1
|
Gao YZ, Liu K, Wu XM, Shi CN, He QL, Wu HP, Yang JJ, Yao H, Ji MH. Oxidative Stress-mediated Loss of Hippocampal Parvalbumin Interneurons Contributes to Memory Precision Decline After Acute Sleep Deprivation. Mol Neurobiol 2025; 62:5377-5394. [PMID: 39546120 DOI: 10.1007/s12035-024-04628-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
Sleep is pivotal to memory consolidation, and sleep deprivation (SD) after learning can impede this process, leading to memory disorders. In the present study, we aimed to explore the effects of acute sleep deprivation (ASD) on memory disorders and the underlying mechanisms. ASD model was induced by subjecting the mice to 6 h of SD following fear conditioning training. Different cohorts were used for behavioral, biochemical, and electrophysiological tests. Here, we showed that memory precision decline was induced by ASD, concomitant with a notable elevation in oxidative stress within PV interneurons, loss of PV, and disturbed neuronal oscillation in the CA1 region. Notably, chemogenetic activation of PV interneurons effectively ameliorated abnormal gamma oscillation and memory precision decline observed in ASD mice. Meanwhile, chemogenetic inhibition of PV interneurons successfully mimicked the abnormal brain oscillations and memory precision decline observed in ASD mice. Additionally, prior administration of the antioxidant medication N-acetylcysteine effectively reversed memory precision decline and mitigated PV loss and abnormal oscillation triggered by ASD. Collectively, our findings indicated that ASD increased oxidative stress in PV interneurons, thereby disrupting neural oscillation in the CA1 and ultimately leading to memory precision decline.
Collapse
Affiliation(s)
- Yu-Zhu Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Miao Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui-Na Shi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiu-Li He
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Peng Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Hao Yao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Mu-Huo Ji
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Selten M, Bernard C, Mukherjee D, Hamid F, Hanusz-Godoy A, Oozeer F, Zimmer C, Marín O. Regulation of PV interneuron plasticity by neuropeptide-encoding genes. Nature 2025:10.1038/s41586-025-08933-z. [PMID: 40307547 DOI: 10.1038/s41586-025-08933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
Neuronal activity must be regulated in a narrow permissive band for the proper operation of neural networks. Changes in synaptic connectivity and network activity-for example, during learning-might disturb this balance, eliciting compensatory mechanisms to maintain network function1-3. In the neocortex, excitatory pyramidal cells and inhibitory interneurons exhibit robust forms of stabilizing plasticity. However, although neuronal plasticity has been thoroughly studied in pyramidal cells4-8, little is known about how interneurons adapt to persistent changes in their activity. Here we describe a critical cellular process through which cortical parvalbumin-expressing (PV+) interneurons adapt to changes in their activity levels. We found that changes in the activity of individual PV+ interneurons drive bidirectional compensatory adjustments of the number and strength of inhibitory synapses received by these cells, specifically from other PV+ interneurons. High-throughput profiling of ribosome-associated mRNA revealed that increasing the activity of a PV+ interneuron leads to upregulation of two genes encoding multiple secreted neuropeptides: Vgf and Scg2. Functional experiments demonstrated that VGF is critically required for the activity-dependent scaling of inhibitory PV+ synapses onto PV+ interneurons. Our findings reveal an instructive role for neuropeptide-encoding genes in regulating synaptic connections among PV+ interneurons in the adult mouse neocortex.
Collapse
Affiliation(s)
- Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Clémence Bernard
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Diptendu Mukherjee
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Alicia Hanusz-Godoy
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Christoph Zimmer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
3
|
Aton SJ. Melody for a memory: sleep boosts the brain's representation of sequential events. Sleep 2025; 48:zsae316. [PMID: 39749971 PMCID: PMC11893525 DOI: 10.1093/sleep/zsae316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Indexed: 01/04/2025] Open
Affiliation(s)
- Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
4
|
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
|
5
|
E Said S, Miyamoto D. Multi-region processing during sleep for memory and cognition. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2025; 101:107-128. [PMID: 40074337 DOI: 10.2183/pjab.101.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Over the past decades, the understanding of sleep has evolved to be a fundamental physiological mechanism integral to the processing of different types of memory rather than just being a passive brain state. The cyclic sleep substates, namely, rapid eye movement (REM) sleep and non-REM (NREM) sleep, exhibit distinct yet complementary oscillatory patterns that form inter-regional networks between different brain regions crucial to learning, memory consolidation, and memory retrieval. Technical advancements in imaging and manipulation approaches have provided deeper understanding of memory formation processes on multi-scales including brain-wide, synaptic, and molecular levels. The present review provides a short background and outlines the current state of research and future perspectives in understanding the role of sleep and its substates in memory processing from both humans and rodents, with a focus on cross-regional brain communication, oscillation coupling, offline reactivations, and engram studies. Moreover, we briefly discuss how sleep contributes to other higher-order cognitive functions.
Collapse
Affiliation(s)
- Salma E Said
- Laboratory for Sleeping-Brain Dynamics, Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Daisuke Miyamoto
- Laboratory for Sleeping-Brain Dynamics, Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
6
|
Ognjanovski N, Kim DS, Charlett-Green E, Goldiez E, van Koppen S, Aton SJ, Watson BO. Daily rhythms drive dynamism in sleep, oscillations and interneuron firing, while excitatory firing remains stable across 24 h. Eur J Neurosci 2025; 61:e16619. [PMID: 39663213 DOI: 10.1111/ejn.16619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/25/2024] [Accepted: 11/10/2024] [Indexed: 12/13/2024]
Abstract
The adaptation to the daily 24-h light-dark cycle is ubiquitous across animal species and is crucial for maintaining fitness. This free-running cycle occurs innately within multiple bodily systems, such as endogenous circadian rhythms in clock-gene expression and synaptic plasticity. These phenomena are well studied; however, it is unknown if and how the 24-h clock affects electrophysiologic network function in vivo. The hippocampus is a region of interest for long timescale (>8 h) studies because it is critical for cognitive function and exhibits time-of-day effects in learning. We recorded single cell spiking activity and local field potentials (LFPs) in mouse hippocampus across the 24-h (12:12-h light/dark) cycle to quantify how electrophysiological network function is modulated across the 24-h day. We found that while inhibitory population firing rates and LFP oscillations exhibit modulation across the day, average excitatory population firing is static. This excitatory stability, despite inhibitory dynamism, may enable consistent around-the-clock function of neural circuits.
Collapse
Affiliation(s)
- Nicolette Ognjanovski
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - David S Kim
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Emma Charlett-Green
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Ethan Goldiez
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Sofie van Koppen
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Brendon O Watson
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
7
|
Burkart ME, Kurzke J, Jacobi R, Vera J, Ashcroft FM, Eilers J, Lippmann K. KATP channel mutation disrupts hippocampal network activity and nocturnal gamma shifts. Brain 2024; 147:4200-4212. [PMID: 38748482 DOI: 10.1093/brain/awae157] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/31/2024] [Accepted: 05/02/2024] [Indexed: 12/14/2024] Open
Abstract
ATP-sensitive potassium (KATP) channels couple cell metabolism to cellular electrical activity. Humans affected by severe activating mutations in KATP channels suffer from developmental delay, epilepsy and neonatal diabetes (DEND syndrome). While the aetiology of diabetes in DEND syndrome is well understood, the pathophysiology of the neurological symptoms remains unclear. We hypothesized that impaired activity of parvalbumin-positive interneurons (PV-INs) may result in seizures and cognitive problems. We found, by performing electrophysiological experiments, that expressing the DEND mutation Kir6.2-V59M selectively in mouse PV-INs reduced intrinsic gamma frequency preference and short-term depression as well as disturbed cognition-associated gamma oscillations and hippocampal sharp waves. Furthermore, the risk of seizures was increased and the day-night shift in gamma activity disrupted. Blocking KATP channels with tolbutamide partially rescued the network oscillations. The non-reversible part may, to some extent, result from observed altered PV-IN dendritic branching and PV-IN arrangement within CA1. In summary, PV-INs play a key role in DEND syndrome, and this provides a framework for establishing treatment options.
Collapse
Affiliation(s)
- Marie-Elisabeth Burkart
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| | - Josephine Kurzke
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| | - Robert Jacobi
- Department for Neurophysiology, Institute for Physiology, Julius-Maximilians-University Würzburg, Würzburg 97070, Germany
| | - Jorge Vera
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Frances M Ashcroft
- Henry Wellcome Centre for Gene Function, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Jens Eilers
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| | - Kristina Lippmann
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| |
Collapse
|
8
|
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
|
9
|
Swift KM, Gary NC, Urbanczyk PJ. On the basis of sex and sleep: the influence of the estrous cycle and sex on sleep-wake behavior. Front Neurosci 2024; 18:1426189. [PMID: 39268035 PMCID: PMC11390649 DOI: 10.3389/fnins.2024.1426189] [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: 04/30/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
The recurrent hormonal fluctuations within reproductive cycles impact sleep-wake behavior in women and in rats and mice used in preclinical models of sleep research. Strides have been made in sleep-related clinical trials to include equal numbers of women; however, the inclusion of female rodents in neuroscience and sleep research is lacking. Female animals are commonly omitted from studies over concerns of the effect of estrus cycle hormones on measured outcomes. This review highlights the estrous cycle's broad effects on sleep-wake behavior: from changes in sleep macroarchitecture to regionally specific alterations in neural oscillations. These changes are largely driven by cycle-dependent ovarian hormonal fluctuations occurring during proestrus and estrus that modulate neural circuits regulating sleep-wake behavior. Removal of estrous cycle influence by ovariectomy ablates characteristic sleep changes. Further, sex differences in sleep are present between gonadally intact females and males. Removal of reproductive hormones via gonadectomy in both sexes mitigates some, but not all sex differences. We examine the extent to which reproductive hormones and sex chromosomes contribute to sex differences in sleep-wake behavior. Finally, this review addresses the limitations in our understanding of the estrous cycle's impact on sleep-wake behavior, gaps in female sleep research that are well studied in males, and the implications that ignoring the estrous cycle has on studies of sleep-related processes.
Collapse
Affiliation(s)
- Kevin M Swift
- Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Nicholas C Gary
- Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Phillip J Urbanczyk
- Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
10
|
Yang D, Qi G, Ort J, Witzig V, Bak A, Delev D, Koch H, Feldmeyer D. Modulation of large rhythmic depolarizations in human large basket cells by norepinephrine and acetylcholine. Commun Biol 2024; 7:885. [PMID: 39033173 PMCID: PMC11271271 DOI: 10.1038/s42003-024-06546-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
Rhythmic brain activity is critical to many brain functions and is sensitive to neuromodulation, but so far very few studies have investigated this activity on the cellular level in vitro in human brain tissue samples. This study reveals and characterizes a novel rhythmic network activity in the human neocortex. Using intracellular patch-clamp recordings of human cortical neurons, we identify large rhythmic depolarizations (LRDs) driven by glutamate release but not by GABA. These LRDs are intricate events made up of multiple depolarizing phases, occurring at ~0.3 Hz, have large amplitudes and long decay times. Unlike human tissue, rat neocortex layers 2/3 exhibit no such activity under identical conditions. LRDs are mainly observed in a subset of L2/3 interneurons that receive substantial excitatory inputs and are likely large basket cells based on their morphology. LRDs are highly sensitive to norepinephrine (NE) and acetylcholine (ACh), two neuromodulators that affect network dynamics. NE increases LRD frequency through β-adrenergic receptor activity while ACh decreases it via M4 muscarinic receptor activation. Multi-electrode array recordings show that NE enhances and synchronizes oscillatory network activity, whereas ACh causes desynchronization. Thus, NE and ACh distinctly modulate LRDs, exerting specific control over human neocortical activity.
Collapse
Affiliation(s)
- Danqing Yang
- Research Center Juelich, Institute of Neuroscience and Medicine 10, Research Center Juelich, 52425, Juelich, Germany
- Department of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Guanxiao Qi
- Research Center Juelich, Institute of Neuroscience and Medicine 10, Research Center Juelich, 52425, Juelich, Germany
| | - Jonas Ort
- Department of Neurosurgery, Faculty of Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Neurosurgical Artificial Intelligence Laboratory Aachen (NAILA), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Center for Integrated Oncology, Universities Aachen, Bonn, Cologne, Düsseldorf (CIO ABCD), Bonn, Germany
| | - Victoria Witzig
- Department of Neurology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Aniella Bak
- Department of Neurology, Section Epileptology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Daniel Delev
- Department of Neurosurgery, Faculty of Medicine, RWTH Aachen University Hospital, Aachen, Germany
- Neurosurgical Artificial Intelligence Laboratory Aachen (NAILA), RWTH Aachen University Hospital, 52074, Aachen, Germany
- Center for Integrated Oncology, Universities Aachen, Bonn, Cologne, Düsseldorf (CIO ABCD), Bonn, Germany
| | - Henner Koch
- Department of Neurology, Section Epileptology, RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Dirk Feldmeyer
- Research Center Juelich, Institute of Neuroscience and Medicine 10, Research Center Juelich, 52425, Juelich, Germany.
- Department of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University Hospital, 52074, Aachen, Germany.
- Jülich-Aachen Research Alliance, Translational Brain Medicine (JARA Brain), Aachen, Germany.
| |
Collapse
|
11
|
Hadler MD, Alle H, Geiger JRP. Parvalbumin interneuron cell-to-network plasticity: mechanisms and therapeutic avenues. Trends Pharmacol Sci 2024; 45:586-601. [PMID: 38763836 DOI: 10.1016/j.tips.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Alzheimer's disease (AD) and schizophrenia (SCZ) represent two major neuropathological conditions with a high disease burden. Despite their distinct etiologies, patients suffering from AD or SCZ share a common burden of disrupted memory functions unattended by current therapies. Recent preclinical analyses highlight cell-type-specific contributions of parvalbumin interneurons (PVIs), particularly the plasticity of their cellular excitability, towards intact neuronal network function (cell-to-network plasticity) and memory performance. Here we argue that deficits of PVI cell-to-network plasticity may underlie memory deficits in AD and SCZ, and we explore two therapeutic avenues: the targeting of PVI-specific neuromodulation, including by neuropeptides, and the recruitment of network synchrony in the gamma frequency range (40 Hz) by external stimulation. We finally propose that these approaches be merged under consideration of recent insights into human brain physiology.
Collapse
Affiliation(s)
- Michael D Hadler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R P Geiger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Bodea GO, Botto JM, Ferreiro ME, Sanchez-Luque FJ, de Los Rios Barreda J, Rasmussen J, Rahman MA, Fenlon LR, Jansz N, Gubert C, Gerdes P, Bodea LG, Ajjikuttira P, Da Costa Guevara DJ, Cumner L, Bell CC, Kozulin P, Billon V, Morell S, Kempen MJHC, Love CJ, Saha K, Palmer LM, Ewing AD, Jhaveri DJ, Richardson SR, Hannan AJ, Faulkner GJ. LINE-1 retrotransposons contribute to mouse PV interneuron development. Nat Neurosci 2024; 27:1274-1284. [PMID: 38773348 PMCID: PMC11239520 DOI: 10.1038/s41593-024-01650-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/14/2024] [Indexed: 05/23/2024]
Abstract
Retrotransposons are mobile DNA sequences duplicated via transcription and reverse transcription of an RNA intermediate. Cis-regulatory elements encoded by retrotransposons can also promote the transcription of adjacent genes. Somatic LINE-1 (L1) retrotransposon insertions have been detected in mammalian neurons. It is, however, unclear whether L1 sequences are mobile in only some neuronal lineages or therein promote neurodevelopmental gene expression. Here we report programmed L1 activation by SOX6, a transcription factor critical for parvalbumin (PV) interneuron development. Mouse PV interneurons permit L1 mobilization in vitro and in vivo, harbor unmethylated L1 promoters and express full-length L1 mRNAs and proteins. Using nanopore long-read sequencing, we identify unmethylated L1s proximal to PV interneuron genes, including a novel L1 promoter-driven Caps2 transcript isoform that enhances neuron morphological complexity in vitro. These data highlight the contribution made by L1 cis-regulatory elements to PV interneuron development and transcriptome diversity, uncovered due to L1 mobility in this milieu.
Collapse
Affiliation(s)
- Gabriela O Bodea
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia.
| | - Juan M Botto
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Maria E Ferreiro
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Francisco J Sanchez-Luque
- Institute of Parasitology and Biomedicine 'López-Neyra', Spanish National Research Council, Granada, Spain
| | | | - Jay Rasmussen
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Muhammed A Rahman
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Laura R Fenlon
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Natasha Jansz
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Patricia Gerdes
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Liviu-Gabriel Bodea
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Prabha Ajjikuttira
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Darwin J Da Costa Guevara
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Linda Cumner
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Charles C Bell
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Peter Kozulin
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Victor Billon
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Biology Department, École Normale Supérieure Paris-Saclay, Gif-sur-Yvette, France
| | - Santiago Morell
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Marie-Jeanne H C Kempen
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Chloe J Love
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Karabi Saha
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, USA
| | - Lucy M Palmer
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Adam D Ewing
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Sandra R Richardson
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Faulkner
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
- Mater Research Institute - University of Queensland, TRI Building, Woolloongabba, Queensland, Australia.
| |
Collapse
|
13
|
De Paolis ML, Paoletti I, Zaccone C, Capone F, D'Amelio M, Krashia P. Transcranial alternating current stimulation (tACS) at gamma frequency: an up-and-coming tool to modify the progression of Alzheimer's Disease. Transl Neurodegener 2024; 13:33. [PMID: 38926897 PMCID: PMC11210106 DOI: 10.1186/s40035-024-00423-y] [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: 01/08/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
The last decades have witnessed huge efforts devoted to deciphering the pathological mechanisms underlying Alzheimer's Disease (AD) and to testing new drugs, with the recent FDA approval of two anti-amyloid monoclonal antibodies for AD treatment. Beyond these drug-based experimentations, a number of pre-clinical and clinical trials are exploring the benefits of alternative treatments, such as non-invasive stimulation techniques on AD neuropathology and symptoms. Among the different non-invasive brain stimulation approaches, transcranial alternating current stimulation (tACS) is gaining particular attention due to its ability to externally control gamma oscillations. Here, we outline the current knowledge concerning the clinical efficacy, safety, ease-of-use and cost-effectiveness of tACS on early and advanced AD, applied specifically at 40 Hz frequency, and also summarise pre-clinical results on validated models of AD and ongoing patient-centred trials.
Collapse
Affiliation(s)
- Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21 - 00128, Rome, Italy
| | - Ilaria Paoletti
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21 - 00128, Rome, Italy
| | - Claudio Zaccone
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21 - 00128, Rome, Italy
| | - Fioravante Capone
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21 - 00128, Rome, Italy
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200 - 00128, Rome, Italy
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21 - 00128, Rome, Italy.
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64 - 00143, Rome, Italy.
| | - Paraskevi Krashia
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64 - 00143, Rome, Italy
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21 - 00128, Rome, Italy
| |
Collapse
|
14
|
Abbaspoor S, Hoffman KL. Circuit dynamics of superficial and deep CA1 pyramidal cells and inhibitory cells in freely-moving macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.06.570369. [PMID: 38106053 PMCID: PMC10723348 DOI: 10.1101/2023.12.06.570369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Diverse neuron classes in hippocampal CA1 have been identified through the heterogeneity of their cellular/molecular composition. How these classes relate to hippocampal function and the network dynamics that support cognition in primates remains unclear. Here we report inhibitory functional cell groups in CA1 of freely-moving macaques whose diverse response profiles to network states and each other suggest distinct and specific roles in the functional microcircuit of CA1. In addition, pyramidal cells that were segregated into superficial and deep layers differed in firing rate, burstiness, and sharp-wave ripple-associated firing. They also showed strata-specific spike-timing interactions with inhibitory cell groups, suggestive of segregated neural populations. Furthermore, ensemble recordings revealed that cell assemblies were preferentially organized according to these strata. These results suggest sublayer-specific circuit organization in hippocampal CA1 of the freely-moving macaques that may underlie its role in cognition.
Collapse
Affiliation(s)
- S Abbaspoor
- Department of Psychology, Vanderbilt Vision Research Center, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee
| | - K L Hoffman
- Department of Psychology, Vanderbilt Vision Research Center, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
15
|
Leitch B. Parvalbumin Interneuron Dysfunction in Neurological Disorders: Focus on Epilepsy and Alzheimer's Disease. Int J Mol Sci 2024; 25:5549. [PMID: 38791587 PMCID: PMC11122153 DOI: 10.3390/ijms25105549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Parvalbumin expressing (PV+) GABAergic interneurons are fast spiking neurons that provide powerful but relatively short-lived inhibition to principal excitatory cells in the brain. They play a vital role in feedforward and feedback synaptic inhibition, preventing run away excitation in neural networks. Hence, their dysfunction can lead to hyperexcitability and increased susceptibility to seizures. PV+ interneurons are also key players in generating gamma oscillations, which are synchronized neural oscillations associated with various cognitive functions. PV+ interneuron are particularly vulnerable to aging and their degeneration has been associated with cognitive decline and memory impairment in dementia and Alzheimer's disease (AD). Overall, dysfunction of PV+ interneurons disrupts the normal excitatory/inhibitory balance within specific neurocircuits in the brain and thus has been linked to a wide range of neurodevelopmental and neuropsychiatric disorders. This review focuses on the role of dysfunctional PV+ inhibitory interneurons in the generation of epileptic seizures and cognitive impairment and their potential as targets in the design of future therapeutic strategies to treat these disorders. Recent research using cutting-edge optogenetic and chemogenetic technologies has demonstrated that they can be selectively manipulated to control seizures and restore the balance of neural activity in the brains of animal models. This suggests that PV+ interneurons could be important targets in developing future treatments for patients with epilepsy and comorbid disorders, such as AD, where seizures and cognitive decline are directly linked to specific PV+ interneuron deficits.
Collapse
Affiliation(s)
- Beulah Leitch
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
16
|
Mota B, Brás AR, Araújo-Andrade L, Silva A, Pereira PA, Madeira MD, Cardoso A. High-Caloric Diets in Adolescence Impair Specific GABAergic Subpopulations, Neurogenesis, and Alter Astrocyte Morphology. Int J Mol Sci 2024; 25:5524. [PMID: 38791562 PMCID: PMC11122083 DOI: 10.3390/ijms25105524] [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: 04/10/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
We compared the effects of two different high-caloric diets administered to 4-week-old rats for 12 weeks: a diet rich in sugar (30% sucrose) and a cafeteria diet rich in sugar and high-fat foods. We focused on the hippocampus, particularly on the gamma-aminobutyric acid (GABA)ergic system, including the Ca2+-binding proteins parvalbumin (PV), calretinin (CR), calbindin (CB), and the neuropeptides somatostatin (SST) and neuropeptide Y (NPY). We also analyzed the density of cholinergic varicosities, brain-derived neurotrophic factor (BDNF), reelin (RELN), and cyclin-dependent kinase-5 (CDK-5) mRNA levels, and glial fibrillary acidic protein (GFAP) expression. The cafeteria diet reduced PV-positive neurons in the granular layer, hilus, and CA1, as well as NPY-positive neurons in the hilus, without altering other GABAergic populations or overall GABA levels. The high-sugar diet induced a decrease in the number of PV-positive cells in CA3 and an increase in CB-positive cells in the hilus and CA1. No alterations were observed in the cholinergic varicosities. The cafeteria diet also reduced the relative mRNA expression of RELN without significant changes in BDNF and CDK5 levels. The cafeteria diet increased the number but reduced the length of the astrocyte processes. These data highlight the significance of determining the mechanisms mediating the observed effects of these diets and imply that the cognitive impairments previously found might be related to both the neuroinflammation process and the reduction in PV, NPY, and RELN expression in the hippocampal formation.
Collapse
Affiliation(s)
- Bárbara Mota
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Rita Brás
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
| | - Leonardo Araújo-Andrade
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Silva
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Pedro A. Pereira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - M. Dulce Madeira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Armando Cardoso
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
17
|
Dirven BCJ, van Melis L, Daneva T, Dillen L, Homberg JR, Kozicz T, Henckens MJAG. Hippocampal Trauma Memory Processing Conveying Susceptibility to Traumatic Stress. Neuroscience 2024; 540:87-102. [PMID: 38220126 DOI: 10.1016/j.neuroscience.2024.01.007] [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: 02/27/2023] [Revised: 12/04/2023] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
While the majority of the population is ever exposed to a traumatic event during their lifetime, only a fraction develops posttraumatic stress disorder (PTSD). Disrupted trauma memory processing has been proposed as a core factor underlying PTSD symptomatology. We used transgenic Targeted-Recombination-in-Active-Populations (TRAP) mice to investigate potential alterations in trauma-related hippocampal memory engrams associated with the development of PTSD-like symptomatology. Mice were exposed to a stress-enhanced fear learning paradigm, in which prior exposure to a stressor affects the learning of a subsequent fearful event (contextual fear conditioning using foot shocks), during which neuronal activity was labeled. One week later, mice were behaviorally phenotyped to identify mice resilient and susceptible to developing PTSD-like symptomatology. Three weeks post-learning, mice were re-exposed to the conditioning context to induce remote fear memory recall, and associated hippocampal neuronal activity was assessed. While no differences in the size of the hippocampal neuronal ensemble activated during fear learning were observed between groups, susceptible mice displayed a smaller ensemble activated upon remote fear memory recall in the ventral CA1, higher regional hippocampal parvalbuminneuronal density and a relatively lower activity of parvalbumininterneurons upon recall. Investigation of potential epigenetic regulators of the engram revealed rather generic (rather than engram-specific) differences between groups, with susceptible mice displaying lower hippocampal histone deacetylase 2 expression, and higher methylation and hydroxymethylation levels. These finding implicate variation in epigenetic regulation within the hippocampus, as well as reduced regional hippocampal activity during remote fear memory recall in interindividual differences in susceptibility to traumatic stress.
Collapse
Affiliation(s)
- Bart C J Dirven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Lennart van Melis
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Teya Daneva
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Lieke Dillen
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Tamas Kozicz
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Center for Individualized Medicine, Department of Clinical Genomics, and Biochemical Genetics Laboratory, Mayo Clinic, Rochester, MN 55905, USA; University of Pecs Medical School, Department of Anatomy, Pecs, Hungary
| | - Marloes J A G Henckens
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Wilson DA, Sullivan RM, Smiley JF, Saito M, Raineki C. Developmental alcohol exposure is exhausting: Sleep and the enduring consequences of alcohol exposure during development. Neurosci Biobehav Rev 2024; 158:105567. [PMID: 38309498 PMCID: PMC10923002 DOI: 10.1016/j.neubiorev.2024.105567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Prenatal alcohol exposure is the leading nongenetic cause of human intellectual impairment. The long-term impacts of prenatal alcohol exposure on health and well-being are diverse, including neuropathology leading to behavioral, cognitive, and emotional impairments. Additionally negative effects also occur on the physiological level, such as the endocrine, cardiovascular, and immune systems. Among these diverse impacts is sleep disruption. In this review, we describe how prenatal alcohol exposure affects sleep, and potential mechanisms of those effects. Furthermore, we outline the evidence that sleep disruption across the lifespan may be a mediator of some cognitive and behavioral impacts of developmental alcohol exposure, and thus may represent a promising target for treatment.
Collapse
Affiliation(s)
- Donald A Wilson
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, USA.
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, USA
| | - John F Smiley
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Psychiatry, New York University Medical Center, New York, NY, USA
| | - Mariko Saito
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Psychiatry, New York University Medical Center, New York, NY, USA
| | - Charlis Raineki
- Department of Psychology, Brock University, St. Catharines, ON, Canada; Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
19
|
Hadler MD, Tzilivaki A, Schmitz D, Alle H, Geiger JRP. Gamma oscillation plasticity is mediated via parvalbumin interneurons. SCIENCE ADVANCES 2024; 10:eadj7427. [PMID: 38295164 PMCID: PMC10830109 DOI: 10.1126/sciadv.adj7427] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024]
Abstract
Understanding the plasticity of neuronal networks is an emerging field of (patho-) physiological research, yet the underlying cellular mechanisms remain poorly understood. Gamma oscillations (30 to 80 hertz), a biomarker of cognitive performance, require and potentiate glutamatergic transmission onto parvalbumin-positive interneurons (PVIs), suggesting an interface for cell-to-network plasticity. In ex vivo local field potential recordings, we demonstrate long-term potentiation of hippocampal gamma power. Gamma potentiation obeys established rules of PVI plasticity, requiring calcium-permeable AMPA receptors (CP-AMPARs) and metabotropic glutamate receptors (mGluRs). A microcircuit computational model of CA3 gamma oscillations predicts CP-AMPAR plasticity onto PVIs critically outperforms pyramidal cell plasticity in increasing gamma power and completely accounts for gamma potentiation. We reaffirm this ex vivo in three PVI-targeting animal models, demonstrating that gamma potentiation requires PVI-specific signaling via a Gq/PKC pathway comprising mGluR5 and a Gi-sensitive, PKA-dependent pathway. Gamma activity-dependent, metabotropically mediated CP-AMPAR plasticity on PVIs may serve as a guiding principle in understanding network plasticity in health and disease.
Collapse
Affiliation(s)
- Michael D. Hadler
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Neurocure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Neurocure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle-Straße 10, 13125 Berlin, Germany
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R. P. Geiger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
20
|
Satchell M, Fry B, Noureddine Z, Simmons A, Ognjanovski NN, Aton SJ, Zochowski MR. Neuromodulation via muscarinic acetylcholine pathway can facilitate distinct, complementary, and sequential roles for NREM and REM states during sleep-dependent memory consolidation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.19.541465. [PMID: 38293183 PMCID: PMC10827095 DOI: 10.1101/2023.05.19.541465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Across vertebrate species, sleep consists of repeating cycles of NREM followed by REM. However, the respective functions of NREM, REM, and their stereotypic cycling pattern are not well understood. Using a simplified biophysical network model, we show that NREM and REM sleep can play differential and critical roles in memory consolidation primarily regulated, based on state-specific changes in cholinergic signaling. Within this network, decreasing and increasing muscarinic acetylcholine (ACh) signaling during bouts of NREM and REM, respectively, differentially alters neuronal excitability and excitatory/inhibitory balance. During NREM, deactivation of inhibitory neurons leads to network-wide disinhibition and bursts of synchronized activity led by firing in engram neurons. These features strengthen connections from the original engram neurons to less-active network neurons. In contrast, during REM, an increase in network inhibition suppresses firing in all but the most-active excitatory neurons, leading to competitive strengthening/pruning of the memory trace. We tested the predictions of the model against in vivo recordings from mouse hippocampus during active sleep-dependent memory storage. Consistent with modeling results, we find that functional connectivity between CA1 neurons changes differentially at transition from NREM to REM sleep during learning. Returning to the model, we find that an iterative sequence of state-specific activations during NREM/REM cycling is essential for memory storage in the network, serving a critical role during simultaneous consolidation of multiple memories. Together these results provide a testable mechanistic hypothesis for the respective roles of NREM and REM sleep, and their universal relative timing, in memory consolidation. Significance statement Using a simplified computational model and in vivo recordings from mouse hippocampus, we show that NREM and REM sleep can play differential roles in memory consolidation. The specific neurophysiological features of the two sleep states allow for expansion of memory traces (during NREM) and prevention of overlap between different memory traces (during REM). These features are likely essential in the context of storing more than one new memory simultaneously within a brain network.
Collapse
|
21
|
Zhao M, Gu H, Pan W, Liu P, Zhu T, Shang H, Jia M, Yang J. SynCAM1 deficiency in the hippocampal parvalbumin interneurons contributes to sevoflurane-induced cognitive impairment in neonatal rats. CNS Neurosci Ther 2024; 30:e14554. [PMID: 38105652 PMCID: PMC10805405 DOI: 10.1111/cns.14554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
AIMS Sevoflurane is widely used for general anesthesia in children. Previous studies reported that multiple neonatal exposures to sevoflurane can induce long-term cognitive impairment in adolescent rats, but the underlying mechanisms were not defined. METHODS Postnatal day 6 (P6) to P8 rat pups were exposed to 30% oxygen with or without 3% sevoflurane balanced with air. The Y maze test (YMT) and Morris water maze (MWM) tests were performed in some cohorts from age P35 to assess cognitive functions, and their brain samples were harvested at age P14, 21, 28, 35, and 42 for measurements of various molecular entities and in vivo electrophysiology experiments at age P35. RESULTS Sevoflurane exposure resulted in cognitive impairment that was associated with decreased synCAM1 expression in parvalbumin (PV) interneurons, a reduction of PV phenotype, disturbed gamma oscillations, and dendritic spine loss in the hippocampal CA3 region. Enriched environment (EE) increased synCAM1 expression in the PV interneurons and attenuated sevoflurane-induced cognitive impairment. The synCAM1 overexpression by the adeno-associated virus vector in the hippocampal CA3 region restored sevoflurane-induced cognitive impairment, PV phenotype loss, gamma oscillations decrease, and dendritic spine loss. CONCLUSION Our data suggested that neonatal sevoflurane exposure results in cognitive impairment through decreased synCAM1 expression in PV interneurons in the hippocampus.
Collapse
Affiliation(s)
- Ming‐ming Zhao
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Han‐wen Gu
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Wei‐tong Pan
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Pan‐miao Liu
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Ting‐ting Zhu
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Hui‐jie Shang
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Min Jia
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jian‐jun Yang
- Department of Anesthesiology, Pain and Perioperative MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Neuroscience Research Institute, Zhengzhou University Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
22
|
Hijazi S, Smit AB, van Kesteren RE. Fast-spiking parvalbumin-positive interneurons in brain physiology and Alzheimer's disease. Mol Psychiatry 2023; 28:4954-4967. [PMID: 37419975 PMCID: PMC11041664 DOI: 10.1038/s41380-023-02168-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023]
Abstract
Fast-spiking parvalbumin (PV) interneurons are inhibitory interneurons with unique morphological and functional properties that allow them to precisely control local circuitry, brain networks and memory processing. Since the discovery in 1987 that PV is expressed in a subset of fast-spiking GABAergic inhibitory neurons, our knowledge of the complex molecular and physiological properties of these cells has been expanding. In this review, we highlight the specific properties of PV neurons that allow them to fire at high frequency and with high reliability, enabling them to control network oscillations and shape the encoding, consolidation and retrieval of memories. We next discuss multiple studies reporting PV neuron impairment as a critical step in neuronal network dysfunction and cognitive decline in mouse models of Alzheimer's disease (AD). Finally, we propose potential mechanisms underlying PV neuron dysfunction in AD and we argue that early changes in PV neuron activity could be a causal step in AD-associated network and memory impairment and a significant contributor to disease pathogenesis.
Collapse
Affiliation(s)
- Sara Hijazi
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Tzilivaki A, Tukker JJ, Maier N, Poirazi P, Sammons RP, Schmitz D. Hippocampal GABAergic interneurons and memory. Neuron 2023; 111:3154-3175. [PMID: 37467748 PMCID: PMC10593603 DOI: 10.1016/j.neuron.2023.06.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/04/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
One of the most captivating questions in neuroscience revolves around the brain's ability to efficiently and durably capture and store information. It must process continuous input from sensory organs while also encoding memories that can persist throughout a lifetime. What are the cellular-, subcellular-, and network-level mechanisms that underlie this remarkable capacity for long-term information storage? Furthermore, what contributions do distinct types of GABAergic interneurons make to this process? As the hippocampus plays a pivotal role in memory, our review focuses on three aspects: (1) delineation of hippocampal interneuron types and their connectivity, (2) interneuron plasticity, and (3) activity patterns of interneurons during memory-related rhythms, including the role of long-range interneurons and disinhibition. We explore how these three elements, together showcasing the remarkable diversity of inhibitory circuits, shape the processing of memories in the hippocampus.
Collapse
Affiliation(s)
- Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany; NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany
| | - John J Tukker
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Nikolaus Maier
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
| | - Panayiota Poirazi
- Foundation for Research and Technology Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), N. Plastira 100, Heraklion, Crete, Greece
| | - Rosanna P Sammons
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany; NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany; Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Philippstrasse. 13, 10115 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany.
| |
Collapse
|
24
|
Yamada J, Maeda S, Tojo M, Hayashida M, Iinuma KM, Jinno S. Altered regulation of oligodendrocytes associated with parvalbumin neurons in the ventral hippocampus underlies fear generalization in male mice. Neuropsychopharmacology 2023; 48:1668-1679. [PMID: 37277574 PMCID: PMC10516901 DOI: 10.1038/s41386-023-01611-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/07/2023]
Abstract
Fear generalization is a neurobiological process by which an organism interprets a novel stimulus as threatening because of its similarity to previously learned fear-inducing stimuli. Because recent studies have suggested that the communication between oligodendrocyte precursor cells (OPCs) and parvalbumin (PV)-expressing GABAergic neurons (PV neurons) may play critical roles in stress-related disorders, we examined the involvement of these cells in fear generalization. We first tested the behavioral characteristics of mouse models for conventional fear conditioning (cFC) and modified FC (mFC) with severe electric foot shocks and found that fear generalization was observed in mice treated with mFC but not in mice treated with cFC. The expression levels of genes related to OPCs, oligodendrocytes (OLs), and myelin in the ventral hippocampus were lower in mFC mice than in cFC mice. The densities of OPCs and OLs were decreased in the ventral hippocampus of mFC mice compared to cFC mice. The myelination ratios of PV neurons in the ventral hippocampus were lower in mFC mice than in cFC mice. The chemogenetic activation of PV neurons in the ventral hippocampus of mFC mice reduced fear generalization. The expression levels of genes related to OPCs, OLs, and myelin were recovered following the activation of PV neurons. Finally, the myelination ratios of PV neurons were increased after the activation of PV neurons. Our results suggest that altered regulation of OLs specifically associated with axons of PV neurons in the ventral hippocampus may underlie the generalization of remote fear memory following severe stress exposure.
Collapse
Affiliation(s)
- Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Miori Tojo
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Miyuki Hayashida
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
25
|
Çalışkan G, Demiray YE, Stork O. Comparison of three common inbred mouse strains reveals substantial differences in hippocampal GABAergic interneuron populations and in vitro network oscillations. Eur J Neurosci 2023; 58:3383-3401. [PMID: 37550182 DOI: 10.1111/ejn.16112] [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: 11/15/2022] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 08/09/2023]
Abstract
A major challenge in neuroscience is to pinpoint neurobiological correlates of specific cognitive and neuropsychiatric traits. At the mesoscopic level, promising candidates for establishing such connections are brain oscillations that can be robustly recorded as local field potentials with varying frequencies in the hippocampus in vivo and in vitro. Inbred mouse strains show natural variation in hippocampal synaptic plasticity (e.g. long-term potentiation), a cellular correlate of learning and memory. However, their diversity in expression of different types of hippocampal network oscillations has not been fully explored. Here, we investigated hippocampal network oscillations in three widely used inbred mouse strains: C57BL/6J (B6J), C57BL/6NCrl (B6N) and 129S2/SvPasCrl (129) with the aim to identify common oscillatory characteristics in inbred mouse strains that show aberrant emotional/cognitive behaviour (B6N and 129) and compare them to "control" B6J strain. First, we detected higher gamma oscillation power in the hippocampal CA3 of both B6N and 129 strains. Second, higher incidence of hippocampal sharp wave-ripple (SPW-R) transients was evident in these strains. Third, we observed prominent differences in the densities of distinct interneuron types and CA3 associative network activity, which are indispensable for sustainment of mesoscopic network oscillations. Together, these results add further evidence to profound physiological differences among inbred mouse strains commonly used in neuroscience research.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Research Group "Synapto-Oscillopathies", Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Yunus E Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying MentalHealth (C-I-R-C), Jena-Magdeburg-Halle, Germany
- German Center for Mental Health (DZPG), Site Jena-Magdeburg-Halle, Jena-Magdeburg-Halle, Germany
| |
Collapse
|
26
|
Etter G, Carmichael JE, Williams S. Linking temporal coordination of hippocampal activity to memory function. Front Cell Neurosci 2023; 17:1233849. [PMID: 37720546 PMCID: PMC10501408 DOI: 10.3389/fncel.2023.1233849] [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: 06/02/2023] [Accepted: 08/01/2023] [Indexed: 09/19/2023] Open
Abstract
Oscillations in neural activity are widespread throughout the brain and can be observed at the population level through the local field potential. These rhythmic patterns are associated with cycles of excitability and are thought to coordinate networks of neurons, in turn facilitating effective communication both within local circuits and across brain regions. In the hippocampus, theta rhythms (4-12 Hz) could contribute to several key physiological mechanisms including long-range synchrony, plasticity, and at the behavioral scale, support memory encoding and retrieval. While neurons in the hippocampus appear to be temporally coordinated by theta oscillations, they also tend to fire in sequences that are developmentally preconfigured. Although loss of theta rhythmicity impairs memory, these sequences of spatiotemporal representations persist in conditions of altered hippocampal oscillations. The focus of this review is to disentangle the relative contribution of hippocampal oscillations from single-neuron activity in learning and memory. We first review cellular, anatomical, and physiological mechanisms underlying the generation and maintenance of hippocampal rhythms and how they contribute to memory function. We propose candidate hypotheses for how septohippocampal oscillations could support memory function while not contributing directly to hippocampal sequences. In particular, we explore how theta rhythms could coordinate the integration of upstream signals in the hippocampus to form future decisions, the relevance of such integration to downstream regions, as well as setting the stage for behavioral timescale synaptic plasticity. Finally, we leverage stimulation-based treatment in Alzheimer's disease conditions as an opportunity to assess the sufficiency of hippocampal oscillations for memory function.
Collapse
Affiliation(s)
| | | | - Sylvain Williams
- Department of Psychiatry, Douglas Mental Health Research Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Martinez JD, Wilson LG, Brancaleone WP, Peterson KG, Popke DS, Garzon VC, Perez Tremble RE, Donnelly MJ, Mendez Ortega SL, Torres D, Shaver JJ, Clawson BC, Jiang S, Yang Z, Aton SJ. Hypnotic treatment reverses NREM sleep disruption and EEG desynchronization in a mouse model of Fragile X syndrome to rescue memory consolidation deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549070. [PMID: 37502832 PMCID: PMC10370139 DOI: 10.1101/2023.07.14.549070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Fragile X syndrome (FXS) is a highly-prevalent genetic cause of intellectual disability, associated with disrupted cognition and sleep abnormalities. Sleep loss itself negatively impacts cognitive function, yet the contribution of sleep loss to impaired cognition in FXS is vastly understudied. One untested possibility is that disrupted cognition in FXS is exacerbated by abnormal sleep. We hypothesized that restoration of sleep-dependent mechanisms could improve functions such as memory consolidation in FXS. We examined whether administration of ML297, a hypnotic drug acting on G-protein-activated inward-rectifying potassium channels, could restore sleep phenotypes and improve disrupted memory consolidation in Fmr1 -/y mice. Using 24-h polysomnographic recordings, we found that Fmr1 -/y mice exhibit reduced non-rapid eye movement (NREM) sleep and fragmented NREM sleep architecture, alterations in NREM EEG spectral power (including reductions in sleep spindles), and reduced EEG coherence between cortical areas. These alterations were reversed in the hours following ML297 administration. Hypnotic treatment following contextual fear or spatial learning also ameliorated disrupted memory consolidation in Fmr1 -/y mice. Hippocampal activation patterns during memory recall was altered in Fmr1 -/y mice, reflecting an altered balance of activity among principal neurons vs. parvalbumin-expressing (PV+) interneurons. This phenotype was partially reversed by post-learning ML297 administration. These studies suggest that sleep disruption could have a major impact on neurophysiological and behavioral phenotypes in FXS, and that hypnotic therapy may significantly improve disrupted cognition in this disorder.
Collapse
|
28
|
Sierra RO, Pedraza LK, Barcsai L, Pejin A, Li Q, Kozák G, Takeuchi Y, Nagy AJ, Lőrincz ML, Devinsky O, Buzsáki G, Berényi A. Closed-loop brain stimulation augments fear extinction in male rats. Nat Commun 2023; 14:3972. [PMID: 37407557 DOI: 10.1038/s41467-023-39546-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 06/16/2023] [Indexed: 07/07/2023] Open
Abstract
Dysregulated fear reactions can result from maladaptive processing of trauma-related memories. In post-traumatic stress disorder (PTSD) and other psychiatric disorders, dysfunctional extinction learning prevents discretization of trauma-related memory engrams and generalizes fear responses. Although PTSD may be viewed as a memory-based disorder, no approved treatments target pathological fear memory processing. Hippocampal sharp wave-ripples (SWRs) and concurrent neocortical oscillations are scaffolds to consolidate contextual memory, but their role during fear processing remains poorly understood. Here, we show that closed-loop, SWR triggered neuromodulation of the medial forebrain bundle (MFB) can enhance fear extinction consolidation in male rats. The modified fear memories became resistant to induced recall (i.e., 'renewal' and 'reinstatement') and did not reemerge spontaneously. These effects were mediated by D2 receptor signaling-induced synaptic remodeling in the basolateral amygdala. Our results demonstrate that SWR-triggered closed-loop stimulation of the MFB reward system enhances extinction of fearful memories and reducing fear expression across different contexts and preventing excessive and persistent fear responses. These findings highlight the potential of neuromodulation to augment extinction learning and provide a new avenue to develop treatments for anxiety disorders.
Collapse
Affiliation(s)
- Rodrigo Ordoñez Sierra
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Lizeth Katherine Pedraza
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Lívia Barcsai
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary
- Neunos Inc, Boston, MA, 02108, USA
| | - Andrea Pejin
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary
- Neunos Inc, Boston, MA, 02108, USA
| | - Qun Li
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Gábor Kozák
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
| | - Yuichi Takeuchi
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- Department of Biopharmaceutical Sciences and Pharmacy, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Anett J Nagy
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary
- Neunos Inc, Boston, MA, 02108, USA
| | - Magor L Lőrincz
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary
- Department of Physiology, Anatomy and Neuroscience, Faculty of Sciences University of Szeged, Szeged, 6726, Hungary
- Neuroscience Division, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Orrin Devinsky
- Department of Neurology, NYU Langone Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - György Buzsáki
- Neuroscience Institute, New York University, New York, NY, 10016, USA
- Center for Neural Science, New York University, New York, NY, 10016, USA
| | - Antal Berényi
- MTA-SZTE 'Momentum' Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged, 6720, Hungary.
- HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged, 6720, Hungary.
- Neunos Inc, Boston, MA, 02108, USA.
- Neuroscience Institute, New York University, New York, NY, 10016, USA.
| |
Collapse
|
29
|
Wyroślak M, Dobrzański G, Mozrzymas JW. Bidirectional plasticity of GABAergic tonic inhibition in hippocampal somatostatin- and parvalbumin-containing interneurons. Front Cell Neurosci 2023; 17:1193383. [PMID: 37448697 PMCID: PMC10336215 DOI: 10.3389/fncel.2023.1193383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
GABAA receptors present in extrasynaptic areas mediate tonic inhibition in hippocampal neurons regulating the performance of neural networks. In this study, we investigated the effect of NMDA-induced plasticity on tonic inhibition in somatostatin- and parvalbumin-containing interneurons. Using pharmacological methods and transgenic mice (SST-Cre/PV-Cre x Ai14), we induced the plasticity of GABAergic transmission in somatostatin- and parvalbumin-containing interneurons by a brief (3 min) application of NMDA. In the whole-cell patch-clamp configuration, we measured tonic currents enhanced by specific agonists (etomidate or gaboxadol). Furthermore, in both the control and NMDA-treated groups, we examined to what extent these changes depend on the regulation of distinct subtypes of GABAA receptors. Tonic conductance in the somatostatin-containing (SST+) interneurons is enhanced after NMDA application, and the observed effect is associated with an increased content of α5-containing GABAARs. Both fast-spiking and non-fast-spiking parvalbumin-positive (PV+) cells showed a reduction of tonic inhibition after plasticity induction. This effect was accompanied in both PV+ interneuron types by a strongly reduced proportion of δ-subunit-containing GABAARs and a relatively small increase in currents mediated by α5-containing GABAARs. Both somatostatin- and parvalbumin-containing interneurons show cell type-dependent and opposite sign plasticity of tonic inhibition. The underlying mechanisms depend on the cell-specific balance of plastic changes in the contents of α5 and δ subunit-containing GABAARs.
Collapse
Affiliation(s)
- Marcin Wyroślak
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | | | - Jerzy W. Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
30
|
Martinez JD, Donnelly MJ, Popke DS, Torres D, Wilson LG, Brancaleone WP, Sheskey S, Lin CM, Clawson BC, Jiang S, Aton SJ. Enriched binocular experience followed by sleep optimally restores binocular visual cortical responses in a mouse model of amblyopia. Commun Biol 2023; 6:408. [PMID: 37055505 PMCID: PMC10102075 DOI: 10.1038/s42003-023-04798-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/03/2023] [Indexed: 04/15/2023] Open
Abstract
Studies of primary visual cortex have furthered our understanding of amblyopia, long-lasting visual impairment caused by imbalanced input from the two eyes during childhood, which is commonly treated by patching the dominant eye. However, the relative impacts of monocular vs. binocular visual experiences on recovery from amblyopia are unclear. Moreover, while sleep promotes visual cortex plasticity following loss of input from one eye, its role in recovering binocular visual function is unknown. Using monocular deprivation in juvenile male mice to model amblyopia, we compared recovery of cortical neurons' visual responses after identical-duration, identical-quality binocular or monocular visual experiences. We demonstrate that binocular experience is quantitatively superior in restoring binocular responses in visual cortex neurons. However, this recovery was seen only in freely-sleeping mice; post-experience sleep deprivation prevented functional recovery. Thus, both binocular visual experience and subsequent sleep help to optimally renormalize bV1 responses in a mouse model of amblyopia.
Collapse
Affiliation(s)
- Jessy D Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Marcus J Donnelly
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI, USA
| | - Donald S Popke
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Torres
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lydia G Wilson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | | | - Sarah Sheskey
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Cheng-Mao Lin
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Brittany C Clawson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
31
|
Martinez JD, Brancaleone WP, Peterson KG, Wilson LG, Aton SJ. Atypical hypnotic compound ML297 restores sleep architecture immediately following emotionally valenced learning, to promote memory consolidation and hippocampal network activation during recall. Sleep 2023; 46:zsac301. [PMID: 36510822 PMCID: PMC9995787 DOI: 10.1093/sleep/zsac301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
Sleep plays a critical role in consolidating many forms of hippocampus-dependent memory. While various classes of hypnotic drugs have been developed in recent years, it remains unknown whether, or how, some of them affect sleep-dependent memory consolidation mechanisms. We find that ML297, a recently developed candidate hypnotic agent targeting a new mechanism (activating GIRK1/2-subunit containing G-protein coupled inwardly rectifying potassium [GIRK] channels), alters sleep architecture in mice over the first 6 hr following a single-trial learning event. Following contextual fear conditioning (CFC), ML297 reversed post-CFC reductions in NREM sleep spindle power and REM sleep amounts and architecture, renormalizing sleep features to what was observed at baseline, prior to CFC. Renormalization of post-CFC REM sleep latency, REM sleep amounts, and NREM spindle power were all associated with improved contextual fear memory (CFM) consolidation. We find that improvements in CFM consolidation due to ML297 are sleep-dependent, and are associated with increased numbers of highly activated dentate gyrus (DG), CA1, and CA3 neurons during CFM recall. Together our findings suggest that GIRK1/2 channel activation restores normal sleep architecture- including REM sleep, which is normally suppressed following CFC-and increases the number of hippocampal neurons incorporated into the CFM engram during memory consolidation.
Collapse
Affiliation(s)
- Jessy D Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William P Brancaleone
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn G Peterson
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lydia G Wilson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
32
|
Shih YT, Alipio JB, Sahay A. An inhibitory circuit-based enhancer of Dyrk1a function reverses Dyrk1a -associated impairment in social recognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.526955. [PMID: 36778241 PMCID: PMC9915696 DOI: 10.1101/2023.02.03.526955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Heterozygous mutations in the Dual specificity tyrosine-phosphorylation-regulated kinase 1a Dyrk1a gene define a syndromic form of Autism Spectrum Disorder. The synaptic and circuit mechanisms mediating Dyrk1a functions in social cognition are unclear. Here, we identify a social experience-sensitive mechanism in hippocampal mossy fiber-parvalbumin interneuron (PV IN) synapses by which Dyrk1a recruits feedforward inhibition of CA3 and CA2 to promote social recognition. We employ genetic epistasis logic to identify a cytoskeletal protein, Ablim3, as a synaptic substrate of Dyrk1a. We demonstrate that Ablim3 downregulation in dentate granule cells of adult hemizygous Dyrk1a mice is sufficient to restore PV IN mediated inhibition of CA3 and CA2 and social recognition. Acute chemogenetic activation of PV INs in CA3/CA2 of adult hemizygous Dyrk1a mice also rescued social recognition. Together, these findings illustrate how targeting Dyrk1a synaptic and circuit substrates as "enhancers of Dyrk1a function" harbors potential to reverse Dyrk1a haploinsufficiency-associated circuit and cognition impairments. Highlights Dyrk1a in mossy fibers recruits PV IN mediated feed-forward inhibition of CA3 and CA2Dyrk1a-Ablim3 signaling in mossy fiber-PV IN synapses promotes inhibition of CA3 and CA2 Downregulating Ablim3 restores PV IN excitability, CA3/CA2 inhibition and social recognition in Dyrk1a+/- mice Chemogenetic activation of PV INs in CA3/CA2 rescues social recognition in Dyrk1a+/- mice.
Collapse
|
33
|
Hall AF, Wang DV. The two tales of hippocampal sharp-wave ripple content: The rigid and the plastic. Prog Neurobiol 2023; 221:102396. [PMID: 36563928 PMCID: PMC9899323 DOI: 10.1016/j.pneurobio.2022.102396] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Sharp-wave ripples, prominently in the CA1 region of the hippocampus, are short oscillatory events accompanied by bursts of neural firing. Ripples and associated hippocampal place cell sequences communicate with cortical ensembles during slow-wave sleep, which has been shown to be critical for systems consolidation of episodic memories. This consolidation is not limited to a newly formed memory trace; instead, ripples appear to reactivate and consolidate memories spanning various experiences. Despite this broad spanning influence, ripples remain capable of producing precise memories. The underlying mechanisms that enable ripples to consolidate memories broadly and with specificity across experiences remain unknown. In this review, we discuss data that uncovers circuit-level processes that generate ripples and influence their characteristics during consolidation. Based on current knowledge, we propose that memory emerges from the integration of two parallel consolidation pathways in CA1: the rigid and plastic pathways. The rigid pathway generates ripples stochastically, providing a backbone upon which dynamic plastic pathway inputs carrying novel information are integrated.
Collapse
Affiliation(s)
- Arron F Hall
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Dong V Wang
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|
34
|
Roque PS, Thörn Perez C, Hooshmandi M, Wong C, Eslamizade MJ, Heshmati S, Brown N, Sharma V, Lister KC, Goyon VM, Neagu-Lund L, Shen C, Daccache N, Sato H, Sato T, Mogil JS, Nader K, Gkogkas CG, Iordanova MD, Prager-Khoutorsky M, McBride HM, Lacaille JC, Wykes L, Schricker T, Khoutorsky A. Parvalbumin interneuron loss mediates repeated anesthesia-induced memory deficits in mice. J Clin Invest 2023; 133:159344. [PMID: 36394958 PMCID: PMC9843048 DOI: 10.1172/jci159344] [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: 02/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Repeated or prolonged, but not short-term, general anesthesia during the early postnatal period causes long-lasting impairments in memory formation in various species. The mechanisms underlying long-lasting impairment in cognitive function are poorly understood. Here, we show that repeated general anesthesia in postnatal mice induces preferential apoptosis and subsequent loss of parvalbumin-positive inhibitory interneurons in the hippocampus. Each parvalbumin interneuron controls the activity of multiple pyramidal excitatory neurons, thereby regulating neuronal circuits and memory consolidation. Preventing the loss of parvalbumin neurons by deleting a proapoptotic protein, mitochondrial anchored protein ligase (MAPL), selectively in parvalbumin neurons rescued anesthesia-induced deficits in pyramidal cell inhibition and hippocampus-dependent long-term memory. Conversely, partial depletion of parvalbumin neurons in neonates was sufficient to engender long-lasting memory impairment. Thus, loss of parvalbumin interneurons in postnatal mice following repeated general anesthesia critically contributes to memory deficits in adulthood.
Collapse
Affiliation(s)
- Patricia Soriano Roque
- Department of Anesthesia and,School of Human Nutrition, McGill University, Montreal, Canada
| | | | | | | | - Mohammad Javad Eslamizade
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, Canada.,Department of Biochemistry, McGill University, Montreal, Canada.,Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Vijendra Sharma
- Department of Biochemistry, McGill University, Montreal, Canada
| | | | | | | | | | | | | | | | - Jeffrey S. Mogil
- Department of Anesthesia and,Department of Psychology, Faculty of Science, and,Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Karim Nader
- Department of Psychology, Faculty of Science, and
| | - Christos G. Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology–Hellas, University Campus, Ioannina, Greece
| | - Mihaela D. Iordanova
- Department of Psychology/Centre for Studies in Behavioural Neurobiology, Concordia University, Montreal, Canada
| | | | | | - Jean-Claude Lacaille
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, Canada
| | - Linda Wykes
- School of Human Nutrition, McGill University, Montreal, Canada
| | | | - Arkady Khoutorsky
- Department of Anesthesia and,Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada.,Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| |
Collapse
|
35
|
Černotová D, Hrůzová K, Levčík D, Svoboda J, Stuchlík A. Linking Social Cognition, Parvalbumin Interneurons, and Oxytocin in Alzheimer's Disease: An Update. J Alzheimers Dis 2023; 96:861-875. [PMID: 37980658 PMCID: PMC10741376 DOI: 10.3233/jad-230333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 11/21/2023]
Abstract
Finding a cure for Alzheimer's disease (AD) has been notoriously challenging for many decades. Therefore, the current focus is mainly on prevention, timely intervention, and slowing the progression in the earliest stages. A better understanding of underlying mechanisms at the beginning of the disease could aid in early diagnosis and intervention, including alleviating symptoms or slowing down the disease progression. Changes in social cognition and progressive parvalbumin (PV) interneuron dysfunction are among the earliest observable effects of AD. Various AD rodent models mimic these early alterations, but only a narrow field of study has considered their mutual relationship. In this review, we discuss current knowledge about PV interneuron dysfunction in AD and emphasize their importance in social cognition and memory. Next, we propose oxytocin (OT) as a potent modulator of PV interneurons and as a promising treatment for managing some of the early symptoms. We further discuss the supporting evidence on its beneficial effects on AD-related pathology. Clinical trials have employed the use of OT in various neuropsychiatric diseases with promising results, but little is known about its prospective impacts on AD. On the other hand, the modulatory effects of OT in specific structures and local circuits need to be clarified in future studies. This review highlights the connection between PV interneurons and social cognition impairment in the early stages of AD and considers OT as a promising therapeutic agent for addressing these early deficits.
Collapse
Affiliation(s)
- Daniela Černotová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karolína Hrůzová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - David Levčík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Svoboda
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Stuchlík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
36
|
Singh S, Topolnik L. Inhibitory circuits in fear memory and fear-related disorders. Front Neural Circuits 2023; 17:1122314. [PMID: 37035504 PMCID: PMC10076544 DOI: 10.3389/fncir.2023.1122314] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/17/2023] [Indexed: 04/11/2023] Open
Abstract
Fear learning and memory rely on dynamic interactions between the excitatory and inhibitory neuronal populations that make up the prefrontal cortical, amygdala, and hippocampal circuits. Whereas inhibition of excitatory principal cells (PCs) by GABAergic neurons restrains their excitation, inhibition of GABAergic neurons promotes the excitation of PCs through a process called disinhibition. Specifically, GABAergic interneurons that express parvalbumin (PV+) and somatostatin (SOM+) provide inhibition to different subcellular domains of PCs, whereas those that express the vasoactive intestinal polypeptide (VIP+) facilitate disinhibition of PCs by inhibiting PV+ and SOM+ interneurons. Importantly, although the main connectivity motifs and the underlying network functions of PV+, SOM+, and VIP+ interneurons are replicated across cortical and limbic areas, these inhibitory populations play region-specific roles in fear learning and memory. Here, we provide an overview of the fear processing in the amygdala, hippocampus, and prefrontal cortex based on the evidence obtained in human and animal studies. Moreover, focusing on recent findings obtained using genetically defined imaging and intervention strategies, we discuss the population-specific functions of PV+, SOM+, and VIP+ interneurons in fear circuits. Last, we review current insights that integrate the region-specific inhibitory and disinhibitory network patterns into fear memory acquisition and fear-related disorders.
Collapse
Affiliation(s)
- Sanjay Singh
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Quebec City, QC, Canada
- Neuroscience Axis, CRCHUQ, Laval University, Quebec City, QC, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Quebec City, QC, Canada
- Neuroscience Axis, CRCHUQ, Laval University, Quebec City, QC, Canada
- *Correspondence: Lisa Topolnik
| |
Collapse
|
37
|
Spahic H, Parmar P, Miller S, Emerson PC, Lechner C, St. Pierre M, Rastogi N, Nugent M, Duck SA, Kirkwood A, Chavez-Valdez R. Dysregulation of ErbB4 Signaling Pathway in the Dorsal Hippocampus after Neonatal Hypoxia-Ischemia and Late Deficits in PV + Interneurons, Synaptic Plasticity and Working Memory. Int J Mol Sci 2022; 24:ijms24010508. [PMID: 36613949 PMCID: PMC9820818 DOI: 10.3390/ijms24010508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal hypoxic-ischemic (HI) injury leads to deficits in hippocampal parvalbumin (PV)+ interneurons (INs) and working memory. Therapeutic hypothermia (TH) does not prevent these deficits. ErbB4 supports maturation and maintenance of PV+ IN. Thus, we hypothesized that neonatal HI leads to persistent deficits in PV+ INs, working memory and synaptic plasticity associated with ErbB4 dysregulation despite TH. P10 HI-injured mice were randomized to normothermia (NT, 36 °C) or TH (31 °C) for 4 h and compared to sham. Hippocampi were studied for α-fodrin, glial fibrillary acidic protein (GFAP), and neuroregulin (Nrg) 1 levels; erb-b2 receptor tyrosine kinase 4 (ErbB4)/ Ak strain transforming (Akt) activation; and PV, synaptotagmin (Syt) 2, vesicular-glutamate transporter (VGlut) 2, Nrg1, and ErbB4 expression in coronal sections. Extracellular field potentials and behavioral testing were performed. At P40, deficits in PV+ INs correlated with impaired memory and coincided with blunted long-term depression (LTD), heightened long-term potentiation (LTP) and increased Vglut2/Syt2 ratio, supporting excitatory-inhibitory (E/I) imbalance. Hippocampal Nrg1 levels were increased in the hippocampus 24 h after neonatal HI, delaying the decline documented in shams. Paradoxically ErbB4 activation decreased 24 h and again 30 days after HI. Neonatal HI leads to persistent deficits in hippocampal PV+ INs, memory, and synaptic plasticity. While acute decreased ErbB4 activation supports impaired maturation and survival after HI, late deficit reemergence may impair PV+ INs maintenance after HI.
Collapse
Affiliation(s)
- Harisa Spahic
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Pritika Parmar
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sarah Miller
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul Casey Emerson
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Charles Lechner
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mark St. Pierre
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Neetika Rastogi
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Nugent
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah Ann Duck
- Department of Molecular and Cellular Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Alfredo Kirkwood
- Mind-Brain Institute, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Raul Chavez-Valdez
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Correspondence:
| |
Collapse
|
38
|
Banerjee T, Pati S, Tiwari P, Vaidya VA. Chronic hM3Dq-DREADD-mediated chemogenetic activation of parvalbumin-positive inhibitory interneurons in postnatal life alters anxiety and despair-like behavior in adulthood in a task- and sex-dependent manner. J Biosci 2022. [DOI: 10.1007/s12038-022-00308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Frank SM, Becker M, Qi A, Geiger P, Frank UI, Rosedahl LA, Malloni WM, Sasaki Y, Greenlee MW, Watanabe T. Efficient learning in children with rapid GABA boosting during and after training. Curr Biol 2022; 32:5022-5030.e7. [PMID: 36384138 DOI: 10.1016/j.cub.2022.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/10/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022]
Abstract
It is generally thought that children learn more efficiently than adults. One way to accomplish this is to have learning rapidly stabilized such that it is not interfered with by subsequent learning. Although γ-aminobutyric acid (GABA) plays an important role in stabilization, it has been reported that GABAergic inhibitory processing is not fully matured yet in children compared with adults. Does this finding indicate that more efficient learning in children is not due to more rapid stabilization? Here, we measured the concentration of GABA in early visual cortical areas in a time-resolved fashion before, during, and after visual perceptual learning (VPL) within subjects using functional MRS (fMRS) and then compared the concentrations between children (8 to 11 years old) and adults (18 to 35 years old). We found that children exhibited a rapid boost of GABA during visual training that persisted after training ended, whereas the concentration of GABA in adults remained unchanged. Moreover, behavioral experiments showed that children exhibited rapid development of resilience to retrograde interference, which indicates that children stabilize VPL much faster than adults. These results together suggest that inhibitory processing in children's brains is more dynamic and adapts more quickly to stabilize learning than in adults, making learning more efficient in children.
Collapse
Affiliation(s)
- Sebastian M Frank
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany; Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA.
| | - Markus Becker
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Andrea Qi
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA
| | - Patricia Geiger
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Ulrike I Frank
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Luke A Rosedahl
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA
| | - Wilhelm M Malloni
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Yuka Sasaki
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA
| | - Mark W Greenlee
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Takeo Watanabe
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA.
| |
Collapse
|
40
|
Miranda JM, Cruz E, Bessières B, Alberini CM. Hippocampal parvalbumin interneurons play a critical role in memory development. Cell Rep 2022; 41:111643. [PMID: 36384113 PMCID: PMC9737056 DOI: 10.1016/j.celrep.2022.111643] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/16/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
Abstract
Episodic memories formed in early childhood rapidly decay, but their latent traces remain stored long term. These memories require the dorsal hippocampus (dHPC) and seem to undergo a developmental critical period. It remains to be determined whether the maturation of parvalbumin interneurons (PVIs), a major mechanism of critical periods, contributes to memory development. Here, we show that episodic infantile learning significantly increases the levels of parvalbumin in the dHPC 48 h after training. Chemogenetic inhibition of PVIs before learning indicated that these neurons are required for infantile memory formation. A bilateral dHPC injection of the γ-aminobutyric acid type A receptor agonist diazepam after training elicited long-term memory expression in infant rats, although direct PVI chemogenetic activation had no effect. Finally, PVI activity was required for brain-derived neurotrophic factor (BDNF)-dependent maturation of memory competence, i.e., adult-like long-term memory expression. Thus, dHPC PVIs are critical for the formation of infantile memories and for memory development.
Collapse
Affiliation(s)
- Janelle M Miranda
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Emmanuel Cruz
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Benjamin Bessières
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Cristina M Alberini
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
41
|
Sleep and wake cycles dynamically modulate hippocampal inhibitory synaptic plasticity. PLoS Biol 2022; 20:e3001812. [PMID: 36318572 PMCID: PMC9624398 DOI: 10.1371/journal.pbio.3001812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/30/2022] [Indexed: 01/01/2023] Open
Abstract
Sleep is an essential process that consolidates memories by modulating synapses through poorly understood mechanisms. Here, we report that GABAergic synapses in hippocampal CA1 pyramidal neurons undergo daily rhythmic alterations. Specifically, wake inhibits phasic inhibition, whereas it promotes tonic inhibition compared to sleep. We further utilize a model of chemically induced inhibitory long-term potentiation (iLTP) to examine inhibitory plasticity. Intriguingly, while CA1 pyramidal neurons in both wake and sleep mice undergo iLTP, wake mice have a much higher magnitude. We also employ optogenetics and observe that inhibitory inputs from parvalbumin-, but not somatostatin-, expressing interneurons contribute to dynamic iLTP during sleep and wake. Finally, we demonstrate that synaptic insertion of α5-GABAA receptors underlies the wake-specific enhancement of iLTP at parvalbumin-synapses, which is independent of time of the day. These data reveal a previously unappreciated daily oscillation of inhibitory LTP in hippocampal neurons and uncover a dynamic contribution of inhibitory synapses in memory mechanisms across sleep and wake.
Collapse
|
42
|
Khlaifia A, Honoré E, Artinian J, Laplante I, Lacaille JC. mTORC1 function in hippocampal parvalbumin interneurons: regulation of firing and long-term potentiation of intrinsic excitability but not long-term contextual fear memory and context discrimination. Mol Brain 2022; 15:56. [PMID: 35715811 PMCID: PMC9204956 DOI: 10.1186/s13041-022-00941-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/07/2022] [Indexed: 02/03/2023] Open
Abstract
Hippocampal CA1 parvalbumin-expressing interneurons (PV INs) play a central role in controlling principal cell activity and orchestrating network oscillations. PV INs receive excitatory inputs from CA3 Schaffer collaterals and local CA1 pyramidal cells, and they provide perisomatic inhibition. Schaffer collateral excitatory synapses onto PV INs express Hebbian and anti-Hebbian types of long-term potentiation (LTP), as well as elicit LTP of intrinsic excitability (LTPIE). LTPIE requires the activation of type 5 metabotropic glutamate receptors (mGluR5) and is mediated by downregulation of potassium channels Kv1.1. It is sensitive to rapamycin and thus may involve activation of the mammalian target of rapamycin complex 1 (mTORC1). LTPIE facilitates PV INs recruitment in CA1 and maintains an excitatory-inhibitory balance. Impaired CA1 PV INs activity or LTP affects network oscillations and memory. However, whether LTPIE in PV INs plays a role in hippocampus-dependent memory remains unknown. Here, we used conditional deletion of the obligatory component of mTORC1, the Regulatory-Associated Protein of mTOR (Raptor), to directly manipulate mTORC1 in PV INs. We found that homozygous, but not heterozygous, conditional knock-out of Rptor resulted in a decrease in CA1 PV INs of mTORC1 signaling via its downstream effector S6 phosphorylation assessed by immunofluorescence. In whole-cell recordings from hippocampal slices, repetitive firing of CA1 PV INs was impaired in mice with either homozygous or heterozygous conditional knock-out of Rptor. High frequency stimulation of Schaffer collateral inputs that induce LTPIE in PV INs of control mice failed to do so in mice with either heterozygous or homozygous conditional knock-out of Rptor in PV INs. At the behavioral level, mice with homozygous or heterozygous conditional knock-out of Rptor showed similar long-term contextual fear memory or contextual fear memory discrimination relative to control mice. Thus, mTORC1 activity in CA1 PV INs regulates repetitive firing and LTPIE but not consolidation of long-term contextual fear memory and context discrimination. Our results indicate that mTORC1 plays cell-specific roles in synaptic plasticity of hippocampal inhibitory interneurons that are differentially involved in hippocampus-dependent learning and memory.
Collapse
Affiliation(s)
- Abdessattar Khlaifia
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group On Neural Signaling and Circuitry (GRSNC), Université de Montréal, P.O. Box 6128, Station Downtown, QC, H3C 3J7, Montreal, Canada.,Department of Psychology, University of Toronto Scarborough, ON, M1C1A4, Toronto, Canada
| | - Eve Honoré
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group On Neural Signaling and Circuitry (GRSNC), Université de Montréal, P.O. Box 6128, Station Downtown, QC, H3C 3J7, Montreal, Canada
| | - Julien Artinian
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group On Neural Signaling and Circuitry (GRSNC), Université de Montréal, P.O. Box 6128, Station Downtown, QC, H3C 3J7, Montreal, Canada.,NeuroService, Neurocentre Magendie , Bordeaux, France
| | - Isabel Laplante
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group On Neural Signaling and Circuitry (GRSNC), Université de Montréal, P.O. Box 6128, Station Downtown, QC, H3C 3J7, Montreal, Canada
| | - Jean-Claude Lacaille
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group On Neural Signaling and Circuitry (GRSNC), Université de Montréal, P.O. Box 6128, Station Downtown, QC, H3C 3J7, Montreal, Canada.
| |
Collapse
|
43
|
Xu Y, Wang ML, Tao H, Geng C, Guo F, Hu B, Wang R, Hou XY. ErbB4 in parvalbumin-positive interneurons mediates proactive interference in olfactory associative reversal learning. Neuropsychopharmacology 2022; 47:1292-1303. [PMID: 34707248 PMCID: PMC9117204 DOI: 10.1038/s41386-021-01205-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 09/04/2021] [Accepted: 10/02/2021] [Indexed: 11/09/2022]
Abstract
Consolidated memories influence later learning and cognitive processes when new information is overlapped with previous events. To reveal which cellular and molecular factors are associated with this proactive interference, we challenged mice with odor-reward associative learning followed by a reversal-learning task. The results showed that genetical ablation of ErbB4 in parvalbumin (PV)-positive interneurons improved performance in reversal-learning phase, with no alteration in learning phase, supporting that PV interneuron ErbB4 is required for proactive interference. Mechanistically, olfactory learning promoted PV interneuron excitatory synaptic plasticity and direct binding of ErbB4 with presynaptic Neurexin1β (NRXN1β) and postsynaptic scaffold PSD-95 in the prefrontal cortex. Interrupting ErbB4-NRXN1β interaction impaired network activity-driven excitatory inputs and excitatory synaptic transmission onto PV interneurons. Neuronal activity-induced ErbB4-PSD-95 association facilitated transsynaptic binding of ErbB4-NRXN1β and excitatory synapse formation in ErbB4-positive interneurons. Furthermore, ErbB4-NRXN1β binding was responsible for the activity-regulated activation of ErbB4 and extracellular signal-regulated kinase (ERK) 1/2 in PV interneurons, as well as synaptic plasticity-related expression of brain-derived neurotrophic factor (BDNF). Correlatedly, blocking ErbB4-NRXN1β coupling in the medial prefrontal cortex of adult mice facilitated reversal learning of an olfactory associative task. These findings provide novel insight into the physiological role of PV interneuron ErbB4 signaling in cognitive processes and reveal an associative learning-related transsynaptic NRXN1β-ErbB4-PSD-95 complex that affects the ERK1/2-BDNF pathway and underlies local inhibitory circuit plasticity and proactive interference.
Collapse
Affiliation(s)
- Yan Xu
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Meng-Lin Wang
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Hui Tao
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China ,grid.254147.10000 0000 9776 7793State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198 China
| | - Chi Geng
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Feng Guo
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Bin Hu
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Ran Wang
- grid.417303.20000 0000 9927 0537Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004 China
| | - Xiao-Yu Hou
- Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China. .,State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
44
|
The role of inhibitory circuits in hippocampal memory processing. Nat Rev Neurosci 2022; 23:476-492. [DOI: 10.1038/s41583-022-00599-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 11/08/2022]
|
45
|
Li Y, Ji M, Yang J. Current Understanding of Long-Term Cognitive Impairment After Sepsis. Front Immunol 2022; 13:855006. [PMID: 35603184 PMCID: PMC9120941 DOI: 10.3389/fimmu.2022.855006] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is recognized as a life-threatening multi-organ dysfunction resulting from a dysregulated host response to infection. Although the incidence and mortality of sepsis decrease significantly due to timely implementation of anti-infective and support therapies, accumulating evidence suggests that a great proportion of survivors suffer from long-term cognitive impairment after hospital discharge, leading to decreased life quality and substantial caregiving burdens for family members. Several mechanisms have been proposed for long-term cognitive impairment after sepsis, which are not mutually exclusive, including blood-brain barrier disruption, neuroinflammation, neurotransmitter dysfunction, and neuronal loss. Targeting these critical processes might be effective in preventing and treating long-term cognitive impairment. However, future in-depth studies are required to facilitate preventive and/or treatment strategies for long-term cognitive impairment after sepsis.
Collapse
Affiliation(s)
- Ying Li
- Department of Anesthesiology, Jiangyin Hospital, Affiliated to Southeast University Medical School, Jiangyin, China
| | - Muhuo Ji
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
46
|
Potrebić M, Pavković Ž, Puškaš N, Pešić V. The Influence of Social Isolation on Social Orientation, Sociability, Social Novelty Preference, and Hippocampal Parvalbumin-Expressing Interneurons in Peripubertal Rats - Understanding the Importance of Meeting Social Needs in Adolescence. Front Behav Neurosci 2022; 16:872628. [PMID: 35592640 PMCID: PMC9113078 DOI: 10.3389/fnbeh.2022.872628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
The fulfillment of belonging needs underlies a variety of behaviors. In order to understand how social needs unmet during maturation shape everyday life, we examined social motivation and cognition in peripubertal rats, as a rodent model of adolescence, subjected to social isolation (SI) during early and early-to-mid adolescence. The behavioral correlates of social orientation (social space preference), sociability (preference for social over non-social novelty), and social novelty preference (SNP) were examined in group-housed (GH) and single-housed (SH) rats in a 3-chamber test. The response to social odors was examined to gain insights into the developmental role of social odors in motivated social behavior. Differentiation between appetitive (number of visits/approaches) and consummatory (exploratory time) aspects of motivated social behavior was done to determine which facet of social motivation characterizes maturation when social needs are met and which aspect dominates when social needs are unsatisfied. The SI-sensitive parvalbumin-expressing interneurons (PVI) in the hippocampus were examined using immunohistochemistry. The main findings are the following: (1) in GH rats, the preference for social space is not evident regardless of animals' age, while sociability becomes apparent in mid-adolescence strictly through consummatory behavior, along with complete SNP (appetitive, consummatory); (2) SH promotes staying in a social chamber/space regardless of animals' age and produces an appetitive preference for it only in early-adolescent animals; (3) SH promotes sociability (appetitive, consummatory) regardless of the animals' age and prevents the SNP; (4) the preference for a social odor is displayed in all the groups through consummatory behavior, while appetitive behavior is evident only in SH rats; (5) the response to social odors does not commensurate directly to the response to conspecifics; (6) SH does not influence PVI in the hippocampus, except in the case of early-adolescence when a transient decrease in the dentate gyrus is observed. These results accentuate the developmental complexity of social motivation and cognition, and the power of SI in adolescence to infringe social maturation at different functional levels, promoting appetitive behavior toward peers overall but harming the interest for social novelty. The findings emphasize the importance of the fulfillment of basic social needs in the navigation through the social world.
Collapse
Affiliation(s)
- Milica Potrebić
- Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Željko Pavković
- Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nela Puškaš
- Institute of Histology and Embryology “Aleksandar Đ. Kostić”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna Pešić
- Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
47
|
Voss L, Bartos M, Elgueta C, Sauer JF. Interneuron function and cognitive behavior are preserved upon postnatal removal of Lhx6. Sci Rep 2022; 12:4923. [PMID: 35318414 PMCID: PMC8941127 DOI: 10.1038/s41598-022-09003-4] [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: 04/15/2021] [Accepted: 03/09/2022] [Indexed: 11/22/2022] Open
Abstract
LIM homeobox domain transcription factor 6 (Lhx6) is crucial for the prenatal specification and differentiation of hippocampal GABAergic interneuron precursors. Interestingly, Lhx6 remains to be expressed in parvalbumin-positive hippocampal interneurons (PVIs) long after specification and differentiation have been completed, the functional implications of which remain elusive. We addressed the role of adult-expressed Lhx6 in the hippocampus by knocking down Lhx6 in adult mice (> 8 weeks old) using viral or transgenic expression of Cre-recombinase in Lhx6loxP/loxP mice. Late removal of Lhx6 did not affect the number of PVIs and had no impact on the morphological and physiological properties of PVIs. Furthermore, mice lacking Lhx6 in PVIs displayed normal cognitive behavior. Loss of Lhx6 only partially reduced the expression of Sox6 and Arx, downstream transcription factors that depend on Lhx6 during embryonic development of PVIs. Our data thus suggest that while Lhx6 is vitally important to drive interneuron transcriptional networks during early development, it becomes uncoupled from downstream effectors during postnatal life.
Collapse
Affiliation(s)
- Lars Voss
- Institute of Physiology I, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marlene Bartos
- Institute of Physiology I, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Claudio Elgueta
- Institute of Physiology I, Medical Faculty, University of Freiburg, Freiburg, Germany.
| | - Jonas-Frederic Sauer
- Institute of Physiology I, Medical Faculty, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
48
|
Strous JFM, Weeland CJ, van der Draai FA, Daams JG, Denys D, Lok A, Schoevers RA, Figee M. Brain Changes Associated With Long-Term Ketamine Abuse, A Systematic Review. Front Neuroanat 2022; 16:795231. [PMID: 35370568 PMCID: PMC8972190 DOI: 10.3389/fnana.2022.795231] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/10/2022] [Indexed: 12/28/2022] Open
Abstract
Recently, the abuse of ketamine has soared. Therefore, it is of great importance to study its potential risks. The effects of prolonged ketamine on the brain can be observationally studied in chronic recreational users. We performed a systematic review of studies reporting functional and structural brain changes after repeated ketamine abuse. We searched the following electronic databases: Medline, Embase and PsycINFO We screened 11,438 records and 16 met inclusion criteria, totaling 440 chronic recreational ketamine users (2–9.7 years; mean use 2.4 g/day), 259 drug-free controls and 44 poly-drug controls. Long-term recreational ketamine use was associated with lower gray matter volume and less white matter integrity, lower functional thalamocortical and corticocortical connectivity. The observed differences in both structural and functional neuroanatomy between ketamine users and controls may explain some of its long-term cognitive and psychiatric side effects, such as memory impairment and executive functioning. Given the effect that long-term ketamine exposure may yield, an effort should be made to curb its abuse.
Collapse
Affiliation(s)
- Jurriaan F. M. Strous
- Department of Psychiatry, University Medical Center Groningen, Groningen, Netherlands
- *Correspondence: Jurriaan F. M. Strous
| | - Cees J. Weeland
- Amsterdam University Medical Center, Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Joost G. Daams
- Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, Netherlands
| | - Damiaan Denys
- Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Anja Lok
- Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, Netherlands
| | - Robert A. Schoevers
- Department of Psychiatry, University Medical Center Groningen, Groningen, Netherlands
| | - Martijn Figee
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
49
|
Twarkowksi H, Steininger V, Kim MJ, Sahay A. A dentate gyrus- CA3 inhibitory circuit promotes evolution of hippocampal-cortical ensembles during memory consolidation. eLife 2022; 11:70586. [PMID: 35191834 PMCID: PMC8903830 DOI: 10.7554/elife.70586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Memories encoded in the dentate gyrus (DG) - CA3 circuit of the hippocampus are routed from CA1 to anterior cingulate cortex (ACC) for consolidation. Although CA1 parvalbumin inhibitory neurons (PV INs) orchestrate hippocampal-cortical communication, we know less about CA3 PV INs or DG - CA3 principal neuron - IN circuit mechanisms that contribute to evolution of hippocampal-cortical ensembles during memory consolidation. Using viral genetics to selectively mimic and boost an endogenous learning-dependent circuit mechanism, DG cell recruitment of CA3 PV INs and feed-forward inhibition (FFI) in CA3, in combination with longitudinal in vivo calcium imaging, we demonstrate that FFI facilitates formation and maintenance of context-associated neuronal ensembles in CA1. Increasing FFI in DG - CA3 promoted context specificity of neuronal ensembles in ACC over time and enhanced long-term contextual fear memory. In vivo LFP recordings in mice with increased FFI in DG - CA3 identified enhanced CA1 sharp-wave ripple - ACC spindle coupling as a potential network mechanism facilitating memory consolidation. Our findings illuminate how FFI in DG - CA3 dictates evolution of ensemble properties in CA1 and ACC during memory consolidation and suggest a teacher-like function for hippocampal CA1 in stabilization and re-organization of cortical representations.
Collapse
Affiliation(s)
- Hannah Twarkowksi
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, United States
| | - Victor Steininger
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, United States
| | - Min Jae Kim
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, United States
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, United States
| |
Collapse
|
50
|
Tsoi SY, Öncül M, Svahn E, Robertson M, Bogdanowicz Z, McClure C, Sürmeli G. Telencephalic outputs from the medial entorhinal cortex are copied directly to the hippocampus. eLife 2022; 11:e73162. [PMID: 35188100 PMCID: PMC8940174 DOI: 10.7554/elife.73162] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 02/04/2022] [Indexed: 11/25/2022] Open
Abstract
Complementary actions of the neocortex and the hippocampus enable encoding and long-term storage of experience dependent memories. Standard models for memory storage assume that sensory signals reach the hippocampus from superficial layers of the entorhinal cortex (EC). Deep layers of the EC on the other hand relay hippocampal outputs to the telencephalic structures including many parts of the neocortex. Here, we show that cells in layer 5a of the medial EC send a copy of their telencephalic outputs back to the CA1 region of the hippocampus. Combining cell-type-specific anatomical tracing with high-throughput RNA-sequencing based projection mapping and optogenetics aided circuit mapping, we show that in the mouse brain these projections have a unique topography and target hippocampal pyramidal cells and interneurons. Our results suggest that projections of deep medial EC neurons are anatomically configured to influence the hippocampus and neocortex simultaneously and therefore lead to novel hypotheses on the functional role of the deep EC.
Collapse
Affiliation(s)
- Sau Yee Tsoi
- University of Edinburgh, Centre for Discovery Brain SciencesEdinburghUnited Kingdom
| | - Merve Öncül
- University of Edinburgh, Centre for Discovery Brain SciencesEdinburghUnited Kingdom
| | - Ella Svahn
- University of Edinburgh, Centre for Discovery Brain SciencesEdinburghUnited Kingdom
| | - Mark Robertson
- University of Edinburgh, Centre for Discovery Brain SciencesEdinburghUnited Kingdom
| | - Zuzanna Bogdanowicz
- University of Edinburgh, Centre for Discovery Brain SciencesEdinburghUnited Kingdom
| | - Christina McClure
- University of Edinburgh, Centre for Discovery Brain SciencesEdinburghUnited Kingdom
| | - Gülşen Sürmeli
- University of Edinburgh, Centre for Discovery Brain SciencesEdinburghUnited Kingdom
- Simons Initiative for the Developing Brain, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|