1
|
Kim S, Shin JJ, Kang M, Yang Y, Cho YS, Paik H, Kim J, Yi Y, Lee S, Koo HY, Bok J, Bae YC, Kim JY, Kim E. Alternatively spliced mini-exon B in PTPδ regulates excitatory synapses through cell-type-specific trans-synaptic PTPδ-IL1RAP interaction. Nat Commun 2025; 16:4415. [PMID: 40360498 PMCID: PMC12075705 DOI: 10.1038/s41467-025-59685-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
PTPδ, encoded by PTPRD, is implicated in various neurological, psychiatric, and neurodevelopmental disorders, but the underlying mechanisms remain unclear. PTPδ trans-synaptically interacts with multiple postsynaptic adhesion molecules, which involves its extracellular alternatively spliced mini-exons, meA and meB. While PTPδ-meA functions have been studied in vivo, PTPδ-meB has not been studied. Here, we report that, unlike homozygous PTPδ-meA-mutant mice, homozygous PTPδ-meB-mutant (Ptprd-meB-/-) mice show markedly reduced early postnatal survival. Heterozygous Ptprd-meB+/- male mice show behavioral abnormalities and decreased excitatory synaptic density and transmission in dentate gyrus granule cells (DG-GCs). Proteomic analyses identify decreased postsynaptic density levels of IL1RAP, a known trans-synaptic partner of meB-containing PTPδ. Accordingly, IL1RAP-mutant mice show decreased excitatory synaptic transmission in DG-GCs. Ptprd-meB+/- DG interneurons with minimal IL1RAP expression show increased excitatory synaptic density and transmission. Therefore, PTPδ-meB is important for survival, synaptic, and behavioral phenotypes and regulates excitatory synapses in cell-type-specific and IL1RAP-dependent manners.
Collapse
Affiliation(s)
- Seoyeong Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Jae Jin Shin
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Muwon Kang
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yeji Yang
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, 28119, Korea
| | - Yi Sul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Hyojung Paik
- Center for Biomedical Computing, Korea Institute of Science and Technology Information (KISTI), Daejeon, 34141, Korea
| | - Jimin Kim
- Center for Biomedical Computing, Korea Institute of Science and Technology Information (KISTI), Daejeon, 34141, Korea
| | - Yunho Yi
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Suho Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Hei Yeun Koo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, 28119, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea.
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea.
| |
Collapse
|
2
|
Severin D, Koh MT, Moreno C, Contreras D, Contreras A, Wesselborg C, Bridi M, Atufa J, Branch A, Worley P, Gallagher M, Kirkwood A. NPTX2 transfection improves synaptic E/I balance and performance in learning impaired aged rats. Prog Neurobiol 2025; 247:102746. [PMID: 40057261 DOI: 10.1016/j.pneurobio.2025.102746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Excessive neural activity in the medial temporal lobe commonly associates with cognitive decline in elderly humans and also in rodents.An attractive model pathway to study synaptic mechanisms underlying age-dependent circuit hyperexcitability is the connection made by lateral entorhinal cortex cells onto the dentate gyrus (LEC→DG). Both structures are particularly affected by age and, importantly, in behaviorally characterized aged rats, learning impairment correlates with diminished feedforward inhibition of granule cells recruited by LEC inputs. In this rat model of aging, we evaluated how overexpression of Neuronal Pentraxin 2 (NPTX2) in the LEC, essential for stabilizing excitatory inputs onto fast-spiking inhibitory interneurons (FS-INs), enhances feedforward inhibition and improves spatial memory in impaired individuals. In addition, we found that FS-INs from unimpaired aged individuals have an increased excitatory drive compared to young individuals. These findings support the notion that NPTX2-mediated compensatory mechanisms to enhance the recruitment of FS-INs are crucial to maintaining proficient memory performance during aging.
Collapse
Affiliation(s)
- Daniel Severin
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Ming Teng Koh
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Cristian Moreno
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Darwin Contreras
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Altagracia Contreras
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Christian Wesselborg
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Michelle Bridi
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Jala Atufa
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Audrey Branch
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Paul Worley
- Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA.
| | - Alfredo Kirkwood
- Mind/Brain Institute and Department of Neurosciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA; Department of Psychological and Brain Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA.
| |
Collapse
|
3
|
Müller-Komorowska D, Fujishige T, Fukai T. Recurrent Interneuron Connectivity Does Not Support Synchrony in a Biophysical Dentate Gyrus Model. eNeuro 2025; 12:ENEURO.0097-25.2025. [PMID: 40175092 PMCID: PMC12017885 DOI: 10.1523/eneuro.0097-25.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/07/2025] [Indexed: 04/04/2025] Open
Abstract
Synchronous activity of neuronal networks is found in many brain areas and correlates with cognition and behavior. Gamma synchrony is particularly strong in the dentate gyrus, which is thought to process contextual information in the hippocampus. Several network mechanisms for synchrony generation have been proposed and studied computationally. One such mechanism relies solely on recurrent inhibitory interneuron connectivity, but it requires a large enough number of synapses. Here, we incorporate previously published connectivity data of the dentate gyrus from mice of either sex into a biophysical computational model to test its ability to generate synchronous activity. We find that recurrent interneuron connectivity is insufficient to induce synchronous activity. This applies to an interneuron ring network and the broader dentate gyrus circuitry. Despite asynchronous input, recurrent interneuron connectivity can have small synchronizing effects but can also desynchronize the network for some types of synaptic input. Our results suggest that biologically plausible recurrent inhibitory connectivity alone is likely insufficient to synchronize the dentate gyrus.
Collapse
|
4
|
Santander O, Arredondo SB, García-Rojas F, Estay SF, Belforte JE, Chávez AE, Varela-Nallar L, Fuenzalida M. Ketamine administration during adolescence impairs synaptic integration and inhibitory synaptic transmission in the adult dentate gyrus. Prog Neurobiol 2025; 246:102718. [PMID: 39855537 DOI: 10.1016/j.pneurobio.2025.102718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/10/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Ketamine administration during adolescence affects cognitive performance; however, its long-term impact on synaptic function and neuronal integration in the hippocampus a brain region critical for cognition remains unclear. Using functional and molecular analyses, we found that chronic ketamine administration during adolescence exerts long-term effects on synaptic integration, expanding the temporal window in an input-specific manner affecting the inner molecular layer but not the medial perforant path inputs in the adult mouse dorsal hippocampal dentate gyrus. Ketamine also alters the excitatory/inhibitory balance by reducing the efficacy of inhibitory inputs likely due to a reduction in parvalbumin-positive interneurons number and function. These findings indicate that during adolescence, ketamine exerts a strong effect on inhibitory synaptic function mediated by parvalbumin-positive neurons that ultimately impact synaptic integration in the dorsal adult dentate gyrus, which could help to understand the neurobiological and functional bases that confer greater vulnerability to the adolescent brain.
Collapse
Affiliation(s)
- Odra Santander
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile; Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Sebastián B Arredondo
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago, Chile
| | - Francisca García-Rojas
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Sebastián F Estay
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile; Instituto de Neurociencias y Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Juan E Belforte
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - Andrés E Chávez
- Instituto de Neurociencias y Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Lorena Varela-Nallar
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
| |
Collapse
|
5
|
Liu B, White AJ, Lo CC. Augmenting flexibility: mutual inhibition between inhibitory neurons expands functional diversity. iScience 2025; 28:111718. [PMID: 39898045 PMCID: PMC11787539 DOI: 10.1016/j.isci.2024.111718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/06/2024] [Accepted: 12/27/2024] [Indexed: 02/04/2025] Open
Abstract
Recent advances in microcircuit analysis of nervous systems have revealed a plethora of mutual connections between inhibitory interneurons across many different species and brain regions. The abundance of these mutual connections has not been fully explained. Strikingly, we show that neural circuits with mutually inhibitory connections are able to rapidly and flexibly switch between distinct functions. That is, multiple functions coexist for a single set of synaptic weights. Here, we develop a theoretical framework to explain how inhibitory recurrent circuits give rise to this flexibility and show that mutual inhibition doubles the number of cusp bifurcations in small neural circuits. As a concrete example, we study a class of functional motifs we call coupled recurrent inhibitory and recurrent excitatory loops (CRIRELs). These CRIRELs have the advantage of being both multi-functional and controllable, performing a plethora of functions, including decisions, memory, toggle, and so forth. Finally, we demonstrate how mutual inhibition maximizes storage capacity for larger networks.
Collapse
Affiliation(s)
- Belle Liu
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu City 30080, Taiwan
- Department of Physics, National Tsing Hua University, Hsinchu City 30080, Taiwan
| | - Alexander James White
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu City 30080, Taiwan
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu City 30080, Taiwan
| | - Chung-Chuan Lo
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu City 30080, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu City 30080, Taiwan
| |
Collapse
|
6
|
Moseley SM, Meliza CD. A Complex Acoustical Environment During Development Enhances Auditory Perception and Coding Efficiency in the Zebra Finch. J Neurosci 2025; 45:e1269242024. [PMID: 39730206 PMCID: PMC11823350 DOI: 10.1523/jneurosci.1269-24.2024] [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: 07/03/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/29/2024] Open
Abstract
Sensory experience during development has lasting effects on perception and neural processing. Exposing juvenile animals to artificial stimuli influences the tuning and functional organization of the auditory cortex, but less is known about how the rich acoustical environments experienced by vocal communicators affect the processing of complex vocalizations. Here, we show that in zebra finches (Taeniopygia guttata), a colonial-breeding songbird species, exposure to a naturalistic social-acoustical environment during development has a profound impact on auditory perceptual behavior and on cortical-level auditory responses to conspecific song. Compared to birds raised by pairs in acoustic isolation, male and female birds raised in a breeding colony were better in an operant discrimination task at recognizing conspecific songs with and without masking colony noise. Neurons in colony-reared birds had higher average firing rates, selectivity, and discriminability, especially in the narrow-spiking, putatively inhibitory neurons of a higher-order auditory area, the caudomedial nidopallium (NCM). Neurons in colony-reared birds were also less correlated in their tuning, more efficient at encoding the spectrotemporal structure of conspecific song, and better at filtering out masking noise. These results suggest that the auditory cortex adapts to noisy, complex acoustical environments by strengthening inhibitory circuitry, functionally decoupling excitatory neurons while maintaining overall excitatory-inhibitory balance.
Collapse
Affiliation(s)
- Samantha M Moseley
- Department of Psychology, University of Virginia, Charlottesville, Virginia 22904
| | - C Daniel Meliza
- Department of Psychology, University of Virginia, Charlottesville, Virginia 22904
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|
7
|
Allami P, Yazdanpanah N, Rezaei N. The role of neuroinflammation in PV interneuron impairments in brain networks; implications for cognitive disorders. Rev Neurosci 2025:revneuro-2024-0153. [PMID: 39842401 DOI: 10.1515/revneuro-2024-0153] [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: 10/22/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Fast spiking parvalbumin (PV) interneuron is an inhibitory gamma-aminobutyric acid (GABA)ergic interneuron diffused in different brain networks, including the cortex and hippocampus. As a key component of brain networks, PV interneurons collaborate in fundamental brain functions such as learning and memory by regulating excitation and inhibition (E/I) balance and generating gamma oscillations. The unique characteristics of PV interneurons, like their high metabolic demands and long branching axons, make them too vulnerable to stressors. Neuroinflammation is one of the most significant stressors that have an adverse, long-lasting impact on PV interneurons. Neuroinflammation affects PV interneurons through specialized inflammatory pathways triggered by cytokines such as tumor necrosis factor (TNF) and interleukin 6 (IL-6). The crucial cells in neuroinflammation, microglia, also play a significant role. The destructive effect of inflammation on PV interneurons can have comprehensive effects and cause neurological disorders such as schizophrenia, Alzheimer's disease (AD), autism spectrum disorder (ASD), and bipolar disorder. In this article, we provide a comprehensive review of mechanisms in which neuroinflammation leads to PV interneuron hypofunction in these diseases. The integrated knowledge about the role of PV interneurons in cognitive networks of the brain and mechanisms involved in PV interneuron impairment in the pathology of these diseases can help us with better therapeutic interventions.
Collapse
Affiliation(s)
- Pantea Allami
- Student's Scientific Research Center, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
| | - Niloufar Yazdanpanah
- Student's Scientific Research Center, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences, Children's Medical Center Hospital , Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences, Children's Medical Center Hospital , Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran 1416634793, Tehran, Iran
| |
Collapse
|
8
|
Lee SH, Kang YJ, Smith BN. Activation of hypoactive parvalbumin-positive fast-spiking interneurons restores dentate inhibition to reduce electrographic seizures in the mouse intrahippocampal kainate model of temporal lobe epilepsy. Neurobiol Dis 2024; 203:106737. [PMID: 39542222 DOI: 10.1016/j.nbd.2024.106737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/20/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
Parvalbumin-positive (PV+) GABAergic interneurons in the dentate gyrus provide powerful perisomatic inhibition of dentate granule cells (DGCs) to prevent overexcitation and maintain the stability of dentate gyrus circuits. Most dentate PV+ interneurons survive status epilepticus, but surviving PV+ interneuron mediated inhibition is compromised in the dentate gyrus shortly after status epilepticus, contributing to epileptogenesis in temporal lobe epilepsy. It is uncertain whether the impaired activity of dentate PV+ interneurons recovers at later times or if it continues for months following status epilepticus. The development of compensatory modifications related to PV+ interneuron circuits in the months following status epilepticus is unknown, although reduced dentate GABAergic inhibition persists long after status epilepticus. We employed whole-cell patch-clamp recordings from dentate PV+ interneurons and DGCs in slices from male and female sham controls and intrahippocampal kainate (IHK) treated mice that developed spontaneous seizures months after status epilepticus to study epilepsy-associated changes in dentate PV+ interneuron circuits. Electrical recordings showed that: 1) Action potential firing rates of dentate PV+ interneurons were reduced in IHK treated mice up to four months after status epilepticus; 2) spontaneous inhibitory postsynaptic currents (sIPSCs) in DGCs exhibited reduced frequency but increased amplitude in IHK treated mice; and 3) the amplitude of IPSCs in DGCs evoked by optogenetic activation of dentate PV+ cells was upregulated without changes in short-term plasticity. Video-EEG recordings revealed that IHK treated mice showed spontaneous electrographic seizures in the dentate gyrus and that chemogenetic activation of PV+ interneurons abolished electrographic seizures. Our results suggest not only that the compensatory changes in PV+ interneuron circuits develop after IHK treatment, but also that increased PV+ interneuron mediated inhibition in the dentate gyrus may compensate for cell loss and reduced intrinsic excitability of dentate PV+ interneurons to stop seizures in temporal lobe epilepsy.
Collapse
Affiliation(s)
- Sang-Hun Lee
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Young-Jin Kang
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Bret N Smith
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
9
|
Leana-Sandoval G, Kolli AV, Chinn CA, Madrid A, Lo I, Sandoval MA, Vera VA, Simms J, Wood MA, Diaz-Alonso J. The GluA1 cytoplasmic tail regulates intracellular AMPA receptor trafficking and synaptic transmission onto dentate gyrus GABAergic interneurons, gating response to novelty. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626277. [PMID: 39677714 PMCID: PMC11643017 DOI: 10.1101/2024.12.01.626277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The GluA1 subunit, encoded by the putative schizophrenia-associated gene GRIA1, is required for activity-regulated AMPA receptor (AMPAR) trafficking, and plays a key role in cognitive and affective function. The cytoplasmic, carboxy-terminal domain (CTD) is the most divergent region across AMPAR subunits. The GluA1 CTD has received considerable attention for its role during long-term potentiation (LTP) at CA1 pyramidal neuron synapses. However, its function at other synapses and, more broadly, its contribution to different GluA1-dependent processes, is poorly understood. Here, we used mice with a constitutive truncation of the GluA1 CTD to dissect its role regulating AMPAR localization and function as well as its contribution to cognitive and affective processes. We found that GluA1 CTD truncation affected AMPAR subunit levels and intracellular trafficking. ΔCTD GluA1 mice exhibited no memory deficits, but presented exacerbated novelty-induced hyperlocomotion and dentate gyrus granule cell (DG GC) hyperactivity, among other behavioral alterations. Mechanistically, we found that AMPAR EPSCs onto DG GABAergic interneurons were significantly reduced, presumably underlying, at least in part, the observed changes in neuronal activity and behavior. In summary, this study dissociates CTD-dependent from CTD-independent GluA1 functions, unveiling the GluA1 CTD as a crucial hub regulating AMPAR function in a cell type-specific manner.
Collapse
Affiliation(s)
- Gerardo Leana-Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Ananth V Kolli
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Carlene A Chinn
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Alexis Madrid
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Iris Lo
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Matthew A Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Vanessa Alizo Vera
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Jeffrey Simms
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Marcelo A Wood
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Javier Diaz-Alonso
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| |
Collapse
|
10
|
Siwiec M, Bobula B, Kielbinski M, Multan N, Hess G, Tokarski K. Activation of 5-HT 7 receptors in the mouse dentate gyrus does not affect theta-burst-induced plasticity at the perforant path synapse. Pharmacol Rep 2024; 76:1377-1389. [PMID: 39487932 PMCID: PMC11582198 DOI: 10.1007/s43440-024-00674-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND The study examined the effects of 5-HT7 receptor activation on GABAergic transmission within the dentate gyrus and plasticity at the glutamatergic perforant path input. METHODS Immunofluorescence imaging was performed using transverse hippocampal slices from transgenic mice expressing green fluorescent protein (GFP) under the Htr7 promoter. This was followed by whole-cell patch clamp electrophysiological recordings assessing the effects of pharmacologically activating 5-HT7 receptors on spontaneous inhibitory postsynaptic currents recorded from dentate granule cells and hilar mossy cells-two glutamatergic neuron types present in the dentate gyrus. Extracellular recordings of field excitatory postsynaptic potentials were then performed to assess whether 5-HT7 receptor activation influenced theta-burst stimulation-evoked plasticity of the perforant path synaptic input. RESULTS It was found that parvalbumin and somatostatin interneurons in the dentate gyrus expressed GFP, which suggests they express 5-HT7 receptors. However, activation of 5-HT7 receptors had no effect on GABAergic transmission targeting mossy cells or granule cells. There was also no effect of 5-HT7 receptor activation on perforant path plasticity either with intact or blocked GABAA receptor signaling. CONCLUSION The presence of 5-HT7 receptors in a subset of parvalbumin and somatostatin interneurons in the mouse dentate gyrus could mean that they are involved in the inhibitory control of dentate gyrus activity. However, this potential effect was not evident in slice recordings of inhibitory transmission targeting principal cells and did not affect perforant path plasticity. Further experiments are needed to fully elucidate the functional role of these receptors in the dentate gyrus.
Collapse
Affiliation(s)
- Marcin Siwiec
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| | - Bartosz Bobula
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Michal Kielbinski
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Nikola Multan
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Grzegorz Hess
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Krzysztof Tokarski
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| |
Collapse
|
11
|
Rozov A, Jappy DJ, Maltseva K, Vazetdinova A, Valiullina-Rakhmatullina F. Dialectics of perisomatic inhibition-The unity and conflict of opposites. Front Neural Circuits 2024; 18:1494300. [PMID: 39534761 PMCID: PMC11554531 DOI: 10.3389/fncir.2024.1494300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Over the past three decades, a great deal of attention has been paid to the study of perisomatic inhibition and perisomatic inhibitory basket cells. A growing body of experimental evidence points to the leading role of perisomatic inhibitory cells in the generation of oscillatory activity in various frequency ranges. Recently the link between the activity of basket cells and complex behavior has been demonstrated in several laboratories. However, all this is true only for one type of perisomatic inhibitory interneuron-parvalbumin-positive basket cells. Nevertheless, where parvalbumin-positive basket cells are found, there is another type of basket cell, cholecystokinin-positive interneurons. These two types of interneurons share a number of common features: they innervate the same compartments of target neurons and they often receive excitation from the same sources, but they also differ from each other in the synchrony of their GABA release and expression of receptors. The functional role of cholecystokinin-positive basket cells in oscillatory activity is not so obvious. They were thought to be involved in theta oscillations, however recent measurements in free moving animals have put some doubts on this hypothesis. Therefore, an important question is, whether these two types of basket cells work synergistically or perform opposing actions in functional networks? In this mini-review, we attempt to answer this question by putting forward the idea that these two types of basket cells are functionally united as two entities of the same network, and their opposing actions are necessary to maintain rhythmogenesis in a "healthy", physiological range.
Collapse
Affiliation(s)
- Andrei Rozov
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny Novgorod, Russia
| | - David John Jappy
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny Novgorod, Russia
| | - Ksenia Maltseva
- Institute of Neuroscience, Lobachevsky State University of Nizhniy Novgorod, Nizhny Novgorod, Russia
| | - Alina Vazetdinova
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Fliza Valiullina-Rakhmatullina
- Laboratory of Electrophysiology, Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
12
|
Moseley SM, Meliza CD. A complex acoustical environment during development enhances auditory perception and coding efficiency in the zebra finch. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600670. [PMID: 38979160 PMCID: PMC11230381 DOI: 10.1101/2024.06.25.600670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Sensory experience during development has lasting effects on perception and neural processing. Exposing juvenile animals to artificial stimuli influences the tuning and functional organization of the auditory cortex, but less is known about how the rich acoustical environments experienced by vocal communicators affect the processing of complex vocalizations. Here, we show that in zebra finches (Taeniopygia guttata), a colonial-breeding songbird species, exposure to a naturalistic social-acoustical environment during development has a profound impact on auditory perceptual behavior and on cortical-level auditory responses to conspecific song. Compared to birds raised by pairs in acoustic isolation, male and female birds raised in a breeding colony were better in an operant discrimination task at recognizing conspecific songs with and without masking colony noise. Neurons in colony-reared birds had higher average firing rates, selectivity, and discriminability, especially in the narrow-spiking, putatively inhibitory neurons of a higher-order auditory area, the caudomedial nidopallium (NCM). Neurons in colony-reared birds were also less correlated in their tuning and more efficient at encoding the spectrotemporal structure of conspecific song, and better at filtering out masking noise. These results suggest that the auditory cortex adapts to noisy, complex acoustical environments by strengthening inhibitory circuitry, functionally decoupling excitatory neurons while maintaining overall excitatory-inhibitory balance.
Collapse
Affiliation(s)
- Samantha M Moseley
- Department of Psychology, University of Virginia, Charlottesville VA 22904, USA
| | - C Daniel Meliza
- Department of Psychology, University of Virginia, Charlottesville VA 22904, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville VA 22904, USA
| |
Collapse
|
13
|
Wanjia G, Han S, Kuhl BA. Repulsion of CA3 / dentate gyrus representations is driven by distinct internal beliefs in the face of ambiguous sensory input. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619862. [PMID: 39484581 PMCID: PMC11527005 DOI: 10.1101/2024.10.23.619862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Recent human neuroimaging studies of episodic memory have revealed a counterintuitive phenomenon in the hippocampus: when events are highly similar, corresponding hippocampal activity patterns are sometimes less correlated than activity patterns associated with unrelated events. This phenomenon-repulsion-is not accounted for by most theories of the hippocampus, and the conditions that trigger repulsion remain poorly understood. Here, we used a spatial route-learning task and high-resolution fMRI in humans to test whether hippocampal repulsion is fundamentally driven by internal beliefs about the environment. By precisely measuring participants' internal beliefs and actively manipulating them, we show that repulsion selectively occurred in hippocampal subfields CA3 and dentate gyrus when visual input was ambiguous-or even identical-but internal beliefs were distinct. These findings firmly establish conditions that elicit repulsion and have broad relevance to theories of hippocampal function and to the fields of human episodic memory and rodent spatial navigation.
Collapse
Affiliation(s)
- Guo Wanjia
- University of Oregon, Department of Psychology & Institute of Neuroscience
| | - Subin Han
- University of Virginia, Department of Psychology
| | - Brice A Kuhl
- University of Oregon, Department of Psychology & Institute of Neuroscience
| |
Collapse
|
14
|
Bocchio M, Vorobyev A, Sadeh S, Brustlein S, Dard R, Reichinnek S, Emiliani V, Baude A, Clopath C, Cossart R. Functional networks of inhibitory neurons orchestrate synchrony in the hippocampus. PLoS Biol 2024; 22:e3002837. [PMID: 39401246 PMCID: PMC11501041 DOI: 10.1371/journal.pbio.3002837] [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: 05/16/2024] [Revised: 10/24/2024] [Accepted: 09/06/2024] [Indexed: 10/26/2024] Open
Abstract
Inhibitory interneurons are pivotal components of cortical circuits. Beyond providing inhibition, they have been proposed to coordinate the firing of excitatory neurons within cell assemblies. While the roles of specific interneuron subtypes have been extensively studied, their influence on pyramidal cell synchrony in vivo remains elusive. Employing an all-optical approach in mice, we simultaneously recorded hippocampal interneurons and pyramidal cells and probed the network influence of individual interneurons using optogenetics. We demonstrate that CA1 interneurons form a functionally interconnected network that promotes synchrony through disinhibition during awake immobility, while preserving endogenous cell assemblies. Our network model underscores the importance of both cell assemblies and dense, unspecific interneuron connectivity in explaining our experimental findings, suggesting that interneurons may operate not only via division of labor but also through concerted activity.
Collapse
Affiliation(s)
- Marco Bocchio
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
- Department of Psychology, Durham University, Durham, United Kingdom
| | - Artem Vorobyev
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Sadra Sadeh
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Sophie Brustlein
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Robin Dard
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Susanne Reichinnek
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Valentina Emiliani
- Wavefront-Engineering Microscopy Group, Photonics Department, Vision Institute, Sorbonne University, INSERM, CNRS, Paris, France
| | - Agnes Baude
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Claudia Clopath
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Rosa Cossart
- Aix Marseille, University, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| |
Collapse
|
15
|
Zeppillo T, Ali H, Ravichandran S, Ritter TC, Wenger S, López-Murcia FJ, Gideons E, Signorelli J, Schmeisser MJ, Wiltfang J, Rhee J, Brose N, Taschenberger H, Krueger-Burg D. Functional Neuroligin-2-MDGA1 interactions differentially regulate synaptic GABA ARs and cytosolic gephyrin aggregation. Commun Biol 2024; 7:1157. [PMID: 39284869 PMCID: PMC11405390 DOI: 10.1038/s42003-024-06789-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Neuroligin-2 (Nlgn2) is a key synaptic adhesion protein at virtually all GABAergic synapses, which recruits GABAARs by promoting assembly of the postsynaptic gephyrin scaffold. Intriguingly, loss of Nlgn2 differentially affects subsets of GABAergic synapses, indicating that synapse-specific interactors and redundancies define its function, but the nature of these interactions remain poorly understood. Here we investigated how Nlgn2 function in hippocampal area CA1 is modulated by two proposed interaction partners, MDGA1 and MDGA2. We show that loss of MDGA1 expression, but not heterozygous deletion of MDGA2, ameliorates the abnormal cytosolic gephyrin aggregation, the reduction in inhibitory synaptic transmission and the exacerbated anxiety-related behaviour characterizing Nlgn2 knockout (KO) mice. Additionally, combined Nlgn2 and MDGA1 deletion causes an exacerbated layer-specific loss of gephyrin puncta. Given that both Nlgn2 and the MDGA1 have been correlated with many psychiatric disorders, our data support the notion that cytosolic gephyrin aggregation may represent an interesting target for novel therapeutic strategies.
Collapse
Affiliation(s)
- Tommaso Zeppillo
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Heba Ali
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences (GGNB), Georg-August-University Göttingen, 37077, Göttingen, Germany
| | - Sowbarnika Ravichandran
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Tamara C Ritter
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Sally Wenger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Francisco J López-Murcia
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, and Bellvitge Biomedical Research Institute (IDIBELL), 08907, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Erinn Gideons
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Janetti Signorelli
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
- Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, 1240000, Antofagasta, Chile
| | - Michael J Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center of the Georg-August-University Göttingen Mainz, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany
| | - Dilja Krueger-Burg
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, 37075, Göttingen, Germany.
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany.
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University Mainz, 55128, Mainz, Germany.
- Department of Psychiatry and Psychotherapy, University Medical Center of the Georg-August-University Göttingen Mainz, 37075, Göttingen, Germany.
| |
Collapse
|
16
|
Wang H, Singh S, Trappenberg T, Nunes A. An Information-Geometric Formulation of Pattern Separation and Evaluation of Existing Indices. ENTROPY (BASEL, SWITZERLAND) 2024; 26:737. [PMID: 39330071 PMCID: PMC11431106 DOI: 10.3390/e26090737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024]
Abstract
Pattern separation is a computational process by which dissimilar neural patterns are generated from similar input patterns. We present an information-geometric formulation of pattern separation, where a pattern separator is modeled as a family of statistical distributions on a manifold. Such a manifold maps an input (i.e., coordinates) to a probability distribution that generates firing patterns. Pattern separation occurs when small coordinate changes result in large distances between samples from the corresponding distributions. Under this formulation, we implement a two-neuron system whose probability law forms a three-dimensional manifold with mutually orthogonal coordinates representing the neurons' marginal and correlational firing rates. We use this highly controlled system to examine the behavior of spike train similarity indices commonly used in pattern separation research. We find that all indices (except scaling factor) are sensitive to relative differences in marginal firing rates, but no index adequately captures differences in spike trains that result from altering the correlation in activity between the two neurons. That is, existing pattern separation metrics appear (A) sensitive to patterns that are encoded by different neurons but (B) insensitive to patterns that differ only in relative spike timing (e.g., synchrony between neurons in the ensemble).
Collapse
Affiliation(s)
- Harvey Wang
- Faculty of Computer Science, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Selena Singh
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Thomas Trappenberg
- Faculty of Computer Science, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Abraham Nunes
- Faculty of Computer Science, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Psychiatry, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
17
|
Kim SY, Lim W. Effect of adult-born immature granule cells on pattern separation in the hippocampal dentate gyrus. Cogn Neurodyn 2024; 18:2077-2093. [PMID: 39104672 PMCID: PMC11297892 DOI: 10.1007/s11571-023-09985-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 08/07/2024] Open
Abstract
Young immature granule cells (imGCs) appear via adult neurogenesis in the hippocampal dentate gyrus (DG). In comparison to mature GCs (mGCs) (born during development), the imGCs exhibit two competing distinct properties such as high excitability (increasing activation degree) and low excitatory innervation (reducing activation degree). We develop a spiking neural network for the DG, incorporating both the mGCs and the imGCs. The mGCs are well known to perform "pattern separation" (i.e., a process of transforming similar input patterns into less similar output patterns) to facilitate pattern storage in the hippocampal CA3. In this paper, we investigate the effect of the young imGCs on pattern separation of the mGCs. The pattern separation efficacy (PSE) of the mGCs is found to vary through competition between high excitability and low excitatory innervation of the imGCs. Their PSE becomes enhanced (worsened) when the effect of high excitability is higher (lower) than the effect of low excitatory innervation. In contrast to the mGCs, the imGCs are found to perform "pattern integration" (i.e., making association between dissimilar patterns). Finally, we speculate that memory resolution in the hippocampal CA3 might be optimally maximized via mixed cooperative encoding through pattern separation and pattern integration.
Collapse
Affiliation(s)
- Sang-Yoon Kim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| | - Woochang Lim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| |
Collapse
|
18
|
Ge J, Xie S, Duan J, Tian B, Ren P, Hu E, Huang Q, Mao H, Zou Y, Chen Q, Wang W. Imbalance between hippocampal projection cell and parvalbumin interneuron architecture increases epileptic susceptibility in mouse model of methyl CpG binding protein 2 duplication syndrome. Epilepsia 2024; 65:2483-2496. [PMID: 38819633 DOI: 10.1111/epi.18027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE Methyl CpG-binding protein 2 (MECP2) duplication syndrome is a rare X-linked genomic disorder affecting predominantly males, which is usually manifested as epilepsy and autism spectrum disorder (ASD) comorbidity. The transgenic line MeCP2Tg1 was used for mimicking MECP2 duplication syndrome and showed autism-epilepsy co-occurrence. Previous works suggested that the excitatory/inhibitory (E/I) imbalance is a potential common mechanism for both epilepsy and ASD. The projection neurons and parvalbumin (PV) interneurons account for the majority of E/I balance in the hippocampus. Therefore, we explored how structural changes of projection and PV+ neurons occur in the hippocampus of MeCP2Tg1 mice and whether these morphological changes contribute to epilepsy susceptibility. METHODS We used the interneuron Designer receptors exclusively activated by designer drugs mouse model to inhibit inhibitory neurons in the hippocampus to verify the epilepsy susceptibility of MeCP2Tg1 (FVB, an inbred strain named as sensitivity to Friend leukemia virus) mice. Electroencephalograms were recorded for the definition of seizure. We performed retro-orbital injection of virus in MeCP2Tg1 (FVB):CaMKIIα-Cre (C57BL/6) mice or MeCP2Tg1:PV-Cre (C57BL/6) mice and their littermate controls to specifically label projection and PV+ neurons for structural analysis. RESULTS Epilepsy susceptibility was increased in MeCP2Tg1 mice. There was a reduced number of PV neurons and reduced dendritic complexity in the hippocampus of MeCP2Tg1 mice. The dendritic complexity in MeCP2Tg1 mice was increased compared to wild-type mice, and total dendritic spine density in dentate gyrus of MeCP2Tg1 mice was also increased. Total dendritic spine density was increased in CA1 of MeCP2Tg1 mice. SIGNIFICANCE Overexpression of MeCP2 may disrupt crucial signaling pathways, resulting in decreased dendritic complexity of PV interneurons and increased dendritic spine density of projection neurons. This reciprocal modulation of excitatory and inhibitory neuronal structures associated with MeCP2 implies its significance as a potential target in the development of epilepsy and offers a novel perspective on the co-occurrence of autism and epilepsy.
Collapse
Affiliation(s)
- Junye Ge
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shengjun Xie
- Jingzhou Hospital affiliated with Yangtze University, Jingzhou, China
| | - Jiamei Duan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Biqing Tian
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Pengfei Ren
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Erling Hu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Qiyi Huang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Honghui Mao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yuxin Zou
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Qian Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
19
|
Cui K, Qi X, Liu Z, Sun W, Jiao P, Liu C, Tong J, Sun X, Sun H, Fu S, Wang J, Zheng Y, Liu T, Cui S, Liu F, Mao J, Zheng J, Wan Y, Yi M. Dominant activities of fear engram cells in the dorsal dentate gyrus underlie fear generalization in mice. PLoS Biol 2024; 22:e3002679. [PMID: 38995985 PMCID: PMC11244812 DOI: 10.1371/journal.pbio.3002679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 05/16/2024] [Indexed: 07/14/2024] Open
Abstract
Over-generalized fear is a maladaptive response to harmless stimuli or situations characteristic of posttraumatic stress disorder (PTSD) and other anxiety disorders. The dorsal dentate gyrus (dDG) contains engram cells that play a crucial role in accurate memory retrieval. However, the coordination mechanism of neuronal subpopulations within the dDG network during fear generalization is not well understood. Here, with the Tet-off system combined with immunostaining and two-photon calcium imaging, we report that dDG fear engram cells labeled in the conditioned context constitutes a significantly higher proportion of dDG neurons activated in a similar context where mice show generalized fear. The activation of these dDG fear engram cells encoding the conditioned context is both sufficient and necessary for inducing fear generalization in the similar context. Activities of mossy cells in the ventral dentate gyrus (vMCs) are significantly suppressed in mice showing fear generalization in a similar context, and activating the vMCs-dDG pathway suppresses generalized but not conditioned fear. Finally, modifying fear memory engrams in the dDG with "safety" signals effectively rescues fear generalization. These findings reveal that the competitive advantage of dDG engram cells underlies fear generalization, which can be rescued by activating the vMCs-dDG pathway or modifying fear memory engrams, and provide novel insights into the dDG network as the neuronal basis of fear generalization.
Collapse
Affiliation(s)
- Kun Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
| | - Xuetao Qi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zilong Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Weiqi Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Peijie Jiao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chang Liu
- Beijing Life Science Academy, Beijing, China
| | - Jifu Tong
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaoyan Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Haojie Sun
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- UCL School of Pharmacy, University College London, London, United Kingdom
| | - Su Fu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaxin Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yawen Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tianyu Liu
- Department of Anesthesiology, Peking University People's Hospital, Beijing, China
| | - Shuang Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Jian Mao
- Beijing Life Science Academy, Beijing, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ming Yi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| |
Collapse
|
20
|
Li H, Lai L, Li X, Wang R, Fang X, Xu N, Zhao J. Electroacupuncture Ameliorates Cognitive Impairment by Regulating γ-Amino Butyric Acidergic Interneurons in the Hippocampus of 5 Familial Alzheimer's Disease Mice. Neuromodulation 2024; 27:730-741. [PMID: 36604241 DOI: 10.1016/j.neurom.2022.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVES γ-amino butyric acid (GABA)-ergic dysfunction in excitatory and inhibitory (E/I) imbalance drives the pathogenesis of Alzheimer's disease (AD). Inhibitory interneurons play an important role in the regulation of E/I balance, synaptic transmission, and network oscillation through manipulation of GABAergic functions, showing positive outcomes in AD animal models. Mice expressing 5 familial AD mutation (5xFAD) exhibited a series of AD-like pathology and learning and memory deficits with age. Because electroacupuncture (EA) treatment has been used for a complementary alternative medicine therapy in patients with AD, we aimed to examine any usefulness of EA therapy in GABA interneuron function and its associated synaptic proteins, to determine whether EA could effectively improve inhibitory transmission and network oscillation and eventually alleviate cognitive impairments in 5xFAD mice, and to further elucidate the GABAergic system function underlying the antidementia response of EA. MATERIALS AND METHODS 5xFAD mice were used to evaluate the potential neuroprotective effect of electroacupuncture at Baihui (DU 20) and Dazhui (DU 14) through behavioral testing, immunofluorescence staining, electrophysiology recording, and molecular biology analysis. RESULTS First, we observed that EA improved memory deficits and inhibitory synaptic protein expression. Second, EA treatment alleviated the decrease of somatostatin-positive interneurons in the dorsal hippocampus. Third, EA attenuated E/I imbalance in 5xFAD mice. Last, EA treatment enhanced theta and gamma oscillation in the hippocampus of 5xFAD mice. CONCLUSIONS EA stimulation at DU20 and DU14 acupoints may be a potential alternative therapy to ameliorate cognitive deficits in AD through the regulation of the function of the GABAergic interneuron.
Collapse
Affiliation(s)
- Hongzhu Li
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rehabilitation, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lanfeng Lai
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Li
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runyi Wang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoling Fang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaying Zhao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
21
|
Nakajima N, Kamijo T, Hayakawa H, Sugisaki E, Aihara T. Modification of temporal pattern sensitivity for inputs from medial entorhinal cortex by lateral inputs in hippocampal granule cells. Cogn Neurodyn 2024; 18:1047-1059. [PMID: 38826655 PMCID: PMC11143144 DOI: 10.1007/s11571-023-09964-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/08/2023] [Accepted: 03/23/2023] [Indexed: 06/04/2024] Open
Abstract
The medial dendrites (MDs) of granule cells (GCs) receive spatial information through the medial entorhinal cortex (MEC) from the entorhinal cortex in the rat hippocampus while the distal dendrites (DDs) of GCs receive non-spatial information (sensory inputs) through the lateral entorhinal cortex (LEC). However, it is unclear how information processing through the two pathways is managed in GCs. In this study, we investigated associative information processing between two independent inputs to MDs and DDs. First, in physiological experiments, to compare response characteristics between MDs and DDs, electrical stimuli comprising five pulses were applied to the MPP or LPP in rat hippocampal slices. These stimuli transiently decreased the excitatory postsynaptic potentials (EPSPs) of successive input pulses to MDs, whereas EPSPs to DDs showed sustained responses. Next, in computational experiments using a local network model obtained by fitting of the physiological experimental data, we compared associative information processing between DDs and MDs. The results showed that the temporal pattern sensitivity for burst inputs to MDs depended on the frequency of the random pulse inputs applied to DDs. On the other hand, with lateral inhibition to GCs from interneurons, the temporal pattern sensitivity for burst inputs to MDs was enhanced or tuned up according to the frequency of the random pulse inputs to the other cells. Thus, our results suggest that the temporal pattern sensitivity of spatial information depends on the non-spatial inputs to GCs.
Collapse
Affiliation(s)
- Naoki Nakajima
- Graduated School of Engineering, Tamagawa University, Tokyo, Japan
| | - Tadanobu Kamijo
- Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | | | - Eriko Sugisaki
- Brain Science Institute, Tamagawa University, Tokyo, Japan
| | - Takeshi Aihara
- Graduated School of Engineering, Tamagawa University, Tokyo, Japan
- Brain Science Institute, Tamagawa University, Tokyo, Japan
| |
Collapse
|
22
|
Arutiunian V, Santhosh M, Neuhaus E, Borland H, Tompkins C, Bernier RA, Bookheimer SY, Dapretto M, Gupta AR, Jack A, Jeste S, McPartland JC, Naples A, Van Horn JD, Pelphrey KA, Webb SJ. The relationship between gamma-band neural oscillations and language skills in youth with Autism Spectrum Disorder and their first-degree relatives. Mol Autism 2024; 15:19. [PMID: 38711098 PMCID: PMC11075235 DOI: 10.1186/s13229-024-00598-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Most children with Autism Spectrum Disorder (ASD) have co-occurring language impairments and some of these autism-specific language difficulties are also present in their non-autistic first-degree relatives. One of the possible neural mechanisms associated with variability in language functioning is alterations in cortical gamma-band oscillations, hypothesized to be related to neural excitation and inhibition balance. METHODS We used a high-density 128-channel electroencephalography (EEG) to register brain response to speech stimuli in a large sex-balanced sample of participants: 125 youth with ASD, 121 typically developing (TD) youth, and 40 unaffected siblings (US) of youth with ASD. Language skills were assessed with Clinical Evaluation of Language Fundamentals. RESULTS First, during speech processing, we identified significantly elevated gamma power in ASD participants compared to TD controls. Second, across all youth, higher gamma power was associated with lower language skills. Finally, the US group demonstrated an intermediate profile in both language and gamma power, with nonverbal IQ mediating the relationship between gamma power and language skills. LIMITATIONS We only focused on one of the possible neural contributors to variability in language functioning. Also, the US group consisted of a smaller number of participants in comparison to the ASD or TD groups. Finally, due to the timing issue in EEG system we have provided only non-phase-locked analysis. CONCLUSIONS Autistic youth showed elevated gamma power, suggesting higher excitation in the brain in response to speech stimuli and elevated gamma power was related to lower language skills. The US group showed an intermediate pattern of gamma activity, suggesting that the broader autism phenotype extends to neural profiles.
Collapse
Affiliation(s)
- Vardan Arutiunian
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Megha Santhosh
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Emily Neuhaus
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Heather Borland
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Chris Tompkins
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
| | - Susan Y Bookheimer
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Mirella Dapretto
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Abha R Gupta
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Allison Jack
- Department of Psychology, George Mason University, Fairfax, VA, USA
| | - Shafali Jeste
- Department of Neurology, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | | | - Adam Naples
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - John D Van Horn
- School of Data Science, University of Virginia, Charlottesville, VA, USA
| | - Kevin A Pelphrey
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Sara Jane Webb
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA.
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA.
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA.
| |
Collapse
|
23
|
Shu WC, Jackson MB. Intrinsic and Synaptic Contributions to Repetitive Spiking in Dentate Granule Cells. J Neurosci 2024; 44:e0716232024. [PMID: 38503495 PMCID: PMC11063872 DOI: 10.1523/jneurosci.0716-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 03/21/2024] Open
Abstract
Repetitive firing of granule cells (GCs) in the dentate gyrus (DG) facilitates synaptic transmission to the CA3 region. This facilitation can gate and amplify the flow of information through the hippocampus. High-frequency bursts in the DG are linked to behavior and plasticity, but GCs do not readily burst. Under normal conditions, a single shock to the perforant path in a hippocampal slice typically drives a GC to fire a single spike, and only occasionally more than one spike is seen. Repetitive spiking in GCs is not robust, and the mechanisms are poorly understood. Here, we used a hybrid genetically encoded voltage sensor to image voltage changes evoked by cortical inputs in many mature GCs simultaneously in hippocampal slices from male and female mice. This enabled us to study relatively infrequent double and triple spikes. We found GCs are relatively homogeneous and their double spiking behavior is cell autonomous. Blockade of GABA type A receptors increased multiple spikes and prolonged the interspike interval, indicating inhibitory interneurons limit repetitive spiking and set the time window for successive spikes. Inhibiting synaptic glutamate release showed that recurrent excitation mediated by hilar mossy cells contributes to, but is not necessary for, multiple spiking. Blockade of T-type Ca2+ channels did not reduce multiple spiking but prolonged interspike intervals. Imaging voltage changes in different GC compartments revealed that second spikes can be initiated in either dendrites or somata. Thus, pharmacological and biophysical experiments reveal roles for both synaptic circuitry and intrinsic excitability in GC repetitive spiking.
Collapse
Affiliation(s)
- Wen-Chi Shu
- Department of Neuroscience and Biophysics Program, University of Wisconsin-Madison, Wisconsin 53705
| | - Meyer B Jackson
- Department of Neuroscience and Biophysics Program, University of Wisconsin-Madison, Wisconsin 53705
| |
Collapse
|
24
|
Jeong M, Jang JH, Oh SJ, Park J, Lee J, Hwang S, Oh YS. Maladaptation of dentate gyrus mossy cells mediates contextual discrimination deficit after traumatic stress. Cell Rep 2024; 43:114000. [PMID: 38527063 DOI: 10.1016/j.celrep.2024.114000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 03/27/2024] Open
Abstract
Fear overgeneralization is a maladaptive response to traumatic stress that is associated with the inability to discriminate between threat and safety contexts, a hallmark feature of post-traumatic stress disorder (PTSD). However, the neural mechanisms underlying this deficit remain unclear. Here, we show that traumatic stress exposure impairs contextual discrimination between threat and safety contexts in the learned helplessness (LH) model. Mossy cells (MCs) in the dorsal hippocampus are suppressed in response to traumatic stress. Bidirectional manipulation of MC activity in the LH model reveals that MC inhibition is causally linked to impaired contextual discrimination. Mechanistically, MC inhibition increases the number of active granule cells in a given context, significantly overlapping context-specific ensembles. Our study demonstrates that maladaptive inhibition of MCs after traumatic stress is a substantial mechanism underlying fear overgeneralization with contextual discrimination deficit, suggesting a potential therapeutic target for cognitive symptoms of PTSD.
Collapse
Affiliation(s)
- Minseok Jeong
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Jin-Hyeok Jang
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Seo-Jin Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Jeongrak Park
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Junseop Lee
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Sehyeon Hwang
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Yong-Seok Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea; Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, 61 Cheomdan-ro, Daegu 41062, Republic of Korea.
| |
Collapse
|
25
|
Lee SH, Kang YJ, Smith BN. Activation of hypoactive parvalbumin-positive fast-spiking interneuron restores dentate inhibition to prevent epileptiform activity in the mouse intrahippocampal kainate model of temporal lobe epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588316. [PMID: 38645248 PMCID: PMC11030452 DOI: 10.1101/2024.04.05.588316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Parvalbumin-positive (PV+) GABAergic interneurons in the dentate gyrus provide powerful perisomatic inhibition of dentate granule cells (DGCs) to prevent overexcitation and maintain the stability of dentate gyrus circuits. Most dentate PV+ interneurons survive status epilepticus, but surviving PV+ interneuron mediated inhibition is compromised in the dentate gyrus shortly after status epilepticus, contributing to epileptogenesis in temporal lobe epilepsy. It is uncertain whether the impaired activity of dentate PV+ interneurons recovers at later times or if it continues for months following status epilepticus. The development of compensatory modifications related to PV+ interneuron circuits in the months following status epilepticus is unknown, although reduced dentate GABAergic inhibition persists long after status epilepticus. We employed PV immunostaining and whole-cell patch-clamp recordings from dentate PV+ interneurons and DGCs in slices from male and female sham controls and intrahippocampal kainate (IHK) treated mice that developed spontaneous seizures months after status epilepticus to study epilepsy-associated changes in dentate PV+ interneuron circuits. We found that the number of dentate PV+ cells was reduced in IHK treated mice. Electrical recordings showed that: 1) Action potential firing rates of dentate PV+ interneurons were reduced in IHK treated mice up to four months after status epilepticus; 2) Spontaneous inhibitory postsynaptic currents (sIPSCs) in DGCs exhibited reduced frequency but increased amplitude in IHK treated mice; and 3) The amplitude of evoked IPSCs in DGCs by optogenetic activation of dentate PV+ cells was upregulated without changes in short-term plasticity. Video-EEG recordings revealed that IHK treated mice showed spontaneous epileptiform activity in the dentate gyrus and that chemogenetic activation of PV+ interneurons abolished the epileptiform activity. Our results suggest not only that the compensatory changes in PV+ interneuron circuits develop after IHK treatment, but also that increased PV+ interneuron mediated inhibition in the dentate gyrus may compensate for cell loss and reduced intrinsic excitability of dentate PV+ interneurons to stop seizures in temporal lobe epilepsy. Highlights Reduced number of dentate PV+ interneurons in TLE micePersistently reduced action potential firing rates of dentate PV+ interneurons in TLE miceEnhanced amplitude but decreased frequency of spontaneous IPSCs in the dentate gyrus in TLE miceIncreased amplitude of evoked IPSCs mediated by dentate PV+ interneurons in TLE miceChemogenetic activation of PV+ interneurons prevents epileptiform activity in TLE mice.
Collapse
|
26
|
Tomé DF, Zhang Y, Aida T, Mosto O, Lu Y, Chen M, Sadeh S, Roy DS, Clopath C. Dynamic and selective engrams emerge with memory consolidation. Nat Neurosci 2024; 27:561-572. [PMID: 38243089 PMCID: PMC10917686 DOI: 10.1038/s41593-023-01551-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/12/2023] [Indexed: 01/21/2024]
Abstract
Episodic memories are encoded by experience-activated neuronal ensembles that remain necessary and sufficient for recall. However, the temporal evolution of memory engrams after initial encoding is unclear. In this study, we employed computational and experimental approaches to examine how the neural composition and selectivity of engrams change with memory consolidation. Our spiking neural network model yielded testable predictions: memories transition from unselective to selective as neurons drop out of and drop into engrams; inhibitory activity during recall is essential for memory selectivity; and inhibitory synaptic plasticity during memory consolidation is critical for engrams to become selective. Using activity-dependent labeling, longitudinal calcium imaging and a combination of optogenetic and chemogenetic manipulations in mouse dentate gyrus, we conducted contextual fear conditioning experiments that supported our model's predictions. Our results reveal that memory engrams are dynamic and that changes in engram composition mediated by inhibitory plasticity are crucial for the emergence of memory selectivity.
Collapse
Affiliation(s)
- Douglas Feitosa Tomé
- Department of Bioengineering, Imperial College London, London, UK.
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | - Ying Zhang
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Center for Life Sciences & IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Tomomi Aida
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Olivia Mosto
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yifeng Lu
- Center for Life Sciences & IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Mandy Chen
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sadra Sadeh
- Department of Bioengineering, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Dheeraj S Roy
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Claudia Clopath
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
27
|
Valeri J, Stiplosek C, O'Donovan SM, Sinclair D, Grant KA, Bollavarapu R, Platt DM, Stockmeier CA, Gisabella B, Pantazopoulos H. Extracellular matrix abnormalities in the hippocampus of subjects with substance use disorder. Transl Psychiatry 2024; 14:115. [PMID: 38402197 PMCID: PMC10894211 DOI: 10.1038/s41398-024-02833-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/26/2024] Open
Abstract
Contextual triggers are significant factors contributing to relapse in substance use disorders (SUD). Emerging evidence points to a critical role of extracellular matrix (ECM) molecules as mediators of reward memories. Chondroitin sulfate proteoglycans (CSPGs) are a subset of ECM molecules that form perineuronal nets (PNN) around inhibitory neurons. PNNs restrict synaptic connections and help maintain synapses. Rodent models suggest that modulation of PNNs may strengthen contextual reward memories in SUD. However, there is currently a lack of information regarding PNNs in the hippocampus of people with SUD as well as how comorbidity with major depressive disorder (MDD) may affect PNNs. We used postmortem hippocampal tissues from cohorts of human and nonhuman primates with or without chronic alcohol use to test the hypothesis that PNNs are increased in subjects with SUD. We used histochemical labeling and quantitative microscopy to examine PNNs, and qRT-PCR to examine gene expression for ECM molecules, synaptic markers and related markers. We identified increased densities of PNNs and CSPG-labeled glial cells in SUD, coinciding with decreased expression of the ECM protease matrix metalloproteinase 9 (Mmp9), and increased expression for the excitatory synaptic marker vesicle associated membrane protein 2 (Vamp2). Similar increases in PNNs were observed in monkeys with chronic alcohol self-administration. Subjects with MDD displayed changes opposite to SUD, and subjects with SUD and comorbid MDD had minimal changes in any of the outcome measures examined. Our findings demonstrate that PNNs are increased in SUD, possibly contributing to stabilizing contextual reward memories as suggested by preclinical studies. Our results also point to a previously unsuspected role for CSPG expression in glial cells in SUD. Evidence for increased hippocampal PNNs in SUD suggests that targeting PNNs to weaken contextual reward memories is a promising therapeutic approach for SUD, however comorbidity with MDD is a significant consideration.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charlotte Stiplosek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - David Sinclair
- Department of Neuroscience, University of Toledo, Toledo, OH, USA
| | | | - Ratna Bollavarapu
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Donna M Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA.
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
28
|
Li H, Tamura R, Hayashi D, Asai H, Koga J, Ando S, Yokota S, Kaneko J, Sakurai K, Sumiyoshi A, Yamamoto T, Hikishima K, Tanaka KZ, McHugh TJ, Hisatsune T. Silencing dentate newborn neurons alters excitatory/inhibitory balance and impairs behavioral inhibition and flexibility. SCIENCE ADVANCES 2024; 10:eadk4741. [PMID: 38198539 PMCID: PMC10780870 DOI: 10.1126/sciadv.adk4741] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
Adult neurogenesis confers the hippocampus with unparalleled neural plasticity, essential for intricate cognitive functions. The specific influence of sparse newborn neurons (NBNs) in modulating neural activities and subsequently steering behavior, however, remains obscure. Using an engineered NBN-tetanus toxin mouse model (NBN-TeTX), we noninvasively silenced NBNs, elucidating their crucial role in impulse inhibition and cognitive flexibility as evidenced through Morris water maze reversal learning and Go/Nogo task in operant learning. Task-based functional MRI (tb-fMRI) paired with operant learning revealed dorsal hippocampal hyperactivation during the Nogo task in male NBN-TeTX mice, suggesting that hippocampal hyperexcitability might underlie the observed behavioral deficits. Additionally, resting-state fMRI (rs-fMRI) exhibited enhanced functional connectivity between the dorsal and ventral dentate gyrus following NBN silencing. Further investigations into the activities of PV+ interneurons and mossy cells highlighted the indispensability of NBNs in maintaining the hippocampal excitation/inhibition balance. Our findings emphasize that the neural plasticity driven by NBNs extensively modulates the hippocampus, sculpting inhibitory control and cognitive flexibility.
Collapse
Affiliation(s)
- Haowei Li
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Risako Tamura
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Daiki Hayashi
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Hirotaka Asai
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Junya Koga
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Shota Ando
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Sayumi Yokota
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Jun Kaneko
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Keisuke Sakurai
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Akira Sumiyoshi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tadashi Yamamoto
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Keigo Hikishima
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kazumasa Z. Tanaka
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Thomas J. McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Tatsuhiro Hisatsune
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
29
|
Valeri J, Stiplosek C, O’Donovan SM, Sinclair D, Grant K, Bollavarapu R, Platt DM, Stockmeier CA, Gisabella B, Pantazopoulos H. Extracellular Matrix Abnormalities in the Hippocampus of Subjects with Substance Use Disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.09.07.23295222. [PMID: 37732207 PMCID: PMC10508799 DOI: 10.1101/2023.09.07.23295222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Contextual triggers are significant factors contributing to relapse in substance use disorders (SUD). Emerging evidence points to a critical role of extracellular matrix (ECM) molecules as mediators of reward memories. Chondroitin sulfate proteoglycans (CSPGs) are a subset of ECM molecules that form perineuronal nets (PNN) around inhibitory neurons. PNNs restrict synaptic connections and help maintain synapses. Rodent models suggest that modulation of PNNs may strengthen contextual reward memories in SUD. However, there is currently a lack of information regarding PNNs in the hippocampus of people with SUD as well as how comorbidity with major depressive disorder (MDD) may affect PNNs. We used postmortem hippocampal tissues from cohorts of human and nonhuman primates with or without chronic alcohol use to test the hypothesis that PNNs are increased in subjects with SUD. We used histochemical labeling and quantitative microscopy to examine PNNs, and qRT-PCR to examine gene expression for ECM molecules, synaptic markers and related markers. We identified increased densities of PNNs and CSPG-labeled glial cells in SUD, coinciding with decreased expression of the ECM protease matrix metalloproteinase 9 (Mmp9), and increased expression for the excitatory synaptic marker vesicle associated membrane protein 2 (Vamp2). Similar increases in PNNs were observed in monkeys with chronic alcohol self-administration. Subjects with MDD displayed changes opposite to SUD, and subjects with SUD and comorbid MDD had minimal changes in any of the outcome measures examined. Our findings demonstrate that PNNs are increased in SUD, possibly contributing to stabilizing contextual reward memories as suggested by preclinical studies. Our results also point to a previously unsuspected role for CSPG expression in glial cells in SUD. Evidence for increased hippocampal PNNs in SUD suggests that targeting PNNs to weaken contextual reward memories is a promising therapeutic approach for SUD, however comorbidity with MDD is a significant consideration.
Collapse
Affiliation(s)
- Jake Valeri
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Charlotte Stiplosek
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
| | | | - David Sinclair
- Department of Neuroscience, University of Toledo, Toledo, OH
| | | | - Ratna Bollavarapu
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Donna M. Platt
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
30
|
Singh S, Becker S, Trappenberg T, Nunes A. Granule cells perform frequency-dependent pattern separation in a computational model of the dentate gyrus. Hippocampus 2024; 34:14-28. [PMID: 37950569 DOI: 10.1002/hipo.23585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/28/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
Mnemonic discrimination (MD) may be dependent on oscillatory perforant path input frequencies to the hippocampus in a "U"-shaped fashion, where some studies show that slow and fast input frequencies support MD, while other studies show that intermediate frequencies disrupt MD. We hypothesize that pattern separation (PS) underlies frequency-dependent MD performance. We aim to study, in a computational model of the hippocampal dentate gyrus (DG), the network and cellular mechanisms governing this putative "U"-shaped PS relationship. We implemented a biophysical model of the DG that produces the hypothesized "U"-shaped input frequency-PS relationship, and its associated oscillatory electrophysiological signatures. We subsequently evaluated the network's PS ability using an adapted spatiotemporal task. We undertook systematic lesion studies to identify the network-level mechanisms driving the "U"-shaped input frequency-PS relationship. A minimal circuit of a single granule cell (GC) stimulated with oscillatory inputs was also used to study potential cellular-level mechanisms. Lesioning synapses onto GCs did not impact the "U"-shaped input frequency-PS relationship. Furthermore, GC inhibition limits PS performance for fast frequency inputs, while enhancing PS for slow frequency inputs. GC interspike interval was found to be input frequency dependent in a "U"-shaped fashion, paralleling frequency-dependent PS observed at the network level. Additionally, GCs showed an attenuated firing response for fast frequency inputs. We conclude that independent of network-level inhibition, GCs may intrinsically be capable of producing a "U"-shaped input frequency-PS relationship. GCs may preferentially decorrelate slow and fast inputs via spike timing reorganization and high frequency filtering.
Collapse
Affiliation(s)
- Selena Singh
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Suzanna Becker
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, Ontario, Canada
| | - Thomas Trappenberg
- Faculty of Computer Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Abraham Nunes
- Faculty of Computer Science, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
31
|
Fincham GW, Kartar A, Uthaug MV, Anderson B, Hall L, Nagai Y, Critchley H, Colasanti A. High ventilation breathwork practices: An overview of their effects, mechanisms, and considerations for clinical applications. Neurosci Biobehav Rev 2023; 155:105453. [PMID: 37923236 DOI: 10.1016/j.neubiorev.2023.105453] [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: 08/01/2023] [Revised: 10/19/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
High Ventilation Breathwork (HVB) refers to practices employing specific volitional manipulation of breathing, with a long history of use to relieve various forms of psychological distress. This paper seeks to offer a consolidative insight into potential clinical application of HVB as a treatment of psychiatric disorders. We thus review the characteristic phenomenological and neurophysiological effects of these practices to inform their mechanism of therapeutic action, safety profiles and future clinical applications. Clinical observations and data from neurophysiological studies indicate that HVB is associated with extraordinary changes in subjective experience, as well as with profound effects on central and autonomic nervous systems functions through modulation of neurometabolic parameters and interoceptive sensory systems. This growing evidence base may guide how the phenomenological effects of HVB can be understood, and potentially harnessed in the context of such volitional perturbation of psychophysiological state. Reports of putative beneficial effects for trauma-related, affective, and somatic disorders invite further research to obtain detailed mechanistic knowledge, and rigorous clinical testing of these potential therapeutic uses.
Collapse
Affiliation(s)
- Guy W Fincham
- Brighton & Sussex Medical School, Department of Neuroscience, University of Sussex, UK; University of Sussex, School of Psychology, Brighton, UK.
| | - Amy Kartar
- Brighton & Sussex Medical School, Department of Neuroscience, University of Sussex, UK
| | - Malin V Uthaug
- The Centre for Psychedelic Research, Division of Psychiatry, Imperial College London, UK; Department of Neuropsychology & Psychopharmacology, Faculty of Psychology & Neuroscience, Maastricht University, The Netherlands
| | - Brittany Anderson
- University of Wisconsin School of Medicine & Public Health, Department of Psychiatry, University of Wisconsin-Madison, USA
| | - Lottie Hall
- Brighton & Sussex Medical School, Department of Neuroscience, University of Sussex, UK
| | - Yoko Nagai
- Brighton & Sussex Medical School, Department of Neuroscience, University of Sussex, UK
| | - Hugo Critchley
- Brighton & Sussex Medical School, Department of Neuroscience, University of Sussex, UK
| | - Alessandro Colasanti
- Brighton & Sussex Medical School, Department of Neuroscience, University of Sussex, UK; Sussex Partnership NHS Foundation Trust.
| |
Collapse
|
32
|
Müller-Komorowska D, Kuru B, Beck H, Braganza O. Phase information is conserved in sparse, synchronous population-rate-codes via phase-to-rate recoding. Nat Commun 2023; 14:6106. [PMID: 37777512 PMCID: PMC10543394 DOI: 10.1038/s41467-023-41803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/19/2023] [Indexed: 10/02/2023] Open
Abstract
Neural computation is often traced in terms of either rate- or phase-codes. However, most circuit operations will simultaneously affect information across both coding schemes. It remains unclear how phase and rate coded information is transmitted, in the face of continuous modification at consecutive processing stages. Here, we study this question in the entorhinal cortex (EC)- dentate gyrus (DG)- CA3 system using three distinct computational models. We demonstrate that DG feedback inhibition leverages EC phase information to improve rate-coding, a computation we term phase-to-rate recoding. Our results suggest that it i) supports the conservation of phase information within sparse rate-codes and ii) enhances the efficiency of plasticity in downstream CA3 via increased synchrony. Given the ubiquity of both phase-coding and feedback circuits, our results raise the question whether phase-to-rate recoding is a recurring computational motif, which supports the generation of sparse, synchronous population-rate-codes in areas beyond the DG.
Collapse
Affiliation(s)
- Daniel Müller-Komorowska
- Neural Coding and Brain Computing Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan.
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany.
| | - Baris Kuru
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
| | - Heinz Beck
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen e.V, Bonn, Germany
| | - Oliver Braganza
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany.
- Institute for Socio-Economics, University of Duisburg-Essen, Duisburg, Germany.
| |
Collapse
|
33
|
Wibawa P, Walterfang M, Malpas CB, Glikmann‐Johnston Y, Poudel G, Razi A, Hannan AJ, Velakoulis D, Georgiou‐Karistianis N. Selective perforant-pathway atrophy in Huntington disease: MRI analysis of hippocampal subfields. Eur J Neurol 2023; 30:2650-2660. [PMID: 37306313 PMCID: PMC10946817 DOI: 10.1111/ene.15918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/17/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION While individuals with Huntington disease (HD) show memory impairment that indicates hippocampal dysfunction, the available literature does not consistently identify structural evidence for involvement of the whole hippocampus but rather suggests that hippocampal atrophy may be confined to certain hippocampal subregions. METHODS We processed T1-weighted MRI from IMAGE-HD study using FreeSurfer 7.0 and compared the volumes of the hippocampal subfields among 36 early motor symptomatic (symp-HD), 40 pre-symptomatic (pre-HD), and 36 healthy control individuals across three timepoints over 36 months. RESULTS Mixed-model analyses revealed significantly lower subfield volumes in symp-HD, compared with pre-HD and control groups, in the subicular regions of the perforant-pathway: presubiculum, subiculum, dentate gyrus, tail, and right molecular layer. These adjoining subfields aggregated into a single principal component, which demonstrated an accelerated rate of atrophy in the symp-HD. Volumes between pre-HD and controls did not show any significant difference. In the combined HD groups, CAG repeat length and disease burden score were associated with presubiculum, molecular layer, tail, and perforant-pathway subfield volumes. Hippocampal left tail and perforant-pathway subfields were associated with motor onset in the pre-HD group. CONCLUSIONS Hippocampal subfields atrophy in early symptomatic HD affects key regions of the perforant-pathway, which may implicate the distinctive memory impairment at this stage of illness. Their volumetric associations with genetic and clinical markers suggest the selective susceptibility of these subfields to mutant Huntingtin and disease progression.
Collapse
Affiliation(s)
- Pierre Wibawa
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Mark Walterfang
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Charles B. Malpas
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
| | - Yifat Glikmann‐Johnston
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Govinda Poudel
- Mary Mackillop Institute for Health ResearchAustralian Catholic UniversityFitzroyVictoriaAustralia
| | - Adeel Razi
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Dennis Velakoulis
- NeuropsychiatryRoyal Melbourne HospitalParkvilleVictoriaAustralia
- Melbourne Neuropsychiatry CenterUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Nellie Georgiou‐Karistianis
- School of Psychological Sciences and Turner Institute for Brain and Mental HealthMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
34
|
Borzello M, Ramirez S, Treves A, Lee I, Scharfman H, Stark C, Knierim JJ, Rangel LM. Assessments of dentate gyrus function: discoveries and debates. Nat Rev Neurosci 2023; 24:502-517. [PMID: 37316588 PMCID: PMC10529488 DOI: 10.1038/s41583-023-00710-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/16/2023]
Abstract
There has been considerable speculation regarding the function of the dentate gyrus (DG) - a subregion of the mammalian hippocampus - in learning and memory. In this Perspective article, we compare leading theories of DG function. We note that these theories all critically rely on the generation of distinct patterns of activity in the region to signal differences between experiences and to reduce interference between memories. However, these theories are divided by the roles they attribute to the DG during learning and recall and by the contributions they ascribe to specific inputs or cell types within the DG. These differences influence the information that the DG is thought to impart to downstream structures. We work towards a holistic view of the role of DG in learning and memory by first developing three critical questions to foster a dialogue between the leading theories. We then evaluate the extent to which previous studies address our questions, highlight remaining areas of conflict, and suggest future experiments to bridge these theories.
Collapse
Affiliation(s)
- Mia Borzello
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | - Steve Ramirez
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | | | - Inah Lee
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, South Korea
| | - Helen Scharfman
- Departments of Child and Adolescent Psychiatry, Neuroscience and Physiology and Psychiatry and the Neuroscience Institute, New York University Langone Health, New York, NY, USA
- The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Craig Stark
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, CA, USA
| | - James J Knierim
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Lara M Rangel
- Department of Cognitive Science, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
35
|
Shi Y, Cui H, Li X, Chen L, Zhang C, Zhao X, Li X, Shao Q, Sun Q, Yan K, Wang G. Laminar and dorsoventral organization of layer 1 interneuronal microcircuitry in superficial layers of the medial entorhinal cortex. Cell Rep 2023; 42:112782. [PMID: 37436894 DOI: 10.1016/j.celrep.2023.112782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/03/2023] [Accepted: 06/24/2023] [Indexed: 07/14/2023] Open
Abstract
Layer 1 (L1) interneurons (INs) participate in various brain functions by gating information flow in the neocortex, but their role in the medial entorhinal cortex (MEC) is still unknown, largely due to scant knowledge of MEC L1 microcircuitry. Using simultaneous triple-octuple whole-cell recordings and morphological reconstructions, we comprehensively depict L1IN networks in the MEC. We identify three morphologically distinct types of L1INs with characteristic electrophysiological properties. We dissect intra- and inter-laminar cell-type-specific microcircuits of L1INs, showing connectivity patterns different from those in the neocortex. Remarkably, motif analysis reveals transitive and clustered features of L1 networks, as well as over-represented trans-laminar motifs. Finally, we demonstrate the dorsoventral gradient of L1IN microcircuits, with dorsal L1 neurogliaform cells receiving fewer intra-laminar inputs but exerting more inhibition on L2 principal neurons. These results thus present a more comprehensive picture of L1IN microcircuitry, which is indispensable for deciphering the function of L1INs in the MEC.
Collapse
Affiliation(s)
- Yuying Shi
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Hui Cui
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xiaoyue Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ligu Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Chen Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xinran Zhao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xiaowan Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Qiming Shao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Qiang Sun
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Kaiyue Yan
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Guangfu Wang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China.
| |
Collapse
|
36
|
Goz RU, Hooks BM. Correlated Somatosensory Input in Parvalbumin/Pyramidal Cells in Mouse Motor Cortex. eNeuro 2023; 10:ENEURO.0488-22.2023. [PMID: 37094939 PMCID: PMC10167893 DOI: 10.1523/eneuro.0488-22.2023] [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: 11/30/2022] [Revised: 04/02/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023] Open
Abstract
In mammalian cortex, feedforward excitatory connections recruit feedforward inhibition. This is often carried by parvalbumin (PV+) interneurons, which may densely connect to local pyramidal (Pyr) neurons. Whether this inhibition affects all local excitatory cells indiscriminately or is targeted to specific subnetworks is unknown. Here, we test how feedforward inhibition is recruited by using two-channel circuit mapping to excite cortical and thalamic inputs to PV+ interneurons and Pyr neurons to mouse primary vibrissal motor cortex (M1). Single Pyr and PV+ neurons receive input from both cortex and thalamus. Connected pairs of PV+ interneurons and excitatory Pyr neurons receive correlated cortical and thalamic inputs. While PV+ interneurons are more likely to form local connections to Pyr neurons, Pyr neurons are much more likely to form reciprocal connections with PV+ interneurons that inhibit them. This suggests that Pyr and PV ensembles may be organized based on their local and long-range connections, an organization that supports the idea of local subnetworks for signal transduction and processing. Excitatory inputs to M1 can thus target inhibitory networks in a specific pattern which permits recruitment of feedforward inhibition to specific subnetworks within the cortical column.
Collapse
Affiliation(s)
- Roman U Goz
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Bryan M Hooks
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| |
Collapse
|
37
|
Geiller T, Priestley JB, Losonczy A. A local circuit-basis for spatial navigation and memory processes in hippocampal area CA1. Curr Opin Neurobiol 2023; 79:102701. [PMID: 36878147 PMCID: PMC10020891 DOI: 10.1016/j.conb.2023.102701] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 03/06/2023]
Abstract
The hippocampus is a multi-stage neural circuit that is critical for memory formation. Its distinct anatomy has long inspired theories that rely on local interactions between neurons within each subregion in order to perform serial operations important for memory encoding and storage. These local computations have received less attention in CA1 area, the primary output node of the hippocampus, where excitatory neurons are thought to be only very sparsely interconnected. However, recent findings have demonstrated the power of local circuitry in CA1, with evidence for strong functional interactions among excitatory neurons, regulation by diverse inhibitory microcircuits, and novel plasticity rules that can profoundly reshape the hippocampal ensemble code. Here we review how these properties expand the dynamical repertoire of CA1 beyond the confines of feedforward processing, and what implications they have for hippocampo-cortical functions in memory formation.
Collapse
Affiliation(s)
- Tristan Geiller
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA; Mortimer B Zuckerman Mind Brain Behavior Institute, New York, NY, 10027, USA. https://twitter.com/tgeiller
| | - James B Priestley
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA; Mortimer B Zuckerman Mind Brain Behavior Institute, New York, NY, 10027, USA; Center for Theoretical Neuroscience, Columbia University, New York, NY, 10027, USA. https://twitter.com/jamespriestley4
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, 10027, USA; Mortimer B Zuckerman Mind Brain Behavior Institute, New York, NY, 10027, USA; Kavli Institute for Brain Science, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
38
|
Arutiunian V, Arcara G, Buyanova I, Davydova E, Pereverzeva D, Sorokin A, Tyushkevich S, Mamokhina U, Danilina K, Dragoy O. Neuromagnetic 40 Hz Auditory Steady-State Response in the left auditory cortex is related to language comprehension in children with Autism Spectrum Disorder. Prog Neuropsychopharmacol Biol Psychiatry 2023; 122:110690. [PMID: 36470421 DOI: 10.1016/j.pnpbp.2022.110690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 11/06/2022] [Accepted: 11/29/2022] [Indexed: 12/08/2022]
Abstract
Language impairment is comorbid in most children with Autism Spectrum Disorder (ASD), but its neural mechanisms are still poorly understood. Some studies hypothesize that the atypical low-level sensory perception in the auditory cortex accounts for the abnormal language development in these children. One of the potential non-invasive measures of such low-level perception can be the cortical gamma-band oscillations registered with magnetoencephalography (MEG), and 40 Hz Auditory Steady-State Response (40 Hz ASSR) is a reliable paradigm for eliciting auditory gamma response. Although there is research in children with and without ASD using 40 Hz ASSR, nothing is known about the relationship between this auditory response in children with ASD and their language abilities measured directly in formal assessment. In the present study, we used MEG and individual brain models to investigate 40 Hz ASSR in primary-school-aged children with and without ASD. It was also used to assess how the strength of the auditory response is related to language abilities of children with ASD, their non-verbal IQ, and social functioning. A total of 40 children were included in the study. The results demonstrated that 40 Hz ASSR was reduced in the right auditory cortex in children with ASD when comparing them to typically developing controls. Importantly, our study provides the first evidence of the association between 40 Hz ASSR in the language-dominant left auditory cortex and language comprehension in children with ASD. This link was domain-specific because the other brain-behavior correlations were non-significant.
Collapse
Affiliation(s)
| | | | - Irina Buyanova
- Center for Language and Brain, HSE University, Moscow, Russia
| | - Elizaveta Davydova
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia; Chair of Differential Psychology and Psychophysiology, Moscow State University of Psychology and Education, Moscow, Russia
| | - Darya Pereverzeva
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Alexander Sorokin
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia; Haskins Laboratories, New Haven, CT, United States of America
| | - Svetlana Tyushkevich
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Uliana Mamokhina
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Kamilla Danilina
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Olga Dragoy
- Center for Language and Brain, HSE University, Moscow, Russia; Institute of Linguistics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
39
|
Wang J, Yang Y, Liu J, Qiu J, Zhang D, Ou M, Kang Y, Zhu T, Zhou C. Loss of sodium leak channel (NALCN) in the ventral dentate gyrus impairs neuronal activity of the glutamatergic neurons for inflammation-induced depression in male mice. Brain Behav Immun 2023; 110:13-29. [PMID: 36796706 DOI: 10.1016/j.bbi.2023.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND The dentate gyrus (DG) has been implicated in the pathophysiology of depression. Many studies have revealed the cellular types, neural circuits, and morphological changes of the DG involved in the development of depression. However, the molecular regulating its intrinsic activity in depression is unknown. METHODS Utilizing the mode of depression induced by lipopolysaccharide (LPS), we investigate the involvement of the sodium leak channel (NALCN) in inflammation-induced depressive-like behaviors of male mice. The expression of NALCN was detected by immunohistochemistry and real-time polymerase chain reaction. DG microinjection of the adeno-associated virus or lentivirus was carried out using a stereotaxic instrument and followed by behavioral tests. Neuronal excitability and NALCN conductance were recorded by whole-cell patch-clamp techniques. RESULTS The expression and function of NALCN were reduced in both the dorsal and ventral DG in LPS-treated mice; whereas, only knocking down NALCN in the ventral pole produced depressive-like behaviors and this effect of NALCN was specific to ventral glutamatergic neurons. The excitability of ventral glutamatergic neurons was impaired by both the knockdown of NALCN and/or the treatment of LPS. Then, the overexpression of NALCN in the ventral glutamatergic neurons decreased the susceptibility of mice to inflammation-induced depression, and the intracranial injection of substance P (non-selective NALCN activator) into the ventral DG rapidly ameliorated inflammation-induced depression-like behaviors in an NALCN-dependent manner. CONCLUSIONS NALCN, which drives the neuronal activity of the ventral DG glutamatergic neurons, uniquely regulates depressive-like behaviors and susceptibility to depression. Therefore, the NALCN of glutamatergic neurons in the ventral DG may present a molecular target for rapid antidepressant drugs.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaoxin Yang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingxuan Qiu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengchan Ou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Kang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
40
|
Sack AS. Adult-Born Granule Cells Contribute to Dentate Gyrus Circuit Reorganization after Traumatic Brain Injury. J Neurosci 2023; 43:879-881. [PMID: 36754637 PMCID: PMC9908312 DOI: 10.1523/jneurosci.1994-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 02/10/2023] Open
Affiliation(s)
- Anne-Sophie Sack
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
41
|
Ash AM, Regele-Blasco E, Seib DR, Chahley E, Skelton PD, Luikart BW, Snyder JS. Adult-born neurons inhibit developmentally-born neurons during spatial learning. Neurobiol Learn Mem 2023; 198:107710. [PMID: 36572174 DOI: 10.1016/j.nlm.2022.107710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Ongoing neurogenesis in the dentate gyrus (DG) subregion of the hippocampus results in a heterogenous population of neurons. Immature adult-born neurons (ABNs) have physiological and anatomical properties that may give them a unique role in learning. For example, compared to older granule neurons, they have greater somatic excitability, which could facilitate their recruitment into memory traces. However, recruitment is also likely to depend on interactions with other DG neurons through processes such as lateral inhibition. Immature ABNs target inhibitory interneurons and, compared to older neurons, they receive less GABAergic inhibition. Thus, they may induce lateral inhibition of mature DG neurons while being less susceptible to inhibition themselves. To test this we used a chemogenetic approach to silence immature ABNs as rats learned a spatial water maze task, and measured activity (Fos expression) in ABNs and developmentally-born neurons (DBNs). A retrovirus expressing the inhibitory DREADD receptor, hM4Di, was injected into the dorsal DG of male rats at 6w to infect neurons born in adulthood. Animals were also injected with BrdU to label DBNs or ABNs. DBNs were significantly more active than immature 4-week-old ABNs. Silencing 4-week-old ABNs did not alter learning but it increased activity in DBNs. However, silencing ABNs did not affect activation in other ABNs within the DG. Silencing ABNs also did not alter Fos expression in parvalbumin- and somatostatin-expressing interneurons. Collectively, these results suggest that ABNs may directly inhibit DBN activity during hippocampal-dependent learning, which may be relevant for maintaining sparse hippocampal representations of experienced events.
Collapse
Affiliation(s)
- Alyssa M Ash
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Elena Regele-Blasco
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Désirée R Seib
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Erin Chahley
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Patrick D Skelton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Jason S Snyder
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
42
|
Kagan BJ, Kitchen AC, Tran NT, Habibollahi F, Khajehnejad M, Parker BJ, Bhat A, Rollo B, Razi A, Friston KJ. In vitro neurons learn and exhibit sentience when embodied in a simulated game-world. Neuron 2022; 110:3952-3969.e8. [PMID: 36228614 DOI: 10.1016/j.neuron.2022.09.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/21/2022] [Accepted: 08/31/2022] [Indexed: 11/06/2022]
Abstract
Integrating neurons into digital systems may enable performance infeasible with silicon alone. Here, we develop DishBrain, a system that harnesses the inherent adaptive computation of neurons in a structured environment. In vitro neural networks from human or rodent origins are integrated with in silico computing via a high-density multielectrode array. Through electrophysiological stimulation and recording, cultures are embedded in a simulated game-world, mimicking the arcade game "Pong." Applying implications from the theory of active inference via the free energy principle, we find apparent learning within five minutes of real-time gameplay not observed in control conditions. Further experiments demonstrate the importance of closed-loop structured feedback in eliciting learning over time. Cultures display the ability to self-organize activity in a goal-directed manner in response to sparse sensory information about the consequences of their actions, which we term synthetic biological intelligence. Future applications may provide further insights into the cellular correlates of intelligence.
Collapse
Affiliation(s)
| | | | - Nhi T Tran
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Forough Habibollahi
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia
| | - Moein Khajehnejad
- Department of Data Science and AI, Monash University, Melbourne, Australia
| | - Bradyn J Parker
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia
| | - Anjali Bhat
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Ben Rollo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Adeel Razi
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK; Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia; Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia; CIFAR Azrieli Global Scholars Program, CIFAR, Toronto, Canada
| | - Karl J Friston
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| |
Collapse
|
43
|
Kim SY, Lim W. Disynaptic effect of hilar cells on pattern separation in a spiking neural network of hippocampal dentate gyrus. Cogn Neurodyn 2022; 16:1427-1447. [PMID: 36408073 PMCID: PMC9666645 DOI: 10.1007/s11571-022-09797-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/25/2022] [Accepted: 03/02/2022] [Indexed: 11/28/2022] Open
Abstract
We study the disynaptic effect of the hilar cells on pattern separation in a spiking neural network of the hippocampal dentate gyrus (DG). The principal granule cells (GCs) in the DG perform pattern separation, transforming similar input patterns into less-similar output patterns. In our DG network, the hilus consists of excitatory mossy cells (MCs) and inhibitory HIPP (hilar perforant path-associated) cells. Here, we consider the disynaptic effects of the MCs and the HIPP cells on the GCs, mediated by the inhibitory basket cells (BCs) in the granular layer; MC → BC → GC and HIPP → BC → GC. The MCs provide disynaptic inhibitory input (mediated by the intermediate BCs) to the GCs, which decreases the firing activity of the GCs. On the other hand, the HIPP cells disinhibit the intermediate BCs, which leads to increasing the firing activity of the GCs. In this way, the disynaptic effects of the MCs and the HIPP cells are opposite. We investigate change in the pattern separation efficacy by varying the synaptic strength K ( BC , X ) [from the pre-synaptic X (= MC or HIPP) to the post-synaptic BC]. Thus, sparsity for the firing activity of the GCs is found to improve the efficacy of pattern separation, and hence the disynaptic effects of the MCs and the HIPP cells on the pattern separation become opposite ones. In the combined case when simultaneously changing both K ( BC , MC ) and K ( BC , HIPP ) , as a result of balance between the two competing disynaptic effects of the MCs and the HIPP cells, the efficacy of pattern separation is found to become the highest at their original default values where the activation degree of the GCs is the lowest. We also note that, while the GCs perform pattern separation, sparsely synchronized rhythm is found to appear in the population of the GCs. Hence, we examine quantitative association between population and individual firing behaviors in the sparsely synchronized rhythm and pattern separation. They are found to be strongly correlated. Consequently, the better the population and individual firing behaviors in the sparsely synchronized rhythm are, the more pattern separation efficacy becomes enhanced.
Collapse
Affiliation(s)
- Sang-Yoon Kim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| | - Woochang Lim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| |
Collapse
|
44
|
Rukundo P, Feng T, Pham V, Pieraut S. Moderate effect of early-life experience on dentate gyrus function. Mol Brain 2022; 15:92. [PMID: 36411441 PMCID: PMC9677655 DOI: 10.1186/s13041-022-00980-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/06/2022] [Indexed: 11/22/2022] Open
Abstract
The development, maturation, and plasticity of neural circuits are strongly influenced by experience and the interaction of an individual with their environment can have a long-lasting effect on cognitive function. Using an enriched environment (EE) paradigm, we have recently demonstrated that enhancing social, physical, and sensory activity during the pre-weaning time in mice led to an increase of inhibitory and excitatory synapses in the dentate gyrus (DG) of the hippocampus. The structural plasticity induced by experience may affect information processing in the circuit. The DG performs pattern separation, a computation that enables the encoding of very similar and overlapping inputs into dissimilar outputs. In the presented study, we have tested the hypothesis that an EE in juvenile mice will affect DG's functions that are relevant for pattern separation: the decorrelation of the inputs from the entorhinal cortex (EC) and the recruitment of the principal excitatory granule cell (GC) during behavior. First, using a novel slice electrophysiology protocol, we found that the transformation of the incoming signal from the EC afferents by individual GC is moderately affected by EE. We further show that EE does not affect behaviorally induced recruitment of principal excitatory GC. Lastly, using the novel object recognition task, a hippocampus-dependent memory test, we show that the ontogeny of this discrimination task was similar among the EE mice and the controls. Taken together, our work demonstrates that pre-weaning enrichment moderately affects DG function.
Collapse
Affiliation(s)
- Pacifique Rukundo
- grid.266818.30000 0004 1936 914XDepartment of Biology, University of Nevada, Reno, NV 89557 USA
| | - Ting Feng
- grid.266818.30000 0004 1936 914XDepartment of Biology, University of Nevada, Reno, NV 89557 USA
| | - Vincent Pham
- grid.266818.30000 0004 1936 914XDepartment of Biology, University of Nevada, Reno, NV 89557 USA
| | - Simon Pieraut
- grid.266818.30000 0004 1936 914XDepartment of Biology, University of Nevada, Reno, NV 89557 USA
| |
Collapse
|
45
|
Khona M, Fiete IR. Attractor and integrator networks in the brain. Nat Rev Neurosci 2022; 23:744-766. [DOI: 10.1038/s41583-022-00642-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 11/06/2022]
|
46
|
Polykretis I, Michmizos KP. The role of astrocytes in place cell formation: A computational modeling study. J Comput Neurosci 2022; 50:505-518. [PMID: 35840871 PMCID: PMC9671849 DOI: 10.1007/s10827-022-00828-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 05/20/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
Abstract
Place cells develop spatially-tuned receptive fields during the early stages of novel environment exploration. The generative mechanism underlying these spatially-selective responses remains largely elusive, but has been associated with theta rhythmicity. An important factor implicating the transformation of silent cells to place cells is a spatially-uniform depolarization that is mediated by a persistent sodium current. This neuronal current is modulated by extracellular calcium concentration, which, in turn, is actively controlled by astrocytes. However, there is no established relationship between the neuronal depolarization and astrocytic activity. To consider this link, we designed a bioplausible computational model of a neuronal-astrocytic network, where astrocytes induced the transient emergence of place fields in silent cells, and accelerated the plasticity-induced consolidation of place cells. Interestingly, theta oscillations emerged naturally at the network level, resulting from the astrocytic modulation of subcellular neuronal properties. Our results suggest that astrocytes participate in spatial mapping and exploration, and further highlight the computational roles of these cells in the brain.
Collapse
Affiliation(s)
- Ioannis Polykretis
- Computational Brain Lab, Department of Computer Science, Rutgers University, New Brunswick, New Jersey, USA
| | - Konstantinos P Michmizos
- Computational Brain Lab, Department of Computer Science, Rutgers University, New Brunswick, New Jersey, USA.
| |
Collapse
|
47
|
John U, Patro N, Patro I. Perineuronal nets: Cruise from a honeycomb to the safety nets. Brain Res Bull 2022; 190:179-194. [DOI: 10.1016/j.brainresbull.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/17/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
|
48
|
Kang YJ, Lee SH, Boychuk JA, Butler CR, Juras JA, Cloyd RA, Smith BN. Adult Born Dentate Granule Cell Mediated Upregulation of Feedback Inhibition in a Mouse Model of Traumatic Brain Injury. J Neurosci 2022; 42:7077-7093. [PMID: 36002261 PMCID: PMC9480876 DOI: 10.1523/jneurosci.2263-21.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Post-traumatic epilepsy (PTE) and behavioral comorbidities frequently develop after traumatic brain injury (TBI). Aberrant neurogenesis of dentate granule cells (DGCs) after TBI may contribute to the synaptic reorganization that occurs in PTE, but how neurogenesis at different times relative to the injury contributes to feedback inhibition and recurrent excitation in the dentate gyrus is unknown. Thus, we examined whether DGCs born at different postnatal ages differentially participate in feedback inhibition and recurrent excitation in the dentate gyrus using the controlled cortical impact (CCI) model of TBI. Both sexes of transgenic mice expressing channelrhodopsin2 (ChR2) in postnatally born DGCs were used for optogenetic activation of three DGC cohorts: postnatally early born DGCs, or those born just before or after CCI. We performed whole-cell patch-clamp recordings from ChR2-negative, mature DGCs and parvalbumin-expressing basket cells (PVBCs) in hippocampal slices to determine whether optogenetic activation of postnatally born DGCs increases feedback inhibition and/or recurrent excitation in mice 8-10 weeks after CCI and whether PVBCs are targets of ChR2-positive DGCs. In the dentate gyrus ipsilateral to CCI, activation of ChR2-expressing DGCs born before CCI produced increased feedback inhibition in ChR2-negative DGCs and increased excitation in PVBCs compared with those from sham controls. This upregulated feedback inhibition was less prominent in DGCs born early in life or after CCI. Surprisingly, ChR2-positive DGC activation rarely evoked recurrent excitation in mature DGCs from any cohort. These results support that DGC birth date-related increased feedback inhibition in of DGCs may contribute to altered excitability after TBI.SIGNIFICANCE STATEMENT Dentate granule cells (DGCs) control excitability of the dentate gyrus through synaptic interactions with inhibitory GABAergic interneurons. Persistent changes in DGC synaptic connectivity develop after traumatic brain injury, contributing to hyperexcitability in post-traumatic epilepsy (PTE). However, the impact of DGC neurogenesis on synaptic reorganization, especially on inhibitory circuits, after brain injury is not adequately described. Here, upregulation of feedback inhibition in mature DGCs from male and female mice was associated with increased excitation of parvalbumin-expressing basket cells by postnatally born DGCs, providing novel insights into underlying mechanisms of altered excitability after brain injury. A better understanding of these inhibitory circuit changes can help formulate hypotheses for development of novel, evidence-based treatments for post-traumatic epilepsy by targeting birth date-specific subsets of DGCs.
Collapse
Affiliation(s)
- Young-Jin Kang
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Sang-Hun Lee
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Epilepsy Research Center, University of Kentucky, Lexington, Kentucky 40536
| | - Jeffery A Boychuk
- Epilepsy Research Center, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Corwin R Butler
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - J Anna Juras
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Ryan A Cloyd
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Bret N Smith
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Epilepsy Research Center, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
49
|
Diederich N, Ziegler M, Kaernbach C. Artificial neural network performance based on correlation analysis qualitatively comparable with human performance in behavioral signal detection experiments. J Neurophysiol 2022; 128:279-289. [PMID: 35766442 DOI: 10.1152/jn.00393.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Standard Gaussian signal detection theory (SDT) is a widely used approach to assess the detection performance of living organisms or technical systems without looking at the inner workings of these systems like neural or electronic mechanisms. Nevertheless, a consideration of the inner mechanisms of a system and how they produce observed behaviors should help to better understand the functioning. It might even offer the possibility to demonstrate isolated pattern separation processes directly in the model. To do so, modeling the interaction between the entorhinal cortex (EC) and the hippocampal subnetwork dentate gyrus (DG) via the perforant path reveals the decorrelation network's mode of operation. We show that the ability to do pattern separation is crucial for high-performance pattern recognition, but also for lure discrimination, and depends on the proportionality between input and output network. NEW & NOTEWORTHY We elucidate the interplay of the entorhinal cortex and the hippocampal dentate gyrus during pattern separation tasks by providing a new simulation model. Functional memory formation and processing of similar memory content is illuminated from within the system. For the first time orthogonalized spiking patterns are evaluated with signal detection theory methods, and the results are applied to clinically established and novel tests.
Collapse
Affiliation(s)
- Nick Diederich
- Micro- and Nanoelectronic Systems, Institute of Micro- and Nanotechnologies-IMN MacroNano®, Technische Universität Ilmenau, Ilmenau, Germany.,Nanoelectronics, Faculty of Engineering, University of Kiel, Kiel, Germany
| | - Martin Ziegler
- Micro- and Nanoelectronic Systems, Institute of Micro- and Nanotechnologies-IMN MacroNano®, Technische Universität Ilmenau, Ilmenau, Germany
| | - Christian Kaernbach
- Department of Psychology, Experimental and Biological Psychology, University of Kiel, Kiel, Germany
| |
Collapse
|
50
|
Arutiunian V, Arcara G, Buyanova I, Gomozova M, Dragoy O. The age-related changes in 40 Hz Auditory Steady-State Response and sustained Event-Related Fields to the same amplitude-modulated tones in typically developing children: A magnetoencephalography study. Hum Brain Mapp 2022; 43:5370-5383. [PMID: 35833318 PMCID: PMC9812253 DOI: 10.1002/hbm.26013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 01/15/2023] Open
Abstract
Recent studies have revealed that gamma-band oscillatory and transient evoked potentials may change with age during childhood. It is hypothesized that these changes can be associated with a maturation of GABAergic neurotransmission and, subsequently, the age-related changes of excitation-inhibition balance in the neural circuits. One of the reliable paradigms for investigating these effects in the auditory cortex is 40 Hz Auditory Steady-State Response (ASSR), where participants are presented with the periodic auditory stimuli. It is known that such stimuli evoke two types of responses in magnetoencephalography (MEG)-40 Hz steady-state gamma response (or 40 Hz ASSR) and auditory evoked response called sustained Event-Related Field (ERF). Although several studies have been conducted in children, focusing on the changes of 40 Hz ASSR with age, almost nothing is known about the age-related changes of the sustained ERF to the same periodic stimuli and their relationships with changes in the gamma strength. Using MEG, we investigated the association between 40 Hz steady-state gamma response and sustained ERF response to the same stimuli and also their age-related changes in the group of 30 typically developing 7-to-12-year-old children. The results revealed a tight relationship between 40 Hz ASSR and ERF, indicating that the age-related increase in strength of 40 Hz ASSR was associated with the age-related decrease of the amplitude of ERF. These effects were discussed in the light of the maturation of the GABAergic system and excitation-inhibition balance development, which may contribute to the changes in ASSR and ERF.
Collapse
Affiliation(s)
| | | | | | | | - Olga Dragoy
- Center for Language and BrainHSE UniversityMoscowRussia,Institute of LinguisticsRussian Academy of SciencesMoscowRussia
| |
Collapse
|