-binding protein SLM2 to hippocampal synapse specification. Focusing on pyramidal cells and somatostatin (SST)-positive GABAergic interneurons, we find that SLM2 preferentially binds and regulates alternative splicing of transcripts encoding synaptic proteins. In the absence of SLM2, neuronal populations exhibit normal intrinsic properties, but there are non-cell-autonomous synaptic phenotypes and associated defects in a hippocampus-dependent memory task. Thus, alternative splicing provides a critical layer of gene regulation that instructs specification of neuronal connectivity in a trans-synaptic manner.
Collapse
Affiliation(s)
| | - Jan Schulz
- Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | - Raul Ortiz
- Biozentrum of the University of Basel, 4056 Basel, Switzerland
| | - Huijuan Feng
- Department of Systems Biology, Department of Biochemistry and Molecular Biophysics, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
| | | | - Andrea M Gomez
- Biozentrum of the University of Basel, 4056 Basel, Switzerland
| | | | | | - Chaolin Zhang
- Department of Systems Biology, Department of Biochemistry and Molecular Biophysics, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
17
|
Lehr AB, Hitti FL, Deibel SH, Stöber TM. Silencing hippocampal CA2 reduces behavioral flexibility in spatial learning. Hippocampus 2023; 33:759-768. [PMID: 36938702 DOI: 10.1002/hipo.23521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 03/21/2023]
Abstract
The hippocampus is a key structure involved in learning and remembering spatial information. However, the extent to which hippocampal region CA2 is involved in these processes remains unclear. Here, we show that chronically silencing dorsal CA2 impairs reversal learning in the Morris water maze. After platform relocation, CA2-silenced mice spent more time in the vicinity of the old platform location and less time in the new target quadrant. Accordingly, behavioral strategy analysis revealed increased perseverance in navigating to the old location during the first day and an increased use of non-spatial strategies during the second day of reversal learning. Confirming previous indirect indications, these results demonstrate that CA2 is recruited when mice must flexibly adapt their behavior as task contingencies change. We discuss how these findings can be explained by recent theories of CA2 function and outline testable predictions to understand the underlying neural mechanisms. Demonstrating a direct involvement of CA2 in spatial learning, this work lends further support to the notion that CA2 plays a fundamental role in hippocampal information processing.
Collapse
Affiliation(s)
- Andrew B Lehr
- Department of Computational Synaptic Physiology, University of Göttingen, Göttingen, Germany
| | - Frederick L Hitti
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Scott H Deibel
- Department of Psychology, University of New Brunswick, Fredericton, New Brunswick, Canada
| | - Tristan M Stöber
- Institute for Neuroinformatics, Ruhr University Bochum, Bochum, Germany.,Department of Neurology, University Hospital Frankfurt, Frankfurt, Germany.,Frankfurt Institute for Advanced Studies, Frankfurt, Germany
| |
Collapse
|
18
|
Vancura B, Geiller T, Losonczy A. Organization and Plasticity of Inhibition in Hippocampal Recurrent Circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532296. [PMID: 36993553 PMCID: PMC10054977 DOI: 10.1101/2023.03.13.532296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Excitatory-inhibitory interactions structure recurrent network dynamics for efficient cortical computations. In the CA3 area of the hippocampus, recurrent circuit dynamics, including experience-induced plasticity at excitatory synapses, are thought to play a key role in episodic memory encoding and consolidation via rapid generation and flexible selection of neural ensembles. However, in vivo activity of identified inhibitory motifs supporting this recurrent circuitry has remained largely inaccessible, and it is unknown whether CA3 inhibition is also modifiable upon experience. Here we use large-scale, 3-dimensional calcium imaging and retrospective molecular identification in the mouse hippocampus to obtain the first comprehensive description of molecularly-identified CA3 interneuron dynamics during both spatial navigation and sharp-wave ripple (SWR)-associated memory consolidation. Our results uncover subtype-specific dynamics during behaviorally distinct brain-states. Our data also demonstrate predictive, reflective, and experience-driven plastic recruitment of specific inhibitory motifs during SWR-related memory reactivation. Together these results assign active roles for inhibitory circuits in coordinating operations and plasticity in hippocampal recurrent circuits.
Collapse
|
19
|
Sheintuch L, Geva N, Deitch D, Rubin A, Ziv Y. Organization of hippocampal CA3 into correlated cell assemblies supports a stable spatial code. Cell Rep 2023; 42:112119. [PMID: 36807137 PMCID: PMC9989830 DOI: 10.1016/j.celrep.2023.112119] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/30/2022] [Accepted: 01/30/2023] [Indexed: 02/19/2023] Open
Abstract
Hippocampal subfield CA3 is thought to stably store memories in assemblies of recurrently connected cells functioning as a collective. However, the collective hippocampal coding properties that are unique to CA3 and how such properties facilitate the stability or precision of the neural code remain unclear. Here, we performed large-scale Ca2+ imaging in hippocampal CA1 and CA3 of freely behaving mice that repeatedly explored the same, initially novel environments over weeks. CA3 place cells have more precise and more stable tuning and show a higher statistical dependence with their peers compared with CA1 place cells, uncovering a cell assembly organization in CA3. Surprisingly, although tuning precision and long-term stability are correlated, cells with stronger peer dependence exhibit higher stability but not higher precision. Overall, our results expose the three-way relationship between tuning precision, long-term stability, and peer dependence, suggesting that a cell assembly organization underlies long-term storage of information in the hippocampus.
Collapse
Affiliation(s)
- Liron Sheintuch
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Nitzan Geva
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Deitch
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Rubin
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| | - Yaniv Ziv
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
20
|
Shinohara Y, Kohara K. Projections of hippocampal CA2 pyramidal neurons: Distinct innervation patterns of CA2 compared to CA3 in rodents. Hippocampus 2023; 33:691-699. [PMID: 36855258 DOI: 10.1002/hipo.23519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023]
Abstract
The hippocampus is a center for spatial and episodic memory formation in rodents. Understanding the composition of subregions and circuitry maps of the hippocampus is essential for elucidating the mechanism of memory formation and recall. For decades, the trisynaptic circuit (entorhinal cortex layer II-dentate gyrus - CA3-CA1) has been considered the neural network substrate responsible for learning and memory. Recently, CA2 has emerged as an important area in the hippocampal circuitry, with distinct functions from those of CA3. In this article, we review the historical definition of the hippocampal area CA2 and the differential projection patterns between CA2 and CA3 pyramidal neurons. We provide a concise and comprehensive map of CA2 outputs by comparing (1) ipsi versus contra projections, (2) septal versus temporal projections, and (3) lamellar structures of CA2 and CA3 pyramidal neurons.
Collapse
Affiliation(s)
- Yoshiaki Shinohara
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Keigo Kohara
- KMU Biobank Center, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
21
|
Nomoto M, Murayama E, Ohno S, Okubo-Suzuki R, Muramatsu SI, Inokuchi K. Hippocampus as a sorter and reverberatory integrator of sensory inputs. Nat Commun 2022; 13:7413. [PMID: 36539403 PMCID: PMC9768143 DOI: 10.1038/s41467-022-35119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
The hippocampus must be capable of sorting and integrating multiple sensory inputs separately but simultaneously. However, it remains to be elucidated how the hippocampus executes these processes simultaneously during learning. Here we found that synchrony between conditioned stimulus (CS)-, unconditioned stimulus (US)- and future retrieval-responsible cells occurs in the CA1 during the reverberatory phase that emerges after sensory inputs have ceased, but not during CS and US inputs. Mutant mice lacking N-methyl-D-aspartate receptors (NRs) in CA3 showed a cued-fear memory impairment and a decrease in synchronized reverberatory activities between CS- and US-responsive CA1 cells. Optogenetic CA3 silencing at the reverberatory phase during learning impaired cued-fear memory. Thus, the hippocampus uses reverberatory activity to link CS and US inputs, and avoid crosstalk during sensory inputs.
Collapse
Affiliation(s)
- Masanori Nomoto
- grid.267346.20000 0001 2171 836XResearch Centre for Idling Brain Science, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XDepartment of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XCREST, JST, University of Toyama, Toyama, 930−0194 Japan
| | - Emi Murayama
- grid.267346.20000 0001 2171 836XResearch Centre for Idling Brain Science, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XDepartment of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XCREST, JST, University of Toyama, Toyama, 930−0194 Japan
| | - Shuntaro Ohno
- grid.267346.20000 0001 2171 836XResearch Centre for Idling Brain Science, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XDepartment of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XCREST, JST, University of Toyama, Toyama, 930−0194 Japan
| | - Reiko Okubo-Suzuki
- grid.267346.20000 0001 2171 836XResearch Centre for Idling Brain Science, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XDepartment of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XCREST, JST, University of Toyama, Toyama, 930−0194 Japan
| | - Shin-ichi Muramatsu
- grid.410804.90000000123090000Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, 329−0498 Japan ,grid.26999.3d0000 0001 2151 536XCenter for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, 108−8639 Japan
| | - Kaoru Inokuchi
- grid.267346.20000 0001 2171 836XResearch Centre for Idling Brain Science, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XDepartment of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930−0194 Japan ,grid.267346.20000 0001 2171 836XCREST, JST, University of Toyama, Toyama, 930−0194 Japan
| |
Collapse
|
22
|
Jeong N, Singer AC. Learning from inhibition: Functional roles of hippocampal CA1 inhibition in spatial learning and memory. Curr Opin Neurobiol 2022; 76:102604. [PMID: 35810533 PMCID: PMC11414469 DOI: 10.1016/j.conb.2022.102604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 11/19/2022]
Abstract
Hippocampal inhibitory interneurons exert a powerful influence on learning and memory. Inhibitory interneurons are known to play a major role in many diseases that affect memory, and to strongly influence brain functions required for memory-related tasks. While previous studies involving genetic, optogenetic, and pharmacological manipulations have shown that hippocampal interneurons play essential roles in spatial and episodic learning and memory, exactly how interneurons affect local circuit computations during spatial navigation is not well understood. Given the significant anatomical, morphological, and functional heterogeneity in hippocampal interneurons, one may suspect cell-type specific roles in circuit computations. Here, we review emerging evidence of CA1 hippocampal interneurons' role in local circuit computations that support spatial learning and memory and discuss open questions about CA1 interneurons in spatial learning.
Collapse
Affiliation(s)
- Nuri Jeong
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA; Neuroscience Graduate Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, 30322, USA. https://twitter.com/nuriscientist
| | - Annabelle C Singer
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
| |
Collapse
|
23
|
Jin SX, Liu L, Li S, Meunier AL, Selkoe DJ. Aβ oligomers from human brain impair mossy fiber LTP in CA3 of hippocampus, but activating cAMP-PKA and cGMP-PKG prevents this. Neurobiol Dis 2022; 172:105816. [PMID: 35820646 PMCID: PMC9809147 DOI: 10.1016/j.nbd.2022.105816] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 01/05/2023] Open
Abstract
Early cognitive impairment in Alzheimer's disease may result in part from synaptic dysfunction caused by the accumulation oligomeric assemblies of amyloid β-protein (Aβ). Changes in hippocampal function seem critical for cognitive impairment in early Alzheimer's disease (AD). Diffusible oligomers of Aβ (oAβ) have been shown to block canonical long-term potentiation (LTP) in the CA1 area of hippocampus, but whether there is also a direct effect of oAβ on synaptic transmission and plasticity at synapses between mossy fibers (axons) from the dentate gyrus granule cells and CA3 pyramidal neurons (mf-CA3 synapses) is unknown. Studies in APP transgenic mice have suggested an age-dependent impairment of mossy fiber LTP. Here we report that although endogenous AD brain-derived soluble oAβ had no effect on mossy-fiber basal transmission, it strongly impaired paired-pulse facilitation in the mossy fiber pathway and presynaptic mossy fiber LTP (mf-LTP). Selective activation of both β1 and β2 adrenergic receptors and their downstream cAMP/PKA signaling pathway prevented oAβ-mediated inhibition of mf-LTP. Unexpectedly, activation of the cGMP/PKG signaling pathway also prevented oAβ-impaired mf-LTP. Our results reveal certain specific pharmacological targets to ameliorate human oAβ-mediated impairment at the mf-CA3 synapse.
Collapse
Affiliation(s)
| | | | | | | | - Dennis J. Selkoe
- Corresponding author at: Hale Building for Transformative Medicine, Rm 10002Q, 60 Fenwood Road, Boston, MA 02115, United States of America. (D.J. Selkoe)
| |
Collapse
|
24
|
Abstract
When navigating through space, we must maintain a representation of our position in real time; when recalling a past episode, a memory can come back in a flash. Interestingly, the brain's spatial representation system, including the hippocampus, supports these two distinct timescale functions. How are neural representations of space used in the service of both real-world navigation and internal mnemonic processes? Recent progress has identified sequences of hippocampal place cells, evolving at multiple timescales in accordance with either navigational behaviors or internal oscillations, that underlie these functions. We review experimental findings on experience-dependent modulation of these sequential representations and consider how they link real-world navigation to time-compressed memories. We further discuss recent work suggesting the prevalence of these sequences beyond hippocampus and propose that these multiple-timescale mechanisms may represent a general algorithm for organizing cell assemblies, potentially unifying the dual roles of the spatial representation system in memory and navigation.
Collapse
Affiliation(s)
- Wenbo Tang
- Graduate Program in Neuroscience, Brandeis University, Waltham, Massachusetts, USA;
| | - Shantanu P Jadhav
- Neuroscience Program, Department of Psychology, and Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts, USA;
| |
Collapse
|
25
|
Ma L, Patel M. Mechanism of carbachol-induced 40 Hz gamma oscillations and the effects of NMDA activation on oscillatory dynamics in a model of the CA3 subfield of the hippocampus. J Theor Biol 2022; 548:111200. [PMID: 35716721 DOI: 10.1016/j.jtbi.2022.111200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/26/2022] [Accepted: 06/08/2022] [Indexed: 11/26/2022]
Abstract
Gamma oscillations are a prominent feature of various neural systems, including the CA3 subfield of the hippocampus. In CA3, in vitro carbachol application induces ∼40 Hz gamma oscillations in the network of glutamatergic excitatory pyramidal neurons (PNs) and local GABAergic inhibitory neurons (INs). Activation of NMDA receptors within CA3 leads to an increase in the frequency of carbachol-induced oscillations to ∼60 Hz, a broadening of the distribution of individual oscillation cycle frequencies, and a decrease in the time lag between PN and IN spike bursts. In this work, we develop a biophysical integrate-and-fire model of the CA3 subfield, we show that the dynamics of our model are in concordance with physiological observations, and we provide computational support for the hypothesis that the 'E-I' mechanism is responsible for the emergence of ∼40 Hz gamma oscillations in the absence of NMDA activation. We then incorporate NMDA receptors into our CA3 model, and we show that our model exhibits the increase in gamma oscillation frequency, broadening of the cycle frequency distribution, and decrease in the time lag between PN and IN spike bursts observed experimentally. Remarkably, we find an inverse relationship in our model between the net NMDA current delivered to PNs and INs in an oscillation cycle and cycle frequency. Furthermore, we find a disparate effect of NMDA receptors on PNs versus INs - we show that NMDA receptors on INs tend to increase oscillation frequency, while NMDA receptors on PNs tend to slightly decrease or not affect oscillation frequency. We find that these observations can be explained if NMDA activity above a threshold level causes a shift in the mechanism underlying gamma oscillations; in the absence of NMDA receptors, the 'E-I' mechanism is primarily responsible for the generation of gamma oscillations (at 40 Hz), while when NMDA receptors are active, the mechanism of gamma oscillations shifts to the 'I-I' mechanism, and we argue that within the 'I-I' regime (which displays a higher baseline oscillation frequency of ∼60 Hz), slight changes in the level of NMDA activity are inversely related to cycle frequency.
Collapse
Affiliation(s)
- Linda Ma
- Department of Mathematics, William & Mary, United States.
| | - Mainak Patel
- Department of Mathematics, William & Mary, United States.
| |
Collapse
|
26
|
Yang S, Gao T, Wang J, Deng B, Azghadi MR, Lei T, Linares-Barranco B. SAM: A Unified Self-Adaptive Multicompartmental Spiking Neuron Model for Learning With Working Memory. Front Neurosci 2022; 16:850945. [PMID: 35527819 PMCID: PMC9074872 DOI: 10.3389/fnins.2022.850945] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Working memory is a fundamental feature of biological brains for perception, cognition, and learning. In addition, learning with working memory, which has been show in conventional artificial intelligence systems through recurrent neural networks, is instrumental to advanced cognitive intelligence. However, it is hard to endow a simple neuron model with working memory, and to understand the biological mechanisms that have resulted in such a powerful ability at the neuronal level. This article presents a novel self-adaptive multicompartment spiking neuron model, referred to as SAM, for spike-based learning with working memory. SAM integrates four major biological principles including sparse coding, dendritic non-linearity, intrinsic self-adaptive dynamics, and spike-driven learning. We first describe SAM's design and explore the impacts of critical parameters on its biological dynamics. We then use SAM to build spiking networks to accomplish several different tasks including supervised learning of the MNIST dataset using sequential spatiotemporal encoding, noisy spike pattern classification, sparse coding during pattern classification, spatiotemporal feature detection, meta-learning with working memory applied to a navigation task and the MNIST classification task, and working memory for spatiotemporal learning. Our experimental results highlight the energy efficiency and robustness of SAM in these wide range of challenging tasks. The effects of SAM model variations on its working memory are also explored, hoping to offer insight into the biological mechanisms underlying working memory in the brain. The SAM model is the first attempt to integrate the capabilities of spike-driven learning and working memory in a unified single neuron with multiple timescale dynamics. The competitive performance of SAM could potentially contribute to the development of efficient adaptive neuromorphic computing systems for various applications from robotics to edge computing.
Collapse
Affiliation(s)
- Shuangming Yang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Tian Gao
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Jiang Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Bin Deng
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | | | - Tao Lei
- School of Electronic Information and Artificial Intelligence, Shaanxi University of Science and Technology, Xi’an, China
| | | |
Collapse
|
27
|
Marks WD, Yokose J, Kitamura T, Ogawa SK. Neuronal Ensembles Organize Activity to Generate Contextual Memory. Front Behav Neurosci 2022; 16:805132. [PMID: 35368306 PMCID: PMC8965349 DOI: 10.3389/fnbeh.2022.805132] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
Contextual learning is a critical component of episodic memory and important for living in any environment. Context can be described as the attributes of a location that are not the location itself. This includes a variety of non-spatial information that can be derived from sensory systems (sounds, smells, lighting, etc.) and internal state. In this review, we first address the behavioral underpinnings of contextual memory and the development of context memory theory, with a particular focus on the contextual fear conditioning paradigm as a means of assessing contextual learning and the underlying processes contributing to it. We then present the various neural centers that play roles in contextual learning. We continue with a discussion of the current knowledge of the neural circuitry and physiological processes that underlie contextual representations in the Entorhinal cortex-Hippocampal (EC-HPC) circuit, as the most well studied contributor to contextual memory, focusing on the role of ensemble activity as a representation of context with a description of remapping, and pattern separation and completion in the processing of contextual information. We then discuss other critical regions involved in contextual memory formation and retrieval. We finally consider the engram assembly as an indicator of stored contextual memories and discuss its potential contribution to contextual memory.
Collapse
Affiliation(s)
- William D. Marks
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jun Yokose
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sachie K. Ogawa
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
28
|
Eom K, Lee HR, Hyun JH, An H, Lee YS, Ho WK, Lee SH. Gradual decorrelation of CA3 ensembles associated with contextual discrimination learning is impaired by Kv1.2 insufficiency. Hippocampus 2022; 32:193-216. [PMID: 34964210 DOI: 10.1002/hipo.23400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/30/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022]
Abstract
The associative network of hippocampal CA3 is thought to contribute to rapid formation of contextual memory from one-trial learning, but the network mechanisms underlying decorrelation of neuronal ensembles in CA3 is largely unknown. Kv1.2 expressions in rodent CA3 pyramidal cells (CA3-PCs) are polarized to distal apical dendrites, and its downregulation specifically enhances dendritic responses to perforant pathway (PP) synaptic inputs. We found that haploinsufficiency of Kv1.2 (Kcna2+/-) in CA3-PCs, but not Kv1.1 (Kcna1+/-), lowers the threshold for long-term potentiation (LTP) at PP-CA3 synapses, and that the Kcna2+/- mice are normal in discrimination of distinct contexts but impaired in discrimination of similar but slightly distinct contexts. We further examined the neuronal ensembles in CA3 and dentate gyrus (DG), which represent the two similar contexts using in situ hybridization of immediate early genes, Homer1a and Arc. The size and overlap of CA3 ensembles activated by the first visit to the similar contexts were not different between wild type and Kcna2+/- mice, but these ensemble parameters diverged over training days between genotypes, suggesting that abnormal plastic changes at PP-CA3 synapses of Kcna2+/- mice is responsible for the impaired pattern separation. Unlike CA3, DG ensembles were not different between two genotype mice. The DG ensembles were already separated on the first day, and their overlap did not further evolve. Eventually, the Kcna2+/- mice exhibited larger CA3 ensemble size and overlap upon retrieval of two contexts, compared to wild type or Kcna1+/- mice. These results suggest that sparse LTP at PP-CA3 synapse probably supervised by mossy fiber inputs is essential for gradual decorrelation of CA3 ensembles.
Collapse
Affiliation(s)
- Kisang Eom
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyoung Ro Lee
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Ho Hyun
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunhoe An
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Brain and Cognitive Science, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| | - Yong-Seok Lee
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Won-Kyung Ho
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Brain and Cognitive Science, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| | - Suk-Ho Lee
- Cell Physiology Laboratory, Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Brain and Cognitive Science, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| |
Collapse
|
29
|
Sheintuch L, Rubin A, Ziv Y. Bias-free estimation of information content in temporally sparse neuronal activity. PLoS Comput Biol 2022; 18:e1009832. [PMID: 35148310 PMCID: PMC8836373 DOI: 10.1371/journal.pcbi.1009832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/13/2022] [Indexed: 11/20/2022] Open
Abstract
Applying information theoretic measures to neuronal activity data enables the quantification of neuronal encoding quality. However, when the sample size is limited, a naïve estimation of the information content typically contains a systematic overestimation (upward bias), which may lead to misinterpretation of coding characteristics. This bias is exacerbated in Ca2+ imaging because of the temporal sparsity of elevated Ca2+ signals. Here, we introduce methods to correct for the bias in the naïve estimation of information content from limited sample sizes and temporally sparse neuronal activity. We demonstrate the higher accuracy of our methods over previous ones, when applied to Ca2+ imaging data recorded from the mouse hippocampus and primary visual cortex, as well as to simulated data with matching tuning properties and firing statistics. Our bias-correction methods allowed an accurate estimation of the information place cells carry about the animal's position (spatial information) and uncovered the spatial resolution of hippocampal coding. Furthermore, using our methods, we found that cells with higher peak firing rates carry higher spatial information per spike and exposed differences between distinct hippocampal subfields in the long-term evolution of the spatial code. These results could be masked by the bias when applying the commonly used naïve calculation of information content. Thus, a bias-free estimation of information content can uncover otherwise overlooked properties of the neural code.
Collapse
Affiliation(s)
- Liron Sheintuch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Rubin
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Yaniv Ziv
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
30
|
Terada S, Geiller T, Liao Z, O'Hare J, Vancura B, Losonczy A. Adaptive stimulus selection for consolidation in the hippocampus. Nature 2022; 601:240-244. [PMID: 34880499 PMCID: PMC9380538 DOI: 10.1038/s41586-021-04118-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
Associative memories guide behavioural adaptation by binding together outcome-predictive sensory stimuli1,2. However, in a feature-rich environment, only a subset of stimuli may predict a desired outcome3,4. How neural circuits enable behavioural adaptation by selectively and durably representing subsets of sensory stimuli that are pertinent to a specific outcome is not known. We investigated this feature selection process in the hippocampus during memory acquisition and subsequent consolidation. Two-photon calcium imaging of CA3 axonal projections to CA1 combined with simultaneous local field potential recordings revealed that CA3 projections that encode behaviourally informative sensory stimuli were selectively recruited during the memory replay events that underlie hippocampal memory consolidation5. These axonal projections formed sequential assemblies that conjunctively link sensory features to spatial location and thus reward proximity. By contrast, axons encoding uninformative, peripatetic sensory cues were notably suppressed during memory replay. Thus, while the hippocampus comprehensively encodes the real-time sensory environment, it implements a flexible filtering mechanism to maximize the utility of memories destined for long-term storage. We propose that utility-dependent recruitment of sensory experience during memory consolidation is a general coding principle for associative learning.
Collapse
Affiliation(s)
- Satoshi Terada
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Tristan Geiller
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Zhenrui Liao
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Justin O'Hare
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Bert Vancura
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
- The Kavli Institute for Brain Science, Columbia University, New York, NY, USA.
| |
Collapse
|
31
|
Zhang HA, Yuan CX, Liu KF, Yang QF, Zhao J, Li H, Yang QH, Song D, Quan ZZ, Qing H. Neural stem cell transplantation alleviates functional cognitive deficits in a mouse model of tauopathy. Neural Regen Res 2022; 17:152-162. [PMID: 34100451 PMCID: PMC8451553 DOI: 10.4103/1673-5374.314324] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The mechanisms of the transplantation of neural stem cells (NSCs) in the treatment of Alzheimer’s disease remain poorly understood. In this study, NSCs were transplanted into the hippocampal CA1 region of the rTg (tau P301L) 4510 mouse model, a tauopathy model that is thought to reflect the tau pathology associated with Alzheimer’s disease. The results revealed that NSC transplantation reduced the abnormal aggregation of tau, resulting in significant improvements in the short-term memory of the tauopathy model mice. Compared with wild-type and phosphate-buffered saline (PBS)-treated mice, mice that received NSC transplantations were characterized by changes in the expression of multiple proteins in brain tissue, particularly those related to the regulation of tau aggregation or misfolding. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and Gene Ontology (GO) function analysis revealed that these proteins were primarily enriched in pathways associated with long-term potentiation, neurogenesis, and other neurobiological processes. Changes in the expression levels of key proteins were verified by western blot assays. These data provided clues to improve the understanding of the functional capacity associated with NSC transplantation in Alzheimer’s disease treatment. This study was approved by the Beijing Animal Ethics Association and Ethics Committee of Beijing Institute of Technology (approval No. SYXK-BIT-school of life science-2017-M03) in 2017.
Collapse
Affiliation(s)
- He-Ao Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Chun-Xu Yuan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Ke-Fu Liu
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Qi-Fan Yang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Juan Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Qing-Hu Yang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhen-Zhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
32
|
Remme MWH, Bergmann U, Alevi D, Schreiber S, Sprekeler H, Kempter R. Hebbian plasticity in parallel synaptic pathways: A circuit mechanism for systems memory consolidation. PLoS Comput Biol 2021; 17:e1009681. [PMID: 34874938 PMCID: PMC8683039 DOI: 10.1371/journal.pcbi.1009681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 12/17/2021] [Accepted: 11/24/2021] [Indexed: 12/03/2022] Open
Abstract
Systems memory consolidation involves the transfer of memories across brain regions and the transformation of memory content. For example, declarative memories that transiently depend on the hippocampal formation are transformed into long-term memory traces in neocortical networks, and procedural memories are transformed within cortico-striatal networks. These consolidation processes are thought to rely on replay and repetition of recently acquired memories, but the cellular and network mechanisms that mediate the changes of memories are poorly understood. Here, we suggest that systems memory consolidation could arise from Hebbian plasticity in networks with parallel synaptic pathways-two ubiquitous features of neural circuits in the brain. We explore this hypothesis in the context of hippocampus-dependent memories. Using computational models and mathematical analyses, we illustrate how memories are transferred across circuits and discuss why their representations could change. The analyses suggest that Hebbian plasticity mediates consolidation by transferring a linear approximation of a previously acquired memory into a parallel pathway. Our modelling results are further in quantitative agreement with lesion studies in rodents. Moreover, a hierarchical iteration of the mechanism yields power-law forgetting-as observed in psychophysical studies in humans. The predicted circuit mechanism thus bridges spatial scales from single cells to cortical areas and time scales from milliseconds to years.
Collapse
Affiliation(s)
- Michiel W. H. Remme
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Urs Bergmann
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Denis Alevi
- Department for Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Susanne Schreiber
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Henning Sprekeler
- Department for Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- Excellence Cluster Science of Intelligence, Berlin, Germany
| | - Richard Kempter
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
33
|
Learning exceptions to the rule in human and model via hippocampal encoding. Sci Rep 2021; 11:21429. [PMID: 34728698 PMCID: PMC8563716 DOI: 10.1038/s41598-021-00864-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/13/2021] [Indexed: 11/09/2022] Open
Abstract
Category learning helps us process the influx of information we experience daily. A common category structure is "rule-plus-exceptions," in which most items follow a general rule, but exceptions violate this rule. People are worse at learning to categorize exceptions than rule-following items, but improved exception categorization has been positively associated with hippocampal function. In light of model-based predictions that the nature of existing memories of related experiences impacts memory formation, here we use behavioural and computational modelling data to explore how learning sequence impacts performance in rule-plus-exception categorization. Our behavioural results indicate that exception categorization accuracy improves when exceptions are introduced later in learning, after exposure to rule-followers. To explore whether hippocampal learning systems also benefit from this manipulation, we simulate our task using a computational model of hippocampus. The model successful replicates our behavioural findings related to exception learning, and representational similarity analysis of the model's hidden layers suggests that model representations are impacted by trial sequence: delaying the introduction of an exception shifts its representation closer to its own category members. Our results provide novel computational evidence of how hippocampal learning systems can be targeted by learning sequence and bolster extant evidence of hippocampus's role in category learning.
Collapse
|
34
|
Hernández-Matias A, Bermúdez-Rattoni F, Osorio-Gómez D. Maintenance of conditioned place avoidance induced by gastric malaise requires NMDA activity within the ventral hippocampus. ACTA ACUST UNITED AC 2021; 28:270-276. [PMID: 34400528 PMCID: PMC8372560 DOI: 10.1101/lm.052720.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/28/2021] [Indexed: 11/24/2022]
Abstract
It has been reported that during chemotherapy treatment, some patients can experience nausea before pharmacological administration, suggesting that contextual stimuli are associated with the nauseating effects. There are attempts to reproduce with animal models the conditions under which this phenomenon is observed to provide a useful paradigm for studying contextual aversion learning and the brain structures involved. This manuscript assessed the hippocampus involvement in acquiring and maintaining long-term conditioned place avoidance (CPA) induced by a gastric malaise-inducing agent, LiCl. Our results demonstrate that a reliable induction of CPA is possible after one acquisition trial. However, CPA establishment requires a 20-min confinement in the compartment associated with LiCl administration. Interestingly, both hippocampal regions seem to be necessary for CPA establishment; nonetheless, inactivation of the ventral hippocampus results in a reversion of avoidance and turns it into preference. Moreover, we demonstrate that activation of dorsal/ventral hippocampal NMDA receptors after CS–US association is required for long-term CPA memory maintenance.
Collapse
Affiliation(s)
- Arturo Hernández-Matias
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Daniel Osorio-Gómez
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| |
Collapse
|
35
|
Cholinergic basal forebrain and hippocampal structure influence visuospatial memory in Parkinson's disease. Brain Imaging Behav 2021; 16:118-129. [PMID: 34176042 DOI: 10.1007/s11682-021-00481-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 10/21/2022]
Abstract
Visuospatial impairment in Parkinson's disease (PD) heralds the onset of a progressive dementia syndrome and might be associated with cholinergic dysfunction. It remains unclear however, whether degeneration of the cholinergic basal forebrain is directly related to cognitive decline, or whether relationships between this region and cognitive function are mediated by closely related brain structures such as those in the medial temporal lobe. To evaluate relationships between structure of the cholinergic basal forebrain, medial temporal lobe and cognition, 27 PD patients without dementia and 20 controls underwent neuropsychological assessment and MRI. Volumes of the cholinergic basal forebrain nuclei, the entorhinal cortex, the hippocampus and its subfields were measured. Regression models utilised basal forebrain and hippocampal volumetric measures to predict cognitive performance. In PD, visuospatial memory (but not verbal memory or executive function) was correlated with hippocampal volume, particularly CA2-3, and basal forebrain subregion Ch1-2, but not Ch4. In addition, hippocampal volume was correlated with Ch1-2 in PD. The relationship between Ch1-2 and visuospatial memory was mediated by CA2-3 integrity. There were no correlations between cognitive and volumetric measures in controls. Our data imply that the integrity of the cholinergic basal forebrain is associated with subregional hippocampal volume. Additionally, a relationship between visuospatial function and cholinergic nuclei does exist, but is fully mediated by variations in hippocampal structure. These findings are consistent with the recent hypothesis that forebrain cholinergic system degeneration results in cognitive deficits via cholinergic denervation, and subsequent structural degeneration, of its target regions.
Collapse
|
36
|
Prathap S, Nagel BJ, Herting MM. Understanding the role of aerobic fitness, spatial learning, and hippocampal subfields in adolescent males. Sci Rep 2021; 11:9311. [PMID: 33927247 PMCID: PMC8084987 DOI: 10.1038/s41598-021-88452-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/07/2021] [Indexed: 02/02/2023] Open
Abstract
Physical exercise during adolescence, a critical developmental window, can facilitate neurogenesis in the dentate gyrus and astrogliogenesis in Cornu Ammonis (CA) hippocampal subfields of rats, and which have been associated with improved hippocampal dependent memory performance. Recent translational studies in humans also suggest that aerobic fitness is associated with hippocampal volume and better spatial memory during adolescence. However, associations between fitness, hippocampal subfield morphology, and learning capabilities in human adolescents remain largely unknown. Employing a translational study design in 34 adolescent males, we explored the relationship between aerobic fitness, hippocampal subfield volumes, and both spatial and verbal memory. Aerobic fitness, assessed by peak oxygen utilization on a high-intensity exercise test (VO2 peak), was positively associated with the volumetric enlargement of the hippocampal head, and the CA1 head region specifically. Larger CA1 volumes were also associated with spatial learning on a Virtual Morris Water Maze task and verbal learning on the Rey Auditory Verbal Learning Test, but not recall memory. In line with previous animal work, the current findings lend support for the long-axis specialization of the hippocampus in the areas of exercise and learning during adolescence.
Collapse
Affiliation(s)
- Sandhya Prathap
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, 90023, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90023, USA
| | - Bonnie J Nagel
- Departments of Psychiatry and Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Megan M Herting
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, 90023, USA.
| |
Collapse
|
37
|
Theta Oscillations Coincide with Sustained Hyperpolarization in CA3 Pyramidal Cells, Underlying Decreased Firing. Cell Rep 2021; 32:107868. [PMID: 32640233 DOI: 10.1016/j.celrep.2020.107868] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/22/2020] [Accepted: 06/16/2020] [Indexed: 11/20/2022] Open
Abstract
Brain states modulate the membrane potential dynamics of neurons, influencing the functional repertoire of the network. Pyramidal cells (PCs) in the hippocampal CA3 are necessary for rapid memory encoding, which preferentially occurs during exploratory behavior in the high-arousal theta state. However, the relationship between the membrane potential dynamics of CA3 PCs and theta has not been explored. Here we characterize the changes in the membrane potential of PCs in relation to theta using electrophysiological recordings in awake mice. During theta, most PCs behave in a stereotypical manner, consistently hyperpolarizing time-locked to the duration of theta. Additionally, PCs display lower membrane potential variance and a reduced firing rate. In contrast, during large irregular activity, PCs show heterogeneous changes in membrane potential. This suggests coordinated hyperpolarization of PCs during theta, possibly caused by increased inhibition. This could lead to a higher signal-to-noise ratio in the small population of PCs active during theta, as observed in ensemble recordings.
Collapse
|
38
|
Abstract
Neural correlates of external variables provide potential internal codes that guide an animal’s behaviour. Notably, first-order features of neural activity, such as single-neuron firing rates, have been implicated in encoding information. However, the extent to which higher-order features, such as multi-neuron coactivity, play primary roles in encoding information or secondary roles in supporting single-neuron codes remains unclear. Here we show that millisecond-timescale coactivity amongst hippocampal CA1 neurons discriminates distinct millisecond-lived behavioural contingencies. This contingency discrimination was unrelated to the tuning of individual neurons but instead an emergent property of their coactivity. Contingency discriminating patterns were reactivated offline after learning and their reinstatement predicted trial-by-trial memory performance. Moreover, optogenetic suppression of inputs from the upstream CA3 region selectively during learning impaired coactivity-based contingency information in CA1 and subsequent dynamic memory retrieval. These findings identify coactivity as a primary feature of neural firing that discriminates distinct behaviourally-relevant variables and supports memory retrieval.
Collapse
|
39
|
Largo-Barrientos P, Apóstolo N, Creemers E, Callaerts-Vegh Z, Swerts J, Davies C, McInnes J, Wierda K, De Strooper B, Spires-Jones T, de Wit J, Uytterhoeven V, Verstreken P. Lowering Synaptogyrin-3 expression rescues Tau-induced memory defects and synaptic loss in the presence of microglial activation. Neuron 2021; 109:767-777.e5. [PMID: 33472038 PMCID: PMC7927913 DOI: 10.1016/j.neuron.2020.12.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 11/24/2020] [Accepted: 12/21/2020] [Indexed: 01/17/2023]
Abstract
Tau is a major driver of neurodegeneration and is implicated in over 20 diseases. Tauopathies are characterized by synaptic loss and neuroinflammation, but it is unclear if these pathological events are causally linked. Tau binds to Synaptogyrin-3 on synaptic vesicles. Here, we interfered with this function to determine the role of pathogenic Tau at pre-synaptic terminals. We show that heterozygous knockout of synaptogyrin-3 is benign in mice but strongly rescues mutant Tau-induced defects in long-term synaptic plasticity and working memory. It also significantly rescues the pre- and post-synaptic loss caused by mutant Tau. However, Tau-induced neuroinflammation remains clearly upregulated when we remove the expression of one allele of synaptogyrin-3. Hence neuroinflammation is not sufficient to cause synaptic loss, and these processes are separately induced in response to mutant Tau. In addition, the pre-synaptic defects caused by mutant Tau are enough to drive defects in cognitive tasks.
Collapse
Affiliation(s)
- Pablo Largo-Barrientos
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Nuno Apóstolo
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Eline Creemers
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | | | - Jef Swerts
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Caitlin Davies
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph McInnes
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joris de Wit
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium
| | - Valerie Uytterhoeven
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium.
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Leuven, Belgium.
| |
Collapse
|
40
|
Schlesiger MI, Ruff T, MacLaren DAA, Barriuso-Ortega I, Saidov KM, Yen TY, Monyer H. Two septal-entorhinal GABAergic projections differentially control coding properties of spatially tuned neurons in the medial entorhinal cortex. Cell Rep 2021; 34:108801. [PMID: 33657367 DOI: 10.1016/j.celrep.2021.108801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 12/23/2020] [Accepted: 02/05/2021] [Indexed: 12/20/2022] Open
Abstract
Septal parvalbumin-expressing (PV+) and calbindin-expressing (CB+) projections inhibit low-threshold and fast-spiking interneurons, respectively, in the medial entorhinal cortex (MEC). We investigate how the two inputs control neuronal activity in the MEC in freely moving mice. Stimulation of PV+ and CB+ terminals causes disinhibition of spatially tuned MEC neurons, but exerts differential effects on temporal coding and burst firing. Thus, recruitment of PV+ projections disrupts theta-rhythmic firing of MEC neurons, while stimulation of CB+ projections increases burst firing of grid cells and enhances phase precession in a cell-type-specific manner. Inactivation of septal PV+ or CB+ neurons differentially affects context, reference, and working memory. Together, our results reveal how specific connectivity of septal GABAergic projections with MEC interneurons translates into differential modulation of MEC neuronal coding.
Collapse
Affiliation(s)
- Magdalene Isabell Schlesiger
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Tobias Ruff
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Duncan Archibald Allan MacLaren
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Isabel Barriuso-Ortega
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Khalid Magomedovich Saidov
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ting-Yun Yen
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
41
|
Kim G, Kwon M, Kang W, Lee SH. Is Reconsolidation a General Property of Memory? Front Hum Neurosci 2021; 15:643106. [PMID: 33732126 PMCID: PMC7959766 DOI: 10.3389/fnhum.2021.643106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/09/2021] [Indexed: 11/13/2022] Open
Abstract
Memory reconsolidation holds great hope for memory modification approaches and clinical treatments of mental disorders associated with maladaptive memories. However, it remains controversial as to whether reconsolidation is a general property of all types of memory. Especially, discrepancies have been reported in research focusing on whether declarative memory undergoes reconsolidation, and whether old memories can be reorganized after retrieval. Here, we discuss how these inconsistent results can be reconciled and what information we need to uncover for the general use of reconsolidation.
Collapse
Affiliation(s)
- Gayoung Kim
- Department of Bio and Brain Engineering, College of Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Minjae Kwon
- Department of Bio and Brain Engineering, College of Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Wonjun Kang
- Department of Bio and Brain Engineering, College of Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Sue-Hyun Lee
- Department of Bio and Brain Engineering, College of Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.,Program of Brain and Cognitive Engineering, College of Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
42
|
Lenck-Santini PP, Sakkaki S. Alterations of Neuronal Dynamics as a Mechanism for Cognitive Impairment in Epilepsy. Curr Top Behav Neurosci 2021; 55:65-106. [PMID: 33454922 DOI: 10.1007/7854_2020_193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Epilepsy is commonly associated with cognitive and behavioral deficits that dramatically affect the quality of life of patients. In order to identify novel therapeutic strategies aimed at reducing these deficits, it is critical first to understand the mechanisms leading to cognitive impairments in epilepsy. Traditionally, seizures and epileptiform activity in addition to neuronal injury have been considered to be the most significant contributors to cognitive dysfunction. In this review we however highlight the role of a new mechanism: alterations of neuronal dynamics, i.e. the timing at which neurons and networks receive and process neural information. These alterations, caused by the underlying etiologies of epilepsy syndromes, are observed in both animal models and patients in the form of abnormal oscillation patterns in unit firing, local field potentials, and electroencephalogram (EEG). Evidence suggests that such mechanisms significantly contribute to cognitive impairment in epilepsy, independently of seizures and interictal epileptiform activity. Therefore, therapeutic strategies directly targeting neuronal dynamics rather than seizure reduction may significantly benefit the quality of life of patients.
Collapse
Affiliation(s)
- Pierre-Pascal Lenck-Santini
- Aix-Marseille Université, INSERM, INMED, Marseille, France. .,Department of Neurological sciences, University of Vermont, Burlington, VT, USA.
| | - Sophie Sakkaki
- Department of Neurological sciences, University of Vermont, Burlington, VT, USA.,Université de. Montpellier, CNRS, INSERM, IGF, Montpellier, France
| |
Collapse
|
43
|
Tessereau C, O’Dea R, Coombes S, Bast T. Reinforcement learning approaches to hippocampus-dependent flexible spatial navigation. Brain Neurosci Adv 2021; 5:2398212820975634. [PMID: 33954259 PMCID: PMC8042550 DOI: 10.1177/2398212820975634] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/21/2020] [Indexed: 11/17/2022] Open
Abstract
Humans and non-human animals show great flexibility in spatial navigation, including the ability to return to specific locations based on as few as one single experience. To study spatial navigation in the laboratory, watermaze tasks, in which rats have to find a hidden platform in a pool of cloudy water surrounded by spatial cues, have long been used. Analogous tasks have been developed for human participants using virtual environments. Spatial learning in the watermaze is facilitated by the hippocampus. In particular, rapid, one-trial, allocentric place learning, as measured in the delayed-matching-to-place variant of the watermaze task, which requires rodents to learn repeatedly new locations in a familiar environment, is hippocampal dependent. In this article, we review some computational principles, embedded within a reinforcement learning framework, that utilise hippocampal spatial representations for navigation in watermaze tasks. We consider which key elements underlie their efficacy, and discuss their limitations in accounting for hippocampus-dependent navigation, both in terms of behavioural performance (i.e. how well do they reproduce behavioural measures of rapid place learning) and neurobiological realism (i.e. how well do they map to neurobiological substrates involved in rapid place learning). We discuss how an actor-critic architecture, enabling simultaneous assessment of the value of the current location and of the optimal direction to follow, can reproduce one-trial place learning performance as shown on watermaze and virtual delayed-matching-to-place tasks by rats and humans, respectively, if complemented with map-like place representations. The contribution of actor-critic mechanisms to delayed-matching-to-place performance is consistent with neurobiological findings implicating the striatum and hippocampo-striatal interaction in delayed-matching-to-place performance, given that the striatum has been associated with actor-critic mechanisms. Moreover, we illustrate that hierarchical computations embedded within an actor-critic architecture may help to account for aspects of flexible spatial navigation. The hierarchical reinforcement learning approach separates trajectory control via a temporal-difference error from goal selection via a goal prediction error and may account for flexible, trial-specific, navigation to familiar goal locations, as required in some arm-maze place memory tasks, although it does not capture one-trial learning of new goal locations, as observed in open field, including watermaze and virtual, delayed-matching-to-place tasks. Future models of one-shot learning of new goal locations, as observed on delayed-matching-to-place tasks, should incorporate hippocampal plasticity mechanisms that integrate new goal information with allocentric place representation, as such mechanisms are supported by substantial empirical evidence.
Collapse
Affiliation(s)
- Charline Tessereau
- School of Mathematical Sciences, University of Nottingham, Nottingham, UK
- School of Psychology, University of Nottingham, Nottingham, UK
- Neuroscience@Nottingham
| | - Reuben O’Dea
- School of Mathematical Sciences, University of Nottingham, Nottingham, UK
- Neuroscience@Nottingham
| | - Stephen Coombes
- School of Mathematical Sciences, University of Nottingham, Nottingham, UK
- Neuroscience@Nottingham
| | - Tobias Bast
- School of Psychology, University of Nottingham, Nottingham, UK
- Neuroscience@Nottingham
| |
Collapse
|
44
|
Yamamoto N, Marks WD, Kitamura T. Cell-Type-Specific Optogenetic Techniques Reveal Neural Circuits Crucial for Episodic Memories. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:429-447. [PMID: 33398831 PMCID: PMC8612024 DOI: 10.1007/978-981-15-8763-4_28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The formation and maintenance of episodic memories are important for our daily life. Accumulating evidence from extensive studies with pharmacological, electrophysiological, and molecular biological approaches has shown that both entorhinal cortex (EC) and hippocampus (HPC) are crucial for the formation and recall of episodic memory. However, to further understand the neural mechanisms of episodic memory processes in the EC-HPC network, cell-type-specific manipulation of neural activity with high temporal resolution during memory process has become necessary. Recently, the technological innovation of optogenetics combined with pharmacological, molecular biological, and electrophysiological approaches has significantly advanced our understanding of the circuit mechanisms for learning and memory. Optogenetic techniques with transgenic mice and/or viral vectors enable us to manipulate the neural activity of specific cell populations as well as specific neural projections with millisecond-scale temporal control during animal behavior. Integrating optogenetics with drug-regulatable activity-dependent gene expression systems has identified memory engram cells, which are a subpopulation of cells that encode a specific episode. Finally, millisecond pulse stimulation of neural activity by optogenetics has further achieved (a) identification of synaptic connectivity between targeted pairs of neural populations, (b) cell-type-specific single-unit electrophysiological recordings, and (c) artificial induction and modification of synaptic plasticity in targeted synapses. In this chapter, we summarize technological and conceptual advancements in the field of neurobiology of learning and memory as revealed by optogenetic approaches in the rodent EC-HPC network for episodic memories.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - William D Marks
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
45
|
Choi I, Beedholm K, Dam VS, Bae SH, Noble DJ, Garraway SM, Aalkjaer C, Boedtkjer E. Sodium bicarbonate cotransporter NBCn1/Slc4a7 affects locomotor activity and hearing in mice. Behav Brain Res 2020; 401:113065. [PMID: 33321164 PMCID: PMC9069564 DOI: 10.1016/j.bbr.2020.113065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/19/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
Despite a widespread expression pattern in the central nervous system, the role of the sodium bicarbonate cotransporter NBCn1/Slc4a7 has not been investigated for locomotor activity, emotion and cognition. Here, we addressed the behavioral consequences of NBCn1 knockout and evaluated hearing and vision that are reportedly impaired in an earlier line of NBCn1 knockout mice and may contribute to behavioral changes. In a circular open field, the knockout mice traveled a shorter distance, especially in the periphery of the chamber, than wildtype littermates. The knockout mice also traveled a shorter total distance in a home cage-like open field. Rearing and grooming behaviors were reduced. The knockout and control mice displayed similar time spent and number of open and closed arms in the elevated plus maze test, indicating negligible change in anxiety. In the Morris water maze test, both groups of mice learned the location of an escape platform within comparable time on the training trials and showed similar platform identification on the probe trial. The knockout mice maintained normal visual responses in the optokinetic drum and produced evoked potentials in response to light stimuli. However, these mice failed to produce auditory evoked potentials. qPCR revealed a robust expression of an alternatively transcribed NBCn1 variant in the knockout mouse retina. These results indicate that NBCn1 deletion leads to reduced locomotor activity in mice by affecting their exploratory behaviors or emotionality. The deletion also causes hearing loss, but its effect on vision varies between different lines of knockout mice.
Collapse
Affiliation(s)
- Inyeong Choi
- Department of Physiology, Emory University School of Medicine, Atlanta, USA.
| | | | - Vibeke S Dam
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Seong-Ho Bae
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, USA
| | - Donald J Noble
- Department of Physiology, Emory University School of Medicine, Atlanta, USA
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, USA
| | | | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
46
|
Lana D, Ugolini F, Giovannini MG. Space-Dependent Glia-Neuron Interplay in the Hippocampus of Transgenic Models of β-Amyloid Deposition. Int J Mol Sci 2020; 21:E9441. [PMID: 33322419 PMCID: PMC7763751 DOI: 10.3390/ijms21249441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
This review is focused on the description and discussion of the alterations of astrocytes and microglia interplay in models of Alzheimer's disease (AD). AD is an age-related neurodegenerative pathology with a slowly progressive and irreversible decline of cognitive functions. One of AD's histopathological hallmarks is the deposition of amyloid beta (Aβ) plaques in the brain. Long regarded as a non-specific, mere consequence of AD pathology, activation of microglia and astrocytes is now considered a key factor in both initiation and progression of the disease, and suppression of astrogliosis exacerbates neuropathology. Reactive astrocytes and microglia overexpress many cytokines, chemokines, and signaling molecules that activate or damage neighboring cells and their mutual interplay can result in virtuous/vicious cycles which differ in different brain regions. Heterogeneity of glia, either between or within a particular brain region, is likely to be relevant in healthy conditions and disease processes. Differential crosstalk between astrocytes and microglia in CA1 and CA3 areas of the hippocampus can be responsible for the differential sensitivity of the two areas to insults. Understanding the spatial differences and roles of glia will allow us to assess how these interactions can influence the state and progression of the disease, and will be critical for identifying therapeutic strategies.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| |
Collapse
|
47
|
Howe T, Blockeel AJ, Taylor H, Jones MW, Bazhenov M, Malerba P. NMDA receptors promote hippocampal sharp-wave ripples and the associated coactivity of CA1 pyramidal cells. Hippocampus 2020; 30:1356-1370. [PMID: 33112474 PMCID: PMC8645203 DOI: 10.1002/hipo.23276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 10/05/2020] [Accepted: 10/15/2020] [Indexed: 10/10/2023]
Abstract
Hippocampal sharp-wave ripples (SWRs) support the reactivation of memory representations, relaying information to neocortex during "offline" and sleep-dependent memory consolidation. While blockade of NMDA receptors (NMDAR) is known to affect both learning and subsequent consolidation, the specific contributions of NMDAR activation to SWR-associated activity remain unclear. Here, we combine biophysical modeling with in vivo local field potential (LFP) and unit recording to quantify changes in SWR dynamics following inactivation of NMDAR. In a biophysical model of CA3-CA1 SWR activity, we find that NMDAR removal leads to reduced SWR density, but spares SWR properties such as duration, cell recruitment and ripple frequency. These predictions are confirmed by experiments in which NMDAR-mediated transmission in rats was inhibited using three different NMDAR antagonists, while recording dorsal CA1 LFP. In the model, loss of NMDAR-mediated conductances also induced a reduction in the proportion of cell pairs that co-activate significantly above chance across multiple events. Again, this prediction is corroborated by dorsal CA1 single-unit recordings, where the NMDAR blocker ketamine disrupted correlated spiking during SWR. Our results are consistent with a framework in which NMDA receptors both promote activation of SWR events and organize SWR-associated spiking content. This suggests that, while SWR are short-lived events emerging in fast excitatory-inhibitory networks, slower network components including NMDAR-mediated currents contribute to ripple density and promote consistency in the spiking content across ripples, underpinning mechanisms for fine-tuning of memory consolidation processes.
Collapse
Affiliation(s)
- Timothy Howe
- School of Physiology, Pharmacology and Neuroscience,
University of Bristol, Bristol, UK
| | - Anthony J. Blockeel
- School of Physiology, Pharmacology and Neuroscience,
University of Bristol, Bristol, UK
| | - Hannah Taylor
- School of Physiology, Pharmacology and Neuroscience,
University of Bristol, Bristol, UK
| | - Matthew W. Jones
- School of Physiology, Pharmacology and Neuroscience,
University of Bristol, Bristol, UK
| | - Maxim Bazhenov
- Department of Medicine, University of California San Diego,
La Jolla, California
| | - Paola Malerba
- School of Physiology, Pharmacology and Neuroscience,
University of Bristol, Bristol, UK
- Battelle Center for Mathematical Medicine, Columbus,
Ohio
| |
Collapse
|
48
|
Piette C, Touboul J, Venance L. Engrams of Fast Learning. Front Cell Neurosci 2020; 14:575915. [PMID: 33250712 PMCID: PMC7676431 DOI: 10.3389/fncel.2020.575915] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/24/2020] [Indexed: 01/22/2023] Open
Abstract
Fast learning designates the behavioral and neuronal mechanisms underlying the acquisition of a long-term memory trace after a unique and brief experience. As such it is opposed to incremental, slower reinforcement or procedural learning requiring repetitive training. This learning process, found in most animal species, exists in a large spectrum of natural behaviors, such as one-shot associative, spatial, or perceptual learning, and is a core principle of human episodic memory. We review here the neuronal and synaptic long-term changes associated with fast learning in mammals and discuss some hypotheses related to their underlying mechanisms. We first describe the variety of behavioral paradigms used to test fast learning memories: those preferentially involve a single and brief (from few hundred milliseconds to few minutes) exposures to salient stimuli, sufficient to trigger a long-lasting memory trace and new adaptive responses. We then focus on neuronal activity patterns observed during fast learning and the emergence of long-term selective responses, before documenting the physiological correlates of fast learning. In the search for the engrams of fast learning, a growing body of evidence highlights long-term changes in gene expression, structural, intrinsic, and synaptic plasticities. Finally, we discuss the potential role of the sparse and bursting nature of neuronal activity observed during the fast learning, especially in the induction plasticity mechanisms leading to the rapid establishment of long-term synaptic modifications. We conclude with more theoretical perspectives on network dynamics that could enable fast learning, with an overview of some theoretical approaches in cognitive neuroscience and artificial intelligence.
Collapse
Affiliation(s)
- Charlotte Piette
- Center for Interdisciplinary Research in Biology, College de France, INSERM U1050, CNRS UMR7241, Université PSL, Paris, France.,Department of Mathematics and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States
| | - Jonathan Touboul
- Department of Mathematics and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, United States
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology, College de France, INSERM U1050, CNRS UMR7241, Université PSL, Paris, France
| |
Collapse
|
49
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
50
|
Fielder E, Tweedy C, Wilson C, Oakley F, LeBeau FEN, Passos JF, Mann DA, von Zglinicki T, Jurk D. Anti-inflammatory treatment rescues memory deficits during aging in nfkb1 -/- mice. Aging Cell 2020; 19:e13188. [PMID: 32915495 PMCID: PMC7576267 DOI: 10.1111/acel.13188] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/29/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation is a common feature of many age-related conditions including neurodegenerative diseases such as Alzheimer's disease. Cellular senescence is a state of irreversible cell-cycle arrest, thought to contribute to neurodegenerative diseases partially via induction of a chronic pro-inflammatory phenotype. In this study, we used a mouse model of genetically enhanced NF-κB activity (nfκb1-/- ), characterized by low-grade chronic inflammation and premature aging, to investigate the impact of inflammaging on cognitive decline. We found that during aging, nfkb1-/- mice show an early onset of memory loss, combined with enhanced neuroinflammation and increased frequency of senescent cells in the hippocampus and cerebellum. Electrophysiological measurements in the hippocampus of nfkb1-/- mice in vitro revealed deficits in gamma frequency oscillations, which could explain the decline in memory capacity. Importantly, treatment with the nonsteroidal anti-inflammatory drug (NASID) ibuprofen reduced neuroinflammation and senescent cell burden resulting in significant improvements in cognitive function and gamma frequency oscillations. These data support the hypothesis that chronic inflammation is a causal factor in the cognitive decline observed during aging.
Collapse
Affiliation(s)
- Edward Fielder
- Biosciences InstituteAgeing Research LaboratoriesCampus for Ageing and VitalityNewcastle UniversityNewcastle upon TyneUK
| | - Clare Tweedy
- Biosciences InstituteFaculty of Medical SciencesNewcastle UniversityNewcastleUK
| | - Caroline Wilson
- Bioscience InstituteImmunity and InflammationNewcastle Fibrosis Research GroupFaculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Fiona Oakley
- Bioscience InstituteImmunity and InflammationNewcastle Fibrosis Research GroupFaculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Fiona E. N. LeBeau
- Biosciences InstituteFaculty of Medical SciencesNewcastle UniversityNewcastleUK
| | - João F. Passos
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMNUSA
| | - Derek A. Mann
- Bioscience InstituteImmunity and InflammationNewcastle Fibrosis Research GroupFaculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Thomas von Zglinicki
- Biosciences InstituteAgeing Research LaboratoriesCampus for Ageing and VitalityNewcastle UniversityNewcastle upon TyneUK
| | - Diana Jurk
- Biosciences InstituteAgeing Research LaboratoriesCampus for Ageing and VitalityNewcastle UniversityNewcastle upon TyneUK
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMNUSA
| |
Collapse
|