1
|
Gazerani P. The neuroplastic brain: current breakthroughs and emerging frontiers. Brain Res 2025; 1858:149643. [PMID: 40280532 DOI: 10.1016/j.brainres.2025.149643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/01/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Neuroplasticity, the brain's capacity to reorganize itself by forming new neural connections, is central to modern neuroscience. Once believed to occur only during early development, research now shows that plasticity continues throughout the lifespan, supporting learning, memory, and recovery from injury or disease. Substantial progress has been made in understanding the mechanisms underlying neuroplasticity and their therapeutic applications. This overview article examines synaptic plasticity, structural remodeling, neurogenesis, and functional reorganization, highlighting both adaptive (beneficial) and maladaptive (harmful) processes across different life stages. Recent strategies to harness neuroplasticity, ranging from pharmacological agents and lifestyle interventions to cutting-edge technologies like brain-computer interfaces (BCIs) and targeted neuromodulation are evaluated in light of current empirical evidence. Contradictory findings in the literature are addressed, and methodological limitations that hamper widespread clinical adoption are discussed. The ethical and societal implications of deploying novel neuroplasticity-based interventions, including issues of equitable access, data privacy, and the blurred line between treatment and enhancement, are then explored in a structured manner. By integrating mechanistic insights, empirical data, and ethical considerations, the aim is to provide a comprehensive and balanced perspective for researchers, clinicians, and policymakers working to optimize brain health across diverse populations.
Collapse
Affiliation(s)
- Parisa Gazerani
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Pilestredet 50, 0167 Oslo, Norway.
| |
Collapse
|
2
|
Srinivasan K, Plenz D, Girvan M. Boosting Reservoir Computing with Brain-inspired Adaptive Dynamics. ARXIV 2025:arXiv:2504.12480v1. [PMID: 40321946 PMCID: PMC12047930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Reservoir computers (RCs) provide a computationally efficient alternative to deep learning while also offering a framework for incorporating brain-inspired computational principles. By using an internal neural network with random, fixed connections-the 'reservoir'-and training only the output weights, RCs simplify the training process but remain sensitive to the choice of hyperparameters that govern activation functions and network architecture. Moreover, typical RC implementations overlook a critical aspect of neuronal dynamics: the balance between excitatory and inhibitory (E-I) signals, which is essential for robust brain function. We show that RCs characteristically perform best in balanced or slightly over-inhibited regimes, outperforming excitation-dominated ones. To reduce the need for precise hyperparameter tuning, we introduce a self-adapting mechanism that locally adjusts E/I balance to achieve target neuronal firing rates, improving performance by up to 130% in tasks like memory capacity and time series prediction compared with globally tuned RCs. Incorporating brain-inspired heterogeneity in target neuronal firing rates further reduces the need for fine-tuning hyperparameters and enables RCs to excel across linear and non-linear tasks. These results support a shift from static optimization to dynamic adaptation in reservoir design, demonstrating how brain-inspired mechanisms can improve RC performance and robustness while deepening our understanding of neural computation.
Collapse
Affiliation(s)
- Keshav Srinivasan
- Biophysics Program, University of Maryland, College Park, MD 20740, USA
- Section on Critical Brain Dynamics, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Dietmar Plenz
- Section on Critical Brain Dynamics, National Institute of Mental Health, Bethesda, MD 20892, USA
| | - Michelle Girvan
- Biophysics Program, University of Maryland, College Park, MD 20740, USA
- Department of Physics, University of Maryland, College Park, MD 20740, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| |
Collapse
|
3
|
Leman DP, Cary BA, Bissen D, Lane BJ, Shanley MR, Wong NF, Bhut KB, Turrigiano GG. Rapid prey capture learning drives a slow resetting of network activity in rodent binocular visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648036. [PMID: 40291689 PMCID: PMC12027325 DOI: 10.1101/2025.04.11.648036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Neocortical neurons possess stable firing rate set points to which they faithfully return when perturbed. These set points are established early and are stable through adulthood, suggesting they are immutable. Here we challenge this idea using an ethological vision-dependent prey capture learning paradigm in juvenile rats. This learning required visual cortex (V1), and enhanced tuning of V1 neurons to specific behavioral epochs. Chronic recordings revealed a slow, state-dependent increase in V1 firing that began after learning was complete and persisted for days. This upward firing rate plasticity was gradual, gated by wake states, and in L2/3 was driven by a TNFα-dependent increase in excitatory synapses onto pyramidal neurons - all features of homeostatic plasticity within V1. Finally, TNFα inhibition after learning reduced retention of hunting skills. Thus, naturalistic learning in juvenile animals co-opts homeostatic forms of plasticity to reset firing rate setpoints within V1, in a process that facilitates skill consolidation.
Collapse
|
4
|
Dai W, Zhang Y, Cheng Y, Dong M, Qian Y, Wang X, Guo C, Liu H, Shen Y. Timing Matters: Preconditioning Effects of Cathodal Transcranial Direct Current Stimulation on Intermittent Theta-Burst Stimulation-Induced Neuroplasticity in the Primary Motor Cortex. Neuromodulation 2025; 28:520-531. [PMID: 39969455 DOI: 10.1016/j.neurom.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Recent advances have highlighted the interplay between intermittent theta-burst stimulation (iTBS) and transcranial direct current stimulation (tDCS) in neuroplasticity modulation. However, the synergistic potential of these modalities in optimizing plasticity, particularly with cathodal tDCS preconditioning before iTBS, remains poorly understood. OBJECTIVE This study examined the effects of cathodal high-definition tDCS (HD-tDCS) preconditioning on iTBS-induced neuroplasticity in the primary motor cortex at different timing intervals. MATERIALS AND METHODS Twenty healthy participants underwent four stimulation sessions in a randomized cross-over design, receiving iTBS either immediately or at 10-minute and 30-minute intervals after cathodal HD-tDCS preconditioning, in addition to a control session with iTBS immediately after sham HD-tDCS. Motor evoked potentials (MEPs) were measured at baseline and 5, 10, 15, and 30 minutes after iTBS to assess changes in neuroplasticity. Each session was separated by ≥one week to prevent carry-over effects. RESULTS Compared with sham sessions, immediate cathodal HD-tDCS preconditioning significantly enhanced MEPs across all measured intervals after iTBS, with sustained neuroplasticity persisting for up to 30 minutes. Immediate preconditioning produced significant MEP enhancements at 5 and 10 minutes when compared with the 30-minute delayed condition. CONCLUSIONS The effectiveness of cathodal tDCS preconditioning in enhancing iTBS-induced neuroplasticity decreased with increasing intervals between tDCS and iTBS application. These findings highlight the essential role of precise timing in tDCS preconditioning for maximizing the neuroplastic effects of iTBS and offer valuable insights for optimizing neurorehabilitation protocols.
Collapse
Affiliation(s)
- Wenjun Dai
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yishu Zhang
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yihui Cheng
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Manyu Dong
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yilun Qian
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyue Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuan Guo
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hanjun Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ying Shen
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Papatheodoropoulos C. Compensatory Regulation of Excitation/Inhibition Balance in the Ventral Hippocampus: Insights from Fragile X Syndrome. BIOLOGY 2025; 14:363. [PMID: 40282228 PMCID: PMC12025323 DOI: 10.3390/biology14040363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
The excitation/inhibition (E/I) balance is a critical feature of neural circuits, which is crucial for maintaining optimal brain function by ensuring network stability and preventing neural hyperexcitability. The hippocampus exhibits the particularly interesting characteristics of having different functions and E/I profiles between its dorsal and ventral segments. Furthermore, the hippocampus is particularly vulnerable to epilepsy and implicated in Fragile X Syndrome (FXS), disorders associated with heightened E/I balance and possible deficits in GABA-mediated inhibition. In epilepsy, the ventral hippocampus shows heightened susceptibility to seizures, while in FXS, recent evidence suggests differential alterations in excitability and inhibition between dorsal and ventral regions. This article explores the mechanisms underlying E/I balance regulation, focusing on the hippocampus in epilepsy and FXS, and emphasizing the possible mechanisms that may confer homeostatic flexibility to the ventral hippocampus in maintaining E/I balance. Notably, the ventral hippocampus in adult FXS models shows enhanced GABAergic inhibition, resistance to epileptiform activity, and physiological network pattern (sharp wave-ripples, SWRs), potentially representing a homeostatic adaptation. In contrast, the dorsal hippocampus in these FXS models is more vulnerable to aberrant discharges and displays altered SWRs. These findings highlight the complex, region-specific nature of E/I balance disruptions in neurological disorders and suggest that the ventral hippocampus may possess unique compensatory mechanisms. Specifically, it is proposed that the ventral hippocampus, the brain region most prone to hyperexcitability, may have unique adaptive capabilities at the cellular and network levels that maintain the E/I balance within a normal range to prevent the transition to hyperexcitability and preserve normal function. Investigating the mechanisms underlying these compensatory responses in the ventral hippocampus and their developmental trajectories may offer novel insights into strategies for mitigating E/I imbalances in epilepsy, FXS, and potentially other neuropsychiatric and neurodevelopmental disorders.
Collapse
|
6
|
Koesters AG, Rich MM, Engisch KL. Homeostatic Synaptic Plasticity of Miniature Excitatory Postsynaptic Currents in Mouse Cortical Cultures Requires Neuronal Rab3A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.06.14.544980. [PMID: 39071374 PMCID: PMC11275788 DOI: 10.1101/2023.06.14.544980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Following prolonged activity blockade, amplitudes of miniature excitatory postsynaptic currents (mEPSCs) increase, a form of plasticity termed "homeostatic synaptic plasticity." We previously showed that a presynaptic protein, the small GTPase Rab3A, is required for full expression of the increase in miniature endplate current amplitudes following prolonged blockade of action potential activity at the mouse neuromuscular junction in vivo, where an increase in postsynaptic receptors does not contribute (Wang et al., 2005; Wang et al., 2011). It is unknown whether this form of Rab3A-dependent homeostatic plasticity at the neuromuscular junction shares any characteristics with central synapses. We show here that homeostatic synaptic plasticity of mEPSCs is impaired in mouse cortical neuron cultures prepared from Rab3A-/- and mutant mice expressing a single point mutation of Rab3A, Rab3A Earlybird mice. To determine if Rab3A is involved in the well-established homeostatic increase in postsynaptic AMPA-type receptors (AMPARs), we performed a series of experiments in which electrophysiological recordings of mEPSCs and confocal imaging of synaptic AMPAR immunofluorescence were assessed within the same cultures. We found that the increase in postsynaptic AMPAR levels in wild type cultures was more variable than that of mEPSC amplitudes, which might be explained by a presynaptic contribution, but we cannot rule out variability in the measurement. Finally, we demonstrate that Rab3A is acting in neurons because only selective loss of Rab3A in neurons, not glia, disrupted the homeostatic increase in mEPSC amplitudes. This is the first demonstration that a protein thought to function presynaptically is required for homeostatic synaptic plasticity of quantal size in central neurons.
Collapse
Affiliation(s)
- Andrew G. Koesters
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mark M. Rich
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45345
| | - Kathrin L. Engisch
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and the College of Science and Mathematics, Wright State University, Dayton, OH 45435
| |
Collapse
|
7
|
Xu M, Liu F, Hu Y, Li H, Wei Y, Zhong S, Pei J, Deng L. Adaptive Synaptic Scaling in Spiking Networks for Continual Learning and Enhanced Robustness. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2025; 36:5151-5165. [PMID: 38536699 DOI: 10.1109/tnnls.2024.3373599] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Synaptic plasticity plays a critical role in the expression power of brain neural networks. Among diverse plasticity rules, synaptic scaling presents indispensable effects on homeostasis maintenance and synaptic strength regulation. In the current modeling of brain-inspired spiking neural networks (SNN), backpropagation through time is widely adopted because it can achieve high performance using a small number of time steps. Nevertheless, the synaptic scaling mechanism has not yet been well touched. In this work, we propose an experience-dependent adaptive synaptic scaling mechanism (AS-SNN) for spiking neural networks. The learning process has two stages: First, in the forward path, adaptive short-term potentiation or depression is triggered for each synapse according to afferent stimuli intensity accumulated by presynaptic historical neural activities. Second, in the backward path, long-term consolidation is executed through gradient signals regulated by the corresponding scaling factor. This mechanism shapes the pattern selectivity of synapses and the information transfer they mediate. We theoretically prove that the proposed adaptive synaptic scaling function follows a contraction map and finally converges to an expected fixed point, in accordance with state-of-the-art results in three tasks on perturbation resistance, continual learning, and graph learning. Specifically, for the perturbation resistance and continual learning tasks, our approach improves the accuracy on the N-MNIST benchmark over the baseline by 44% and 25%, respectively. An expected firing rate callback and sparse coding can be observed in graph learning. Extensive experiments on ablation study and cost evaluation evidence the effectiveness and efficiency of our nonparametric adaptive scaling method, which demonstrates the great potential of SNN in continual learning and robust learning.
Collapse
|
8
|
Marsh B, Chauvette S, Huang M, Timofeev I, Bazhenov M. Network effects of traumatic brain injury: from infra slow to high frequency oscillations and seizures. J Comput Neurosci 2025:10.1007/s10827-025-00895-5. [PMID: 40019646 DOI: 10.1007/s10827-025-00895-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 03/01/2025]
Abstract
Traumatic brain injury (TBI) can have a multitude of effects on neural functioning. In extreme cases, TBI can lead to seizures both immediately following the injury as well as persistent epilepsy over years to a lifetime. However, mechanisms of neural dysfunctioning after TBI remain poorly understood. To address these questions, we analyzed human and animal data and we developed a biophysical network model implementing effects of ion concentration dynamics and homeostatic synaptic plasticity to test effects of TBI on the brain network dynamics. We focus on three primary phenomena that have been reported in vivo after TBI: an increase in infra slow oscillations (<0.1 Hz), increase in Delta power (1 - 4 Hz), and the emergence of broadband Gamma bursts (30 - 100 Hz). Using computational network model, we show that the infra slow oscillations can be directly attributed to extracellular potassium dynamics, while the increase in Delta power and occurrence of Gamma bursts are related to the increase in strength of synaptic weights from homeostatic synaptic scaling triggered by trauma. We also show that the buildup of Gamma bursts in the injured region can lead to seizure-like events that propagate across the entire network; seizures can then be initiated in previously healthy regions. This study brings greater understanding of the network effects of TBI and how they can lead to epileptic activity. This lays the foundation to begin investigating how injured networks can be healed and seizures prevented.
Collapse
Affiliation(s)
- Brianna Marsh
- Neuroscience Graduate Program, The University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA.
| | - Sylvain Chauvette
- Department of Psychiatry and Neuroscience, Université Laval, 2325 Rue de l'Université, Québec, QC G1V 0A6, Canada
| | - Mingxiong Huang
- Department of Radiology, The University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA
| | - Igor Timofeev
- Department of Psychiatry and Neuroscience, Université Laval, 2325 Rue de l'Université, Québec, QC G1V 0A6, Canada
| | - Maxim Bazhenov
- Department of Medicine, The University of California San Diego, 9500 Gilman Dr, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
9
|
Chien C, He K, Perry S, Tchitchkan E, Han Y, Li X, Dickman D. Distinct input-specific mechanisms enable presynaptic homeostatic plasticity. SCIENCE ADVANCES 2025; 11:eadr0262. [PMID: 39951523 PMCID: PMC11827636 DOI: 10.1126/sciadv.adr0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
Synapses are endowed with the flexibility to change through experience, but must be sufficiently stable to last a lifetime. This tension is illustrated at the Drosophila neuromuscular junction (NMJ), where two motor inputs that differ in structural and functional properties coinnervate most muscles to coordinate locomotion. To stabilize NMJ activity, motor neurons augment neurotransmitter release following diminished postsynaptic glutamate receptor functionality, termed presynaptic homeostatic potentiation (PHP). How these distinct inputs contribute to PHP plasticity remains enigmatic. We have used a botulinum neurotoxin to selectively silence each input and resolve their roles in PHP, demonstrating that PHP is input specific: Chronic (genetic) PHP selectively targets the tonic MN-Ib, where active zone remodeling enhances Ca2+ influx to promote increased glutamate release. In contrast, acute (pharmacological) PHP selectively increases vesicle pools to potentiate phasic MN-Is. Thus, distinct homeostatic modulations in active zone nanoarchitecture, vesicle pools, and Ca2+ influx collaborate to enable input-specific PHP expression.
Collapse
Affiliation(s)
- Chun Chien
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Kaikai He
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Tchitchkan
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
- USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Ruggiero A, Heim LR, Susman L, Hreaky D, Shapira I, Katsenelson M, Rosenblum K, Slutsky I. NMDA receptors regulate the firing rate set point of hippocampal circuits without altering single-cell dynamics. Neuron 2025; 113:244-259.e7. [PMID: 39515323 DOI: 10.1016/j.neuron.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/05/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Understanding how neuronal circuits stabilize their activity is a fundamental yet poorly understood aspect of neuroscience. Here, we show that hippocampal network properties, such as firing rate distribution and dimensionality, are actively regulated, despite perturbations and single-cell drift. Continuous inhibition of N-methyl-D-aspartate receptors (NMDARs) ex vivo lowers the excitation/inhibition ratio and network firing rates while preserving resilience to perturbations. This establishes a new network firing rate set point via NMDAR-eEF2K signaling pathway. NMDARs' capacity to modulate and stabilize network firing is mediated by excitatory synapses and the intrinsic excitability of parvalbumin-positive neurons, respectively. In behaving mice, continuous NMDAR blockade in CA1 reduces network firing without altering single-neuron drift or triggering a compensatory response. These findings expand NMDAR function beyond their canonical role in synaptic plasticity and raise the possibility that some NMDAR-dependent behavioral effects are mediated by their unique regulation of population activity set points.
Collapse
Affiliation(s)
- Antonella Ruggiero
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Leore R Heim
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Lee Susman
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, 69978 Tel Aviv, Israel; Center for the Physics of Biological Function, Princeton University, Princeton, NJ 08544, USA
| | - Dema Hreaky
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Maxim Katsenelson
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Kobi Rosenblum
- Sagol Department of Neurobiology, Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel; Sieratzki Institute for Advances in Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
11
|
de León-López CAM, Carretero-Rey M, Khan ZU. AMPA Receptors in Synaptic Plasticity, Memory Function, and Brain Diseases. Cell Mol Neurobiol 2025; 45:14. [PMID: 39841263 PMCID: PMC11754374 DOI: 10.1007/s10571-024-01529-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/26/2024] [Indexed: 01/23/2025]
Abstract
Tetrameric AMPA-type ionotropic glutamate receptors are primary transducers of fast excitatory synaptic transmission in the central nervous system, and their properties and abundance at the synaptic surface are crucial determinants of synaptic efficacy in neuronal communication across the brain. The induction of long-term potentiation (LTP) leads to the insertion of GluA1-containing AMPA receptors at the synaptic surface, whereas during long-term depression (LTD), these receptors are internalized into the cytoplasm of the spine. Disruptions in the trafficking of AMPA receptors to and from the synaptic surface attenuate both forms of synaptic plasticity. Homeostatic scaling up and scaling down, which are additional types of plasticity similar to LTP and LTD, are also regulated by the insertion and removal of GluA1-containing AMPA receptors from the synaptic surface. The trafficking of AMPA receptors is an intricate process assisted by various proteins. Furthermore, AMPA receptors are critical for the formation and consolidation of various types of memory, and alterations in their function are intimately associated with cognitive dysfunction in aging and several neurological and psychiatric diseases. In this review, we will provide an overview of the current understanding of how AMPA receptors regulate various forms of synaptic plasticity, their contribution to memory functions, and their role in aging and brain diseases.
Collapse
Affiliation(s)
- Cristina A Muñoz de León-López
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain
| | - Marta Carretero-Rey
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain
| | - Zafar U Khan
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain.
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain.
- CIBERNED, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
12
|
Gerasimov E, Pchitskaya E, Vlasova O, Bezprozvanny I. Dynamic changes in the hippocampal neuronal circuits activity following acute stress revealed by miniature fluorescence microscopy imaging. Mol Brain 2024; 17:92. [PMID: 39695833 DOI: 10.1186/s13041-024-01168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Coordinated activity of neuronal ensembles is a basis for information processing in the brain. Recent development of miniscope imaging technology enabled recordings of neuronal circuits activity in vivo in freely behaving animals. Acute stress is believed to affect various hippocampal functions, especially memory. In the current study, we utilized miniscope imaging to investigate the hippocampal neuronal circuits properties in a mouse as function of time and immediately in response to an acute stress, induced by passive restraint, 3 h and 10 days after. Comprehensive quantitative analysis of network activity changes at the neuronal ensembles level revealed highly stable neuronal activity parameters, which exhibited a rapid and robust shift in response to acute stress stimulation. This shift was accompanied by the restructuring of the pairwise-correlated neuronal pairs. Remarkably, we discovered that ensembles activity characteristics returned to the initial state following recovery period, demonstrating hippocampal homeostatic stability at the neuronal circuits level. Obtained results provide an evidence about hippocampal neuronal ensembles activity in response to acute stress over time.
Collapse
Affiliation(s)
- Evgenii Gerasimov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia.
| | - Ekaterina Pchitskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia
| | - Olga Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia.
| |
Collapse
|
13
|
Michetti C, Benfenati F. Homeostatic regulation of brain activity: from endogenous mechanisms to homeostatic nanomachines. Am J Physiol Cell Physiol 2024; 327:C1384-C1399. [PMID: 39401424 DOI: 10.1152/ajpcell.00470.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 11/12/2024]
Abstract
After the initial concepts of the constancy of the internal milieu or homeostasis, put forward by Claude Bernard and Walter Cannon, homeostasis emerged as a mechanism to control oscillations of biologically meaningful variables within narrow physiological ranges. This is a primary need in the central nervous system that is continuously subjected to a multitude of stimuli from the internal and external environments that affect its function and structure, allowing to adapt the individual to the ever-changing daily conditions. Preserving physiological levels of activity despite disturbances that could either depress neural computation or excessively stimulate neural activity is fundamental, and failure of these homeostatic mechanisms can lead to brain diseases. In this review, we cover the role and main mechanisms of homeostatic plasticity involving the regulation of excitability and synaptic strength from the single neuron to the network level. We analyze the relationships between homeostatic and Hebbian plasticity and the conditions under which the preservation of the excitatory/inhibitory balance fails, triggering epileptogenesis and eventually epilepsy. Several therapeutic strategies to cure epilepsy have been designed to strengthen homeostasis when endogenous homeostatic plasticity mechanisms have become insufficient or ineffective to contrast hyperactivity. We describe "on demand" gene therapy strategies, including optogenetics, chemogenetics, and chemo-optogenetics, and particularly focus on new closed loop sensor-actuator strategies mimicking homeostatic plasticity that can be endogenously expressed to strengthen the homeostatic defenses against brain diseases.
Collapse
Affiliation(s)
- Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
14
|
Jasińska M, Jasek-Gajda E, Ziaja M, Litwin JA, Lis GJ, Pyza E. Light-Modulated Circadian Synaptic Plasticity in the Somatosensory Cortex: Link to Locomotor Activity. Int J Mol Sci 2024; 25:12870. [PMID: 39684579 DOI: 10.3390/ijms252312870] [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: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
The circadian clock controls various physiological processes, including synaptic function and neuronal activity, affecting the functioning of the entire organism. Light is an important external factor regulating the day-night cycle. This study examined the effects of the circadian clock and light on synaptic plasticity, and explored how locomotor activity contributes to these processes. We analyzed synaptic protein expression and excitatory synapse density in the somatosensory cortex of mice from four groups exposed to different lighting conditions (LD 12:12, DD, LD 16:8, and LL). Locomotor activity was assessed through individual wheel-running monitoring. To explore daily and circadian changes in synaptic proteins, we performed double-immunofluorescence labeling and laser scanning confocal microscopy imaging, targeting three pairs of presynaptic and postsynaptic proteins (Synaptophysin 1/PSD95, Piccolo/Homer 1, Neurexins/PICK1). Excitatory synapse density was evaluated by co-labeling presynaptic and postsynaptic markers. Our results demonstrated that all the analyzed synaptic proteins exhibited circadian regulation modulated by light. Under constant light conditions, only Piccolo and Homer 1 showed rhythmicity. Locomotor activity was also associated with the circadian clock's effects on synaptic proteins, showing a stronger connection to changes in postsynaptic protein levels. Excitatory synapse density peaked during the day/subjective day and exhibited an inverse relationship with locomotor activity. Continued light exposure disrupted cyclic changes in synapse density but kept it consistently elevated. These findings underscore the crucial roles of light and locomotor activity in regulating synaptic plasticity.
Collapse
Affiliation(s)
- Małgorzata Jasińska
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Ewa Jasek-Gajda
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Marek Ziaja
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Jan A Litwin
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Grzegorz J Lis
- Department of Histology, Jagiellonian University Medical College, 31-034 Krakow, Poland
| | - Elżbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Krakow, Poland
| |
Collapse
|
15
|
Chien C, He K, Perry S, Tchitchkan E, Han Y, Li X, Dickman D. Distinct input-specific mechanisms enable presynaptic homeostatic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612361. [PMID: 39314403 PMCID: PMC11419068 DOI: 10.1101/2024.09.10.612361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Synapses are endowed with the flexibility to change through experience, but must be sufficiently stable to last a lifetime. This tension is illustrated at the Drosophila neuromuscular junction (NMJ), where two motor inputs that differ in structural and functional properties co-innervate most muscles to coordinate locomotion. To stabilize NMJ activity, motor neurons augment neurotransmitter release following diminished postsynaptic glutamate receptor functionality, termed presynaptic homeostatic potentiation (PHP). How these distinct inputs contribute to PHP plasticity remains enigmatic. We have used a botulinum neurotoxin to selectively silence each input and resolve their roles in PHP, demonstrating that PHP is input-specific: Chronic (genetic) PHP selectively targets the tonic MN-Ib, where active zone remodeling enhances Ca2+ influx to promote increased glutamate release. In contrast, acute (pharmacological) PHP selectively increases vesicle pools to potentiate phasic MN-Is. Thus, distinct homeostatic modulations in active zone nanoarchitecture, vesicle pools, and Ca2+ influx collaborate to enable input-specific PHP expression.
Collapse
Affiliation(s)
- Chun Chien
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Kaikai He
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Sarah Perry
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| | - Elizabeth Tchitchkan
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| | - Yifu Han
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Xiling Li
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Dion Dickman
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| |
Collapse
|
16
|
Chen X, Bialek W. Searching for long timescales without fine tuning. Phys Rev E 2024; 110:034407. [PMID: 39425360 DOI: 10.1103/physreve.110.034407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 09/03/2024] [Indexed: 10/21/2024]
Abstract
Animal behavior occurs on timescales much longer than the response times of individual neurons. In many cases, it is plausible that these long timescales emerge from the recurrent dynamics of electrical activity in networks of neurons. In linear models, timescales are set by the eigenvalues of a dynamical matrix whose elements measure the strengths of synaptic connections between neurons. It is not clear to what extent these matrix elements need to be tuned to generate long timescales; in some cases, one needs not just a single long timescale but a whole range. Starting from the simplest case of random symmetric connections, we combine maximum entropy and random matrix theory methods to construct ensembles of networks, exploring the constraints required for long timescales to become generic. We argue that a single long timescale can emerge generically from realistic constraints, but a full spectrum of slow modes requires more tuning. Langevin dynamics that generates patterns of synaptic connections drawn from these ensembles involves a combination of Hebbian learning and activity-dependent synaptic scaling.
Collapse
Affiliation(s)
- Xiaowen Chen
- Joseph Henry Laboratories of Physics, and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
- Laboratoire de Physique de l'Ecole Normale Supérieure, ENS, PSL Université, CNRS, Sorbonne Université, Université Paris Cité, F-75005 Paris, France
| | - William Bialek
- Joseph Henry Laboratories of Physics, and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
- Initiative for the Theoretical Sciences, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, New York 10016, USA
| |
Collapse
|
17
|
Shen W, Wang P, Wei G, Yuan S, Chen M, Su Y, Xu B, Li G. SiC@NiO Core-Shell Nanowire Networks-Based Optoelectronic Synapses for Neuromorphic Computing and Visual Systems at High Temperature. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400458. [PMID: 38607289 DOI: 10.1002/smll.202400458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/18/2024] [Indexed: 04/13/2024]
Abstract
1D nanowire networks, sharing similarities of structure, information transfer, and computation with biological neural networks, have emerged as a promising platform for neuromorphic systems. Based on brain-like structures of 1D nanowire networks, neuromorphic synaptic devices can overcome the von Neumann bottleneck, achieving intelligent high-efficient sensing and computing function with high information processing rates and low power consumption. Here, high-temperature neuromorphic synaptic devices based on SiC@NiO core-shell nanowire networks optoelectronic memristors (NNOMs) are developed. Experimental results demonstrate that NNOMs attain synaptic short/long-term plasticity and modulation plasticity under both electrical and optical stimulation, and exhibit advanced functions such as short/long-term memory and "learning-forgetting-relearning" under optical stimulation at both room temperature and 200 °C. Based on the advanced functions under light stimulus, the constructed 5 × 3 optoelectronic synaptic array devices exhibit a stable visual memory function up to 200 °C, which can be utilized to develop artificial visual systems. Additionally, when exposed to multiple electronic or optical stimuli, the NNOMs effectively replicate the principles of Pavlovian classical conditioning, achieving visual heterologous synaptic functionality and refining neural networks. Overall, with abundant synaptic characteristics and high-temperature thermal stability, these neuromorphic synaptic devices offer a promising route for advancing neuromorphic computing and visual systems.
Collapse
Affiliation(s)
- Weikang Shen
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
| | - Pan Wang
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
| | - Guodong Wei
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, Shanxi, 030024, P. R. China
| | - Shuai Yuan
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
| | - Mi Chen
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
| | - Ying Su
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
| | - Bingshe Xu
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, Shanxi, 030024, P. R. China
| | - Guoqiang Li
- Xi'an Key Laboratory of Compound Semiconductor Materials and Devices, School of Physics & Information Science, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, P. R. China
- The School of Integrated Circuits, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, 510641, P. R. China
| |
Collapse
|
18
|
Wen W, Turrigiano GG. Keeping Your Brain in Balance: Homeostatic Regulation of Network Function. Annu Rev Neurosci 2024; 47:41-61. [PMID: 38382543 DOI: 10.1146/annurev-neuro-092523-110001] [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] [Indexed: 02/23/2024]
Abstract
To perform computations with the efficiency necessary for animal survival, neocortical microcircuits must be capable of reconfiguring in response to experience, while carefully regulating excitatory and inhibitory connectivity to maintain stable function. This dynamic fine-tuning is accomplished through a rich array of cellular homeostatic plasticity mechanisms that stabilize important cellular and network features such as firing rates, information flow, and sensory tuning properties. Further, these functional network properties can be stabilized by different forms of homeostatic plasticity, including mechanisms that target excitatory or inhibitory synapses, or that regulate intrinsic neuronal excitability. Here we discuss which aspects of neocortical circuit function are under homeostatic control, how this homeostasis is realized on the cellular and molecular levels, and the pathological consequences when circuit homeostasis is impaired. A remaining challenge is to elucidate how these diverse homeostatic mechanisms cooperate within complex circuits to enable them to be both flexible and stable.
Collapse
Affiliation(s)
- Wei Wen
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| | - Gina G Turrigiano
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| |
Collapse
|
19
|
Chotard V, Trapani F, Glaziou G, Sermet BS, Yger P, Marre O, Rebsam A. Altered Functional Responses of the Retina in B6 Albino Tyrc/c Mice. Invest Ophthalmol Vis Sci 2024; 65:39. [PMID: 39189994 PMCID: PMC11361382 DOI: 10.1167/iovs.65.10.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 08/09/2024] [Indexed: 08/28/2024] Open
Abstract
Purpose Mammals with albinism present low visual discrimination ability and different proportions of certain retinal cell subtypes. As the spatial resolution of the retina depends on the visual field sampling by retinal ganglion cells (RGCs) based on the convergence of upstream cell inputs, it could be affected in albinism and thus modify the RGC function. Methods We used the Tyrc/c line, a mouse model of oculocutaneous albinism type 1 (OCA1), carrying a tyrosinase mutation, and previously characterized by a total absence of pigment and severe visual deficits. To assess their retinal function, we recorded the light responses of hundreds of RGCs ex vivo using multi-electrode array (MEA). We estimated the receptive field (RF)-center diameter of Tyr+/c and Tyrc/c RGCs using a checkerboard stimulation before simultaneously stimulating the center and surround of RGC RFs with full-field flashes. Results Following checkerboard stimulation, the RF-center diameters of RGCs were indistinguishable between Tyrc/c and Tyr+/c retinas. Nevertheless, RGCs from Tyrc/c retinas presented more OFF responses to full-field flashes than RGCs from Tyr+/c retinas. Unlike Tyr+/c retinas, very few OFF-center RGCs switched polarity to ON or ON-OFF responses after full-field flashes in Tyrc/c retinas, suggesting a different surround suppression in these retinas. Conclusions The retinal output signal is affected in Tyrc/c retinas, despite intact RF-center diameters of their RGCs. Adaptive mechanisms during development are probably responsible for this change in RGC responses, related to the absence of ocular pigments.
Collapse
Affiliation(s)
- Virginie Chotard
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Francesco Trapani
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Guilhem Glaziou
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Pierre Yger
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Alexandra Rebsam
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
20
|
Kim S, Jang G, Kim H, Lim D, Han KA, Um JW, Ko J. MDGAs perform activity-dependent synapse type-specific suppression via distinct extracellular mechanisms. Proc Natl Acad Sci U S A 2024; 121:e2322978121. [PMID: 38900791 PMCID: PMC11214077 DOI: 10.1073/pnas.2322978121] [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: 12/29/2023] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
MDGA (MAM domain containing glycosylphosphatidylinositol anchor) family proteins were previously identified as synaptic suppressive factors. However, various genetic manipulations have yielded often irreconcilable results, precluding precise evaluation of MDGA functions. Here, we found that, in cultured hippocampal neurons, conditional deletion of MDGA1 and MDGA2 causes specific alterations in synapse numbers, basal synaptic transmission, and synaptic strength at GABAergic and glutamatergic synapses, respectively. Moreover, MDGA2 deletion enhanced both N-methyl-D-aspartate (NMDA) receptor- and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor-mediated postsynaptic responses. Strikingly, ablation of both MDGA1 and MDGA2 abolished the effect of deleting individual MDGAs that is abrogated by chronic blockade of synaptic activity. Molecular replacement experiments further showed that MDGA1 requires the meprin/A5 protein/PTPmu (MAM) domain, whereas MDGA2 acts via neuroligin-dependent and/or MAM domain-dependent pathways to regulate distinct postsynaptic properties. Together, our data demonstrate that MDGA paralogs act as unique negative regulators of activity-dependent postsynaptic organization at distinct synapse types, and cooperatively contribute to adjustment of excitation-inhibition balance.
Collapse
Affiliation(s)
- Seungjoon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Gyubin Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Dongseok Lim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| |
Collapse
|
21
|
Cheung H, Yu TZ, Yi X, Wu YJ, Wang Q, Gu X, Xu M, Cai M, Wen W, Li XN, Liu YX, Sun Y, Zheng J, Xu TL, Luo Y, Zhang MZ, Li WG. An ultra-short-acting benzodiazepine in thalamic nucleus reuniens undermines fear extinction via intermediation of hippocamposeptal circuits. Commun Biol 2024; 7:728. [PMID: 38877285 PMCID: PMC11178775 DOI: 10.1038/s42003-024-06417-w] [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: 01/19/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
Benzodiazepines, commonly used for anxiolytics, hinder conditioned fear extinction, and the underlying circuit mechanisms are unclear. Utilizing remimazolam, an ultra-short-acting benzodiazepine, here we reveal its impact on the thalamic nucleus reuniens (RE) and interconnected hippocamposeptal circuits during fear extinction. Systemic or RE-specific administration of remimazolam impedes fear extinction by reducing RE activation through A type GABA receptors. Remimazolam enhances long-range GABAergic inhibition from lateral septum (LS) to RE, underlying the compromised fear extinction. RE projects to ventral hippocampus (vHPC), which in turn sends projections characterized by feed-forward inhibition to the GABAergic neurons of the LS. This is coupled with long-range GABAergic projections from the LS to RE, collectively constituting an overall positive feedback circuit construct that promotes fear extinction. RE-specific remimazolam negates the facilitation of fear extinction by disrupting this circuit. Thus, remimazolam in RE disrupts fear extinction caused by hippocamposeptal intermediation, offering mechanistic insights for the dilemma of combining anxiolytics with extinction-based exposure therapy.
Collapse
Affiliation(s)
- Hoiyin Cheung
- Center for Brain Science, Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tong-Zhou Yu
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xin Yi
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yan-Jiao Wu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qi Wang
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xue Gu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Miao Xu
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Meihua Cai
- Center for Brain Science, Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wen Wen
- Center for Brain Science, Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xin-Ni Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Ying-Xiao Liu
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Ying Sun
- Center for Brain Science, Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jijian Zheng
- Center for Brain Science, Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Tian-Le Xu
- Center for Brain Science, Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Ma-Zhong Zhang
- Center for Brain Science, Department of Anesthesiology and Pediatric Clinical Pharmacology Laboratory, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Wei-Guang Li
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
- Ministry of Education-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
22
|
Vitureira N, Rafael A, Abudara V. P2X7 receptors and pannexin1 hemichannels shape presynaptic transmission. Purinergic Signal 2024; 20:223-236. [PMID: 37713157 PMCID: PMC11189373 DOI: 10.1007/s11302-023-09965-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023] Open
Abstract
Over the last decades, since the discovery of ATP as a transmitter, accumulating evidence has been reported about the role of this nucleotide and purinergic receptors, in particular P2X7 receptors, in the modulation of synaptic strength and plasticity. Purinergic signaling has emerged as a crucial player in orchestrating the molecular interaction between the components of the tripartite synapse, and much progress has been made in how this neuron-glia interaction impacts neuronal physiology under basal and pathological conditions. On the other hand, pannexin1 hemichannels, which are functionally linked to P2X7 receptors, have appeared more recently as important modulators of excitatory synaptic function and plasticity under diverse contexts. In this review, we will discuss the contribution of ATP, P2X7 receptors, and pannexin hemichannels to the modulation of presynaptic strength and its impact on motor function, sensory processing, synaptic plasticity, and neuroglial communication, with special focus on the P2X7 receptor/pannexin hemichannel interplay. We also address major hypotheses about the role of this interaction in physiological and pathological circumstances.
Collapse
Affiliation(s)
- Nathalia Vitureira
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Alberto Rafael
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
23
|
Zolezzi DM, Larsen DB, McPhee M, Graven-Nielsen T. Effects of pain on cortical homeostatic plasticity in humans: a systematic review. Pain Rep 2024; 9:e1141. [PMID: 38444774 PMCID: PMC10914232 DOI: 10.1097/pr9.0000000000001141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/09/2023] [Accepted: 12/28/2023] [Indexed: 03/07/2024] Open
Abstract
Homeostatic plasticity (HP) is a negative feedback mechanism that prevents excessive facilitation or depression of cortical excitability (CE). Cortical HP responses in humans have been investigated by using 2 blocks of noninvasive brain stimulation with a no-stimulation block in between. A healthy HP response is characterized by reduced CE after 2 excitatory stimulation blocks and increased CE when using inhibitory stimulation. Conversely, impaired HP responses have been demonstrated in experimental and chronic pain conditions. Therefore, this systematic review aimed to provide an overview of the effect of pain on cortical HP in humans. Scopus, Embase, and PubMed were searched from inception until November 20, 2023. The included studies (1) compared experimental or clinical pain conditions with healthy controls, (2) induced HP using 2 blocks of stimulation with a no-stimulation interval, and (3) evaluated CE measures such as motor-evoked potentials. Four studies were included, consisting of 5 experiments and 146 participants, of whom 63 were patients with chronic pain and 48 were subjected to an experimental pain model. This systematic review found support for an HP impairment in pain compared with that in pain-free states, reflected by a lack of CE reduction after excitatory-excitatory HP induction over the primary motor cortex. Inhibitory-inhibitory HP induction did not produce a consistent HP response across studies, independent of pain or pain-free states. Standardization of HP induction protocols and outcome calculations is needed to ensure reproducibility and study comparison. Future HP studies may consider investigating sensory domains including nociception, which would further our understanding of abnormal HP regulation in pain conditions.
Collapse
Affiliation(s)
- Daniela M. Zolezzi
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Dennis B. Larsen
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Megan McPhee
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Thomas Graven-Nielsen
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
24
|
Huang L, Hardyman F, Edwards M, Galliano E. Deprivation-Induced Plasticity in the Early Central Circuits of the Rodent Visual, Auditory, and Olfactory Systems. eNeuro 2024; 11:ENEURO.0435-23.2023. [PMID: 38195533 PMCID: PMC11059429 DOI: 10.1523/eneuro.0435-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Activity-dependent neuronal plasticity is crucial for animals to adapt to dynamic sensory environments. Traditionally, it has been investigated using deprivation approaches in animal models primarily in sensory cortices. Nevertheless, emerging evidence emphasizes its significance in sensory organs and in subcortical regions where cranial nerves relay information to the brain. Additionally, critical questions started to arise. Do different sensory modalities share common cellular mechanisms for deprivation-induced plasticity at these central entry points? Does the deprivation duration correlate with specific plasticity mechanisms? This study systematically reviews and meta-analyzes research papers that investigated visual, auditory, or olfactory deprivation in rodents of both sexes. It examines the consequences of sensory deprivation in homologous regions at the first central synapse following cranial nerve transmission (vision - lateral geniculate nucleus and superior colliculus; audition - ventral and dorsal cochlear nucleus; olfaction - olfactory bulb). The systematic search yielded 91 papers (39 vision, 22 audition, 30 olfaction), revealing substantial heterogeneity in publication trends, experimental methods, measures of plasticity, and reporting across the sensory modalities. Despite these differences, commonalities emerged when correlating plasticity mechanisms with the duration of sensory deprivation. Short-term deprivation (up to 1 d) reduced activity and increased disinhibition, medium-term deprivation (1 d to a week) involved glial changes and synaptic remodeling, and long-term deprivation (over a week) primarily led to structural alterations. These findings underscore the importance of standardizing methodologies and reporting practices. Additionally, they highlight the value of cross-modal synthesis for understanding how the nervous system, including peripheral, precortical, and cortical areas, respond to and compensate for sensory inputs loss.
Collapse
Affiliation(s)
- Li Huang
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| | - Francesca Hardyman
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| | - Megan Edwards
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| | - Elisa Galliano
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB23EL Cambridge, United Kingdom
| |
Collapse
|
25
|
Jürgensen AM, Sakagiannis P, Schleyer M, Gerber B, Nawrot MP. Prediction error drives associative learning and conditioned behavior in a spiking model of Drosophila larva. iScience 2024; 27:108640. [PMID: 38292165 PMCID: PMC10824792 DOI: 10.1016/j.isci.2023.108640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 11/10/2023] [Accepted: 12/01/2023] [Indexed: 02/01/2024] Open
Abstract
Predicting reinforcement from sensory cues is beneficial for goal-directed behavior. In insect brains, underlying associations between cues and reinforcement, encoded by dopaminergic neurons, are formed in the mushroom body. We propose a spiking model of the Drosophila larva mushroom body. It includes a feedback motif conveying learned reinforcement expectation to dopaminergic neurons, which can compute prediction error as the difference between expected and present reinforcement. We demonstrate that this can serve as a driving force in learning. When combined with synaptic homeostasis, our model accounts for theoretically derived features of acquisition and loss of associations that depend on the intensity of the reinforcement and its temporal proximity to the cue. From modeling olfactory learning over the time course of behavioral experiments and simulating the locomotion of individual larvae toward or away from odor sources in a virtual environment, we conclude that learning driven by prediction errors can explain larval behavior.
Collapse
Affiliation(s)
- Anna-Maria Jürgensen
- Computational Systems Neuroscience, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| | - Panagiotis Sakagiannis
- Computational Systems Neuroscience, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| | - Michael Schleyer
- Leibniz Institute for Neurobiology (LIN), Department of Genetics, 39118 Magdeburg, Germany
- Institute for the Advancement of Higher Education, Faculty of Science, Hokkaido University, Sapporo 060-08080, Japan
| | - Bertram Gerber
- Leibniz Institute for Neurobiology (LIN), Department of Genetics, 39118 Magdeburg, Germany
- Institute for Biology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Brain and Behavioral Sciences (CBBS), Otto-von-Guericke University, 39118 Magdeburg, Germany
| | - Martin Paul Nawrot
- Computational Systems Neuroscience, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| |
Collapse
|
26
|
Zhang P, Yan J, Wei J, Li Y, Sun C. Disrupted synaptic homeostasis and partial occlusion of associative long-term potentiation in the human cortex during social isolation. J Affect Disord 2024; 344:207-218. [PMID: 37832738 DOI: 10.1016/j.jad.2023.10.080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
Social isolation often occurs in the military mission of soldiers but has increased in the general population since the COVID-19 epidemic. Overall synaptic homeostasis along with associative plasticity for the activity-dependent refinement of transmission across single synapses represent basic neural network function and adaptive behavior mechanisms. Here, we use electrophysiological and behavioral indices to non-invasively study the net synaptic strength and long-term potentiation (LTP)-like plasticity of humans in social isolation environments. The theta activity of electroencephalography (EEG) signals and transcranial magnetic stimulation (TMS) intensity to elicit a predefined amplitude of motor-evoked potential (MEP) demonstrate the disrupted synaptic homeostasis in the human cortex during social isolation. Furthermore, the induced MEP change by paired associative stimulation (PAS) demonstrates the partial occlusion of LTP-like plasticity, further behavior performances in a word-pair task are also identified as a potential index. Our study indicates that social isolation disrupts synaptic homeostasis and occludes associative LTP-like plasticity in the human cortex, decreasing behavior performance related to declarative memory.
Collapse
Affiliation(s)
- Peng Zhang
- School of Psychology, Beijing Key Laboratory of Learning and Cognition, Capital Normal University, Beijing 100048, China
| | - Juan Yan
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Jiao Wei
- The First Affiliated Hospital of Shandong First Medical University, Neurosurgery, Jinan 250013, China
| | - Yane Li
- College of Mathematics and Computer Science, Zhejiang A&F University, Hangzhou 311300, China
| | - Chuancai Sun
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100191, China; The First Affiliated Hospital of Shandong First Medical University, Nephrology, Jinan 250013, China.
| |
Collapse
|
27
|
Terzi F, Knabbe J, Cambridge SB. In Vivo Optical Interrogation of Neuronal Responses to Genetic, Cell Type-Specific Silencing. J Neurosci 2023; 43:8607-8620. [PMID: 37923378 PMCID: PMC10727181 DOI: 10.1523/jneurosci.2253-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/05/2023] [Accepted: 06/26/2023] [Indexed: 11/07/2023] Open
Abstract
We established a low background, Cre-dependent version of the inducible Tet-On system for fast, cell type-specific transgene expression in vivo Coexpression of a constitutive, Cre-dependent fluorescent marker selectively allowed single-cell analyses before and after inducible, Tet-dependent transgene expression. Here, we used this method for precise, acute manipulation of neuronal activity in the living brain. The goal was to study neuronal network homeostasis at cellular resolution. Single induction of the potassium channel Kir2.1 produced cell type-specific silencing within hours that lasted for at least 3 d. Longitudinal in vivo imaging of spontaneous calcium transients and neuronal morphology demonstrated that prolonged silencing did not alter spine densities or synaptic input strength. Furthermore, selective induction of Kir2.1 in parvalbumin interneurons increased the activity of surrounding neurons in a distance-dependent manner. This high-resolution, inducible interference and interval imaging of individual cells (high I5, HighFive) method thus allows visualizing temporally precise, genetic perturbations of defined cells.SIGNIFICANCE STATEMENT Gene function is studied by KO or overexpression of a specific gene followed by analyses of phenotypic changes. However, being able to predict and analyze exactly those cells in which genetic manipulation will occur is not possible. We combined two prominent transgene overexpression methods to fluorescently highlight the targeted cells appropriately before cell type-specific transgene induction. By inducing a potassium channel that decreases neuronal firing, we investigated how neuronal networks in the living mouse brain possibly compensate swift changes in cellular activities. Unlike in vitro, known compensatory homeostatic mechanisms, such as changes in synapses, were not observed in vivo Overall, we demonstrated with our method rapid genetic manipulation and analysis of neuronal activities as well as precision transgene expression.
Collapse
Affiliation(s)
- Firat Terzi
- Heidelberg University, Heidelberg, 69120, Germany
| | | | - Sidney B Cambridge
- Heidelberg University, Heidelberg, 69120, Germany
- Dr. Senckenberg Anatomy, Institute for Anatomy II, Goethe-University Frankfurt am Main, Frankfurt, 60590, Germany
| |
Collapse
|
28
|
Wittkopf PG, Boye Larsen D, Gregoret L, Graven-Nielsen T. Disrupted Cortical Homeostatic Plasticity Due to Prolonged Capsaicin-induced Pain. Neuroscience 2023; 533:1-9. [PMID: 37774909 DOI: 10.1016/j.neuroscience.2023.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/08/2023] [Accepted: 09/20/2023] [Indexed: 10/01/2023]
Abstract
Homeostatic plasticity (HP) regulates cortical excitability (CE) stability but is disrupted in persistent pain conditions. This study investigated how prolonged experimental pain affects HP and if pain relief modulates disrupted HP. Twenty-four healthy participants were randomised into a PainRelief or NoPainRelief group and attended four sessions; two sessions on consecutive days, separated by two weeks. Transcranial magnetic stimulation motor-evoked potentials reflecting CE and quantitative sensory testing (QST) measures were recorded. A capsaicin (pain condition) or placebo (control condition) patch was applied to the hand. HP was induced by cathodal-cathodal transcranial direct current stimulation (HP1) with CE assessment before and after. The PainRelief group had ice applied to the patch, while the NoPainRelief group waited for five minutes; subsequently another HP induction (HP2) and CE assessment were performed. After 24 h with the patch on, HP induction (HP3), QST, and CE recordings were repeated. Capsaicin reduced CE and the pain condition showed disrupted homeostatic responses at all time points (HP1: showed CE inhibition instead of facilitation; HP2 & HP3: lack of CE facilitation). Conversely, homeostatic responses were induced at all time points for the placebo condition. Capsaicin pain disrupts HP which is not restored by ice-induced pain relief. Future research may explore the prevention of HP disruption by targeting capsaicin-induced nociception but not pain perception.
Collapse
Affiliation(s)
- Priscilla Geraldine Wittkopf
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark
| | - Dennis Boye Larsen
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark
| | - Luisina Gregoret
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark
| | - Thomas Graven-Nielsen
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark.
| |
Collapse
|
29
|
Kim HJ, Hwang B, Reva M, Lee J, Lee BE, Lee Y, Cho EJ, Jeong M, Lee SE, Myung K, Baik JH, Park JH, Kim JI. GABAergic-like dopamine synapses in the brain. Cell Rep 2023; 42:113239. [PMID: 37819757 DOI: 10.1016/j.celrep.2023.113239] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/18/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023] Open
Abstract
Dopamine synapses play a crucial role in volitional movement and reward-related behaviors, while dysfunction of dopamine synapses causes various psychiatric and neurological disorders. Despite this significance, the true biological nature of dopamine synapses remains poorly understood. Here, we show that dopamine transmission is strongly correlated with GABA co-transmission across the brain and dopamine synapses are structured and function like GABAergic synapses with marked regional heterogeneity. In addition, GABAergic-like dopamine synapses are clustered on the dendrites, and GABA transmission at dopamine synapses has distinct physiological properties. Interestingly, the knockdown of neuroligin-2, a key postsynaptic protein at GABAergic synapses, unexpectedly does not weaken GABA co-transmission but instead facilitates it at dopamine synapses in the striatal neurons. More importantly, the attenuation of GABA co-transmission precedes deficits in dopaminergic transmission in animal models of Parkinson's disease. Our findings reveal the spatial and functional nature of GABAergic-like dopamine synapses in health and disease.
Collapse
Affiliation(s)
- Hyun-Jin Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Byungjae Hwang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Maria Reva
- Institut Pasteur, Unit of Synapse and Circuit Dynamics, CNRS UMR, 3571 Paris, France; Sorbonne University, ED3C, Paris, France
| | - Jieun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Byeong Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Youngeun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eun Jeong Cho
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Minseok Jeong
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Kyungjae Myung
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea; Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Ja-Hyun Baik
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Jung-Hoon Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| |
Collapse
|
30
|
Sharif NA. Electrical, Electromagnetic, Ultrasound Wave Therapies, and Electronic Implants for Neuronal Rejuvenation, Neuroprotection, Axonal Regeneration, and IOP Reduction. J Ocul Pharmacol Ther 2023; 39:477-498. [PMID: 36126293 DOI: 10.1089/jop.2022.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The peripheral nervous system (PNS) of mammals and nervous systems of lower organisms possess significant regenerative potential. In contrast, although neural plasticity can provide some compensation, the central nervous system (CNS) neurons and nerves of adult mammals generally fail to regenerate after an injury or damage. However, use of diverse electrical, electromagnetic and sonographic energy waves are illuminating novel ways to stimulate neuronal differentiation, proliferation, neurite growth, and axonal elongation/regeneration leading to various levels of functional recovery in animals and humans afflicted with disorders of the CNS, PNS, retina, and optic nerve. Tools such as acupuncture, electroacupuncture, electroshock therapy, electrical stimulation, transcranial magnetic stimulation, red light therapy, and low-intensity pulsed ultrasound therapy are demonstrating efficacy in treating many different maladies. These include wound healing, partial recovery from motor dysfunctions, recovery from ischemic/reperfusion insults and CNS and ocular remyelination, retinal ganglion cell (RGC) rejuvenation, and RGC axonal regeneration. Neural rejuvenation and axonal growth/regeneration processes involve activation or intensifying of the intrinsic bioelectric waves (action potentials) that exist in every neuronal circuit of the body. In addition, reparative factors released at the nerve terminals and via neuronal dendrites (transmitter substances), extracellular vesicles containing microRNAs and neurotrophins, and intercellular communication occurring via nanotubes aid in reestablishing lost or damaged connections between the traumatized tissues and the PNS and CNS. Many other beneficial effects of the aforementioned treatment paradigms are mediated via gene expression alterations such as downregulation of inflammatory and death-signal genes and upregulation of neuroprotective and cytoprotective genes. These varied techniques and technologies will be described and discussed covering cell-based and animal model-based studies. Data from clinical applications and linkage to human ocular diseases will also be discussed where relevant translational research has been reported.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, California, USA
- Singapore Eye Research Institute (SERI), Singapore
- SingHealth Duke-NUS Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
- Department of Surgery and Cancer, Imperial College of Science and Technology, London, United Kingdom
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA
- Department of Pharmacy Sciences, Creighton University, Omaha, Nebraska, USA
- Insitute of Ophthalmology, University College London (UCL), London, United Kingdom
| |
Collapse
|
31
|
Ramesh N, Escher M, Turrel O, Lützkendorf J, Matkovic T, Liu F, Sigrist SJ. An antagonism between Spinophilin and Syd-1 operates upstream of memory-promoting presynaptic long-term plasticity. eLife 2023; 12:e86084. [PMID: 37767892 PMCID: PMC10588984 DOI: 10.7554/elife.86084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
We still face fundamental gaps in understanding how molecular plastic changes of synapses intersect with circuit operation to define behavioral states. Here, we show that an antagonism between two conserved regulatory proteins, Spinophilin (Spn) and Syd-1, controls presynaptic long-term plasticity and the maintenance of olfactory memories in Drosophila. While Spn mutants could not trigger nanoscopic active zone remodeling under homeostatic challenge and failed to stably potentiate neurotransmitter release, concomitant reduction of Syd-1 rescued all these deficits. The Spn/Syd-1 antagonism converged on active zone close F-actin, and genetic or acute pharmacological depolymerization of F-actin rescued the Spn deficits by allowing access to synaptic vesicle release sites. Within the intrinsic mushroom body neurons, the Spn/Syd-1 antagonism specifically controlled olfactory memory stabilization but not initial learning. Thus, this evolutionarily conserved protein complex controls behaviorally relevant presynaptic long-term plasticity, also observed in the mammalian brain but still enigmatic concerning its molecular mechanisms and behavioral relevance.
Collapse
Affiliation(s)
- Niraja Ramesh
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | - Marc Escher
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | - Oriane Turrel
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | | | - Tanja Matkovic
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | - Fan Liu
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| |
Collapse
|
32
|
Han Y, Goel P, Chen J, Perry S, Tran N, Nishimura S, Sanjani M, Chien C, Dickman D. Excess glutamate release triggers subunit-specific homeostatic receptor scaling. Cell Rep 2023; 42:112775. [PMID: 37436892 PMCID: PMC10529671 DOI: 10.1016/j.celrep.2023.112775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/06/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023] Open
Abstract
Ionotropic glutamate receptors (GluRs) are targets for modulation in Hebbian and homeostatic synaptic plasticity and are remodeled by development, experience, and disease. We have probed the impact of synaptic glutamate levels on the two postsynaptic GluR subtypes at the Drosophila neuromuscular junction, GluRA and GluRB. We first demonstrate that GluRA and GluRB compete to establish postsynaptic receptive fields, and that proper GluR abundance and composition can be orchestrated in the absence of any synaptic glutamate release. However, excess glutamate adaptively tunes postsynaptic GluR abundance, echoing GluR scaling observed in mammalian systems. Furthermore, when GluRA vs. GluRB competition is eliminated, GluRB becomes insensitive to glutamate modulation. In contrast, GluRA is now homeostatically regulated by excess glutamate to maintain stable miniature activity, where Ca2+ permeability through GluRA receptors is required. Thus, excess glutamate, GluR competition, and Ca2+ signaling collaborate to selectively target GluR subtypes for homeostatic regulation at postsynaptic compartments.
Collapse
Affiliation(s)
- Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jiawen Chen
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Nancy Tran
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Samantha Nishimura
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Manisha Sanjani
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Chun Chien
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
33
|
Gromova KV, Thies E, Janiesch PC, Lützenkirchen FP, Zhu Y, Stajano D, Dürst CD, Schweizer M, Konietzny A, Mikhaylova M, Gee CE, Kneussel M. The kinesin Kif21b binds myosin Va and mediates changes in actin dynamics underlying homeostatic synaptic downscaling. Cell Rep 2023; 42:112743. [PMID: 37418322 DOI: 10.1016/j.celrep.2023.112743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 07/09/2023] Open
Abstract
Homeostatic synaptic plasticity adjusts the strength of synapses to restrain neuronal activity within a physiological range. Postsynaptic guanylate kinase-associated protein (GKAP) controls the bidirectional synaptic scaling of AMPA receptors (AMPARs); however, mechanisms by which chronic activity triggers cytoskeletal remodeling to downscale synaptic transmission are barely understood. Here, we report that the microtubule-dependent kinesin motor Kif21b binds GKAP and likewise is located in dendritic spines in a myosin Va- and neuronal-activity-dependent manner. Kif21b depletion unexpectedly alters actin dynamics in spines, and adaptation of actin turnover following chronic activity is lost in Kif21b-knockout neurons. Consistent with a role of the kinesin in regulating actin dynamics, Kif21b overexpression promotes actin polymerization. Moreover, Kif21b controls GKAP removal from spines and the decrease of GluA2-containing AMPARs from the neuronal surface, thereby inducing homeostatic synaptic downscaling. Our data highlight a critical role of Kif21b at the synaptic actin cytoskeleton underlying homeostatic scaling of neuronal firing.
Collapse
Affiliation(s)
- Kira V Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Philipp C Janiesch
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Felix P Lützenkirchen
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Yipeng Zhu
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Daniele Stajano
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Céline D Dürst
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michaela Schweizer
- Core Facility Morphology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anja Konietzny
- RG Neuronal Protein Transport, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marina Mikhaylova
- RG Neuronal Protein Transport, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, 10099 Berlin, Germany
| | - Christine E Gee
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Hamburg Center of Neuroscience, HCNS, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
34
|
Sawicki J, Berner R, Loos SAM, Anvari M, Bader R, Barfuss W, Botta N, Brede N, Franović I, Gauthier DJ, Goldt S, Hajizadeh A, Hövel P, Karin O, Lorenz-Spreen P, Miehl C, Mölter J, Olmi S, Schöll E, Seif A, Tass PA, Volpe G, Yanchuk S, Kurths J. Perspectives on adaptive dynamical systems. CHAOS (WOODBURY, N.Y.) 2023; 33:071501. [PMID: 37486668 DOI: 10.1063/5.0147231] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/24/2023] [Indexed: 07/25/2023]
Abstract
Adaptivity is a dynamical feature that is omnipresent in nature, socio-economics, and technology. For example, adaptive couplings appear in various real-world systems, such as the power grid, social, and neural networks, and they form the backbone of closed-loop control strategies and machine learning algorithms. In this article, we provide an interdisciplinary perspective on adaptive systems. We reflect on the notion and terminology of adaptivity in different disciplines and discuss which role adaptivity plays for various fields. We highlight common open challenges and give perspectives on future research directions, looking to inspire interdisciplinary approaches.
Collapse
Affiliation(s)
- Jakub Sawicki
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
- Akademie Basel, Fachhochschule Nordwestschweiz FHNW, Leonhardsstrasse 6, 4009 Basel, Switzerland
| | - Rico Berner
- Department of Physics, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| | - Sarah A M Loos
- DAMTP, University of Cambridge, Wilberforce Road, Cambridge CB3 0WA, United Kingdom
| | - Mehrnaz Anvari
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
- Fraunhofer Institute for Algorithms and Scientific Computing, Schloss Birlinghoven, 53757 Sankt-Augustin, Germany
| | - Rolf Bader
- Institute of Systematic Musicology, University of Hamburg, Hamburg, Germany
| | - Wolfram Barfuss
- Transdisciplinary Research Area: Sustainable Futures, University of Bonn, 53113 Bonn, Germany
- Center for Development Research (ZEF), University of Bonn, 53113 Bonn, Germany
| | - Nicola Botta
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
- Department of Computer Science and Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Nuria Brede
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
- Department of Computer Science, University of Potsdam, An der Bahn 2, 14476 Potsdam, Germany
| | - Igor Franović
- Scientific Computing Laboratory, Center for the Study of Complex Systems, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia
| | - Daniel J Gauthier
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
| | - Sebastian Goldt
- Department of Physics, International School of Advanced Studies (SISSA), Trieste, Italy
| | - Aida Hajizadeh
- Research Group Comparative Neuroscience, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Philipp Hövel
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
| | - Omer Karin
- Department of Mathematics, Imperial College London, London SW7 2AZ, United Kingdom
| | - Philipp Lorenz-Spreen
- Center for Adaptive Rationality, Max Planck Institute for Human Development, Lentzeallee 94, 14195 Berlin, Germany
| | - Christoph Miehl
- Akademie Basel, Fachhochschule Nordwestschweiz FHNW, Leonhardsstrasse 6, 4009 Basel, Switzerland
| | - Jan Mölter
- Department of Mathematics, School of Computation, Information and Technology, Technical University of Munich, Boltzmannstraße 3, 85748 Garching bei München, Germany
| | - Simona Olmi
- Akademie Basel, Fachhochschule Nordwestschweiz FHNW, Leonhardsstrasse 6, 4009 Basel, Switzerland
| | - Eckehard Schöll
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
- Akademie Basel, Fachhochschule Nordwestschweiz FHNW, Leonhardsstrasse 6, 4009 Basel, Switzerland
| | - Alireza Seif
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois 60637, USA
| | - Peter A Tass
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Giovanni Volpe
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Serhiy Yanchuk
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
- Department of Physics, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| | - Jürgen Kurths
- Potsdam Institute for Climate Impact Research, Telegrafenberg, 14473 Potsdam, Germany
- Department of Physics, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| |
Collapse
|
35
|
Schröder JK, Abdel-Hafiz L, Ali AAH, Cousin TC, Hallenberger J, Rodrigues Almeida F, Anstötz M, Lenz M, Vlachos A, von Gall C, Tundo-Lavalle F. Effects of the Light/Dark Phase and Constant Light on Spatial Working Memory and Spine Plasticity in the Mouse Hippocampus. Cells 2023; 12:1758. [PMID: 37443792 PMCID: PMC10340644 DOI: 10.3390/cells12131758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Circadian rhythms in behavior and physiology such as rest/activity and hormones are driven by an internal clock and persist in the absence of rhythmic environmental cues. However, the period and phase of the internal clock are entrained by the environmental light/dark cycle. Consequently, aberrant lighting conditions, which are increasing in modern society, have a strong impact on rhythmic body and brain functions. Mice were exposed to three different lighting conditions, 12 h light/12 h dark cycle (LD), constant darkness (DD), and constant light (LL), to study the effects of the light/dark cycle and aberrant lighting on the hippocampus, a critical structure for temporal and spatial memory formation and navigation. Locomotor activity and plasma corticosterone levels were analyzed as readouts for circadian rhythms. Spatial working memory via Y-maze, spine morphology of Golgi-Cox-stained hippocampi, and plasticity of excitatory synapses, measured by number and size of synaptopodin and GluR1-immunreactive clusters, were analyzed. Our results indicate that the light/dark cycle drives diurnal differences in synaptic plasticity in hippocampus. Moreover, spatial working memory, spine density, and size and number of synaptopodin and GluR1 clusters were reduced in LL, while corticosterone levels were increased. This indicates that acute constant light affects hippocampal function and synaptic plasticity.
Collapse
Affiliation(s)
- Jane K. Schröder
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
- Department of Pediatric Hematology and Oncology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Laila Abdel-Hafiz
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Amira A. H. Ali
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, El-Gomhoria St. 1, Mansoura 35516, Egypt
| | - Teresa C. Cousin
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Johanna Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Filipe Rodrigues Almeida
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Max Anstötz
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Maximilian Lenz
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany;
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany;
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany;
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Federica Tundo-Lavalle
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| |
Collapse
|
36
|
Curia G. Hebbian and homeostatic synaptic plasticity of AMPA receptors in epileptogenesis. Cell Rep Med 2023; 4:101047. [PMID: 37196628 DOI: 10.1016/j.xcrm.2023.101047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023]
Abstract
AMPA receptors' synaptic plasticity is involved in epileptogenesis. In this issue, Eiro et al.1 demonstrate that Hebbian plasticity is responsible for increased AMPAR in focal seizures, while homeostatic plasticity induces the reduction of AMPAR in generalized onset seizures.
Collapse
Affiliation(s)
- Giulia Curia
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
37
|
Lenz M, Eichler A, Kruse P, Stöhr P, Kleidonas D, Galanis C, Lu H, Vlachos A. Denervated mouse CA1 pyramidal neurons express homeostatic synaptic plasticity following entorhinal cortex lesion. Front Mol Neurosci 2023; 16:1148219. [PMID: 37122623 PMCID: PMC10130538 DOI: 10.3389/fnmol.2023.1148219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/14/2023] [Indexed: 05/02/2023] Open
Abstract
Structural, functional, and molecular reorganization of denervated neural networks is often observed in neurological conditions. The loss of input is accompanied by homeostatic synaptic adaptations, which can affect the reorganization process. A major challenge of denervation-induced homeostatic plasticity operating in complex neural networks is the specialization of neuronal inputs. It remains unclear whether neurons respond similarly to the loss of distinct inputs. Here, we used in vitro entorhinal cortex lesion (ECL) and Schaffer collateral lesion (SCL) in mouse organotypic entorhino-hippocampal tissue cultures to study denervation-induced plasticity of CA1 pyramidal neurons. We observed microglia accumulation, presynaptic bouton degeneration, and a reduction in dendritic spine numbers in the denervated layers 3 days after SCL and ECL. Transcriptome analysis of the CA1 region revealed complex changes in differential gene expression following SCL and ECL compared to non-lesioned controls with a specific enrichment of differentially expressed synapse-related genes observed after ECL. Consistent with this finding, denervation-induced homeostatic plasticity of excitatory synapses was observed 3 days after ECL but not after SCL. Chemogenetic silencing of the EC but not CA3 confirmed the pathway-specific induction of homeostatic synaptic plasticity in CA1. Additionally, increased RNA oxidation was observed after SCL and ECL. These results reveal important commonalities and differences between distinct pathway lesions and demonstrate a pathway-specific induction of denervation-induced homeostatic synaptic plasticity.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Phyllis Stöhr
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dimitrios Kleidonas
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Han Lu
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
38
|
Wang Y, Lin J, Li J, Yan L, Li W, He X, Ma H. Chronic Neuronal Inactivity Utilizes the mTOR-TFEB Pathway to Drive Transcription-Dependent Autophagy for Homeostatic Up-Scaling. J Neurosci 2023; 43:2631-2652. [PMID: 36868861 PMCID: PMC10089247 DOI: 10.1523/jneurosci.0146-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
Activity-dependent changes in protein expression are critical for neuronal plasticity, a fundamental process for the processing and storage of information in the brain. Among the various forms of plasticity, homeostatic synaptic up-scaling is unique in that it is induced primarily by neuronal inactivity. However, precisely how the turnover of synaptic proteins occurs in this homeostatic process remains unclear. Here, we report that chronically inhibiting neuronal activity in primary cortical neurons prepared from embryonic day (E)18 Sprague Dawley rats (both sexes) induces autophagy, thereby regulating key synaptic proteins for up-scaling. Mechanistically, chronic neuronal inactivity causes dephosphorylation of ERK and mTOR, which induces transcription factor EB (TFEB)-mediated cytonuclear signaling and drives transcription-dependent autophagy to regulate αCaMKII and PSD95 during synaptic up-scaling. Together, these findings suggest that mTOR-dependent autophagy, which is often triggered by metabolic stressors such as starvation, is recruited and sustained during neuronal inactivity to maintain synaptic homeostasis, a process that ensures proper brain function and if impaired can cause neuropsychiatric disorders such as autism.SIGNIFICANCE STATEMENT In the mammalian brain, protein turnover is tightly controlled by neuronal activation to ensure key neuronal functions during long-lasting synaptic plasticity. However, a long-standing question is how this process occurs during synaptic up-scaling, a process that requires protein turnover but is induced by neuronal inactivation. Here, we report that mTOR-dependent signaling, which is often triggered by metabolic stressors such as starvation, is "hijacked" by chronic neuronal inactivation, which then serves as a nucleation point for transcription factor EB (TFEB) cytonuclear signaling that drives transcription-dependent autophagy for up-scaling. These results provide the first evidence of a physiological role of mTOR-dependent autophagy in enduing neuronal plasticity, thereby connecting major themes in cell biology and neuroscience via a servo loop that mediates autoregulation in the brain.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Jingran Lin
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Jiarui Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Lu Yan
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Wenwen Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
- Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing 100050, China
| |
Collapse
|
39
|
Yamakou ME, Kuehn C. Combined effects of spike-timing-dependent plasticity and homeostatic structural plasticity on coherence resonance. Phys Rev E 2023; 107:044302. [PMID: 37198865 DOI: 10.1103/physreve.107.044302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/23/2023] [Indexed: 05/19/2023]
Abstract
Efficient processing and transfer of information in neurons have been linked to noise-induced resonance phenomena such as coherence resonance (CR), and adaptive rules in neural networks have been mostly linked to two prevalent mechanisms: spike-timing-dependent plasticity (STDP) and homeostatic structural plasticity (HSP). Thus this paper investigates CR in small-world and random adaptive networks of Hodgkin-Huxley neurons driven by STDP and HSP. Our numerical study indicates that the degree of CR strongly depends, and in different ways, on the adjusting rate parameter P, which controls STDP, on the characteristic rewiring frequency parameter F, which controls HSP, and on the parameters of the network topology. In particular, we found two robust behaviors. (i) Decreasing P (which enhances the weakening effect of STDP on synaptic weights) and decreasing F (which slows down the swapping rate of synapses between neurons) always leads to higher degrees of CR in small-world and random networks, provided that the synaptic time delay parameter τ_{c} has some appropriate values. (ii) Increasing the synaptic time delay τ_{c} induces multiple CR (MCR)-the occurrence of multiple peaks in the degree of coherence as τ_{c} changes-in small-world and random networks, with MCR becoming more pronounced at smaller values of P and F. Our results imply that STDP and HSP can jointly play an essential role in enhancing the time precision of firing necessary for optimal information processing and transfer in neural systems and could thus have applications in designing networks of noisy artificial neural circuits engineered to use CR to optimize information processing and transfer.
Collapse
Affiliation(s)
- Marius E Yamakou
- Department of Data Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 11, 91058 Erlangen, Germany
- Max-Planck-Institut für Mathematik in den Naturwissenschaften, Inselstr. 22, 04103 Leipzig, Germany
| | - Christian Kuehn
- Faculty of Mathematics, Technical University of Munich, Boltzmannstrasse 3, 85748 Garching bei München, Germany
- Complexity Science Hub Vienna, Josefstädter Strasse 39, 1080 Vienna, Austria
| |
Collapse
|
40
|
Plautz EJ, Barbay S, Frost SB, Stowe AM, Dancause N, Zoubina EV, Eisner-Janowicz I, Guggenmos DJ, Nudo RJ. Spared Premotor Areas Undergo Rapid Nonlinear Changes in Functional Organization Following a Focal Ischemic Infarct in Primary Motor Cortex of Squirrel Monkeys. J Neurosci 2023; 43:2021-2032. [PMID: 36788028 PMCID: PMC10027035 DOI: 10.1523/jneurosci.1452-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 02/16/2023] Open
Abstract
Recovery of motor function after stroke is accompanied by reorganization of movement representations in spared cortical motor regions. It is widely assumed that map reorganization parallels recovery, suggesting a causal relationship. We examined this assumption by measuring changes in motor representations in eight male and six female squirrel monkeys in the first few weeks after injury, a time when motor recovery is most rapid. Maps of movement representations were derived using intracortical microstimulation techniques in primary motor cortex (M1), ventral premotor cortex (PMv), and dorsal premotor cortex (PMd) in 14 adult squirrel monkeys before and after a focal infarct in the M1 distal forelimb area. Maps were derived at baseline and at either 2 (n = 7) or 3 weeks (n = 7) postinfarct. In PMv the forelimb maps remained unchanged at 2 weeks but contracted significantly (-42.4%) at 3 weeks. In PMd the forelimb maps expanded significantly (+110.6%) at 2 weeks but contracted significantly (-57.4%) at 3 weeks. Motor deficits were equivalent at both time points. These results highlight two features of plasticity after M1 lesions. First, significant contraction of distal forelimb motor maps in both PMv and PMd is evident by 3 weeks. Second, an unpredictable nonlinear pattern of reorganization occurs in the distal forelimb representation in PMd, first expanding at 2 weeks, and then contracting at 3 weeks postinjury. Together with previous results demonstrating reliable map expansions in PMv several weeks to months after M1 injury, the subacute time period may represent a critical window for the timing of therapeutic interventions.SIGNIFICANCE STATEMENT The relationship between motor recovery and motor map reorganization after cortical injury has rarely been examined in acute/subacute periods. In nonhuman primates, premotor maps were examined at 2 and 3 weeks after injury to primary motor cortex. Although maps are known to expand late after injury, the present study demonstrates early map expansion at 2 weeks (dorsal premotor cortex) followed by contraction at 3 weeks (dorsal and ventral premotor cortex). This nonlinear map reorganization during a time of gradual behavioral recovery suggests that the relationship between map plasticity and motor recovery is much more complex than previously thought. It also suggests that rehabilitative motor training may have its most potent effects during this early dynamic phase of map reorganization.
Collapse
Affiliation(s)
- Erik J Plautz
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
- Landon Center on Aging, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Scott Barbay
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
- Landon Center on Aging, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Shawn B Frost
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
- Landon Center on Aging, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Ann M Stowe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Numa Dancause
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Elena V Zoubina
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
- Landon Center on Aging, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Ines Eisner-Janowicz
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - David J Guggenmos
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Randolph J Nudo
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
- Landon Center on Aging, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
41
|
Djemil S, Sames AM, Pak DTS. ACh Transfers: Homeostatic Plasticity of Cholinergic Synapses. Cell Mol Neurobiol 2023; 43:697-709. [PMID: 35643882 PMCID: PMC11415198 DOI: 10.1007/s10571-022-01227-2] [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: 01/14/2022] [Accepted: 04/25/2022] [Indexed: 11/03/2022]
Abstract
The field of homeostatic plasticity continues to advance rapidly, highlighting the importance of stabilizing neuronal activity within functional limits in the context of numerous fundamental processes such as development, learning, and memory. Most homeostatic plasticity studies have been focused on glutamatergic synapses, while the rules that govern homeostatic regulation of other synapse types are less understood. While cholinergic synapses have emerged as a critical component in the etiology of mammalian neurodegenerative disease mechanisms, relatively few studies have been conducted on the homeostatic plasticity of such synapses, particularly in the mammalian nervous system. An exploration of homeostatic mechanisms at the cholinergic synapse may illuminate potential therapeutic targets for disease management and treatment. We will review cholinergic homeostatic plasticity in the mammalian neuromuscular junction, the autonomic nervous system, central synapses, and in relation to pathological conditions including Alzheimer disease and DYT1 dystonia. This work provides a historical context for the field of cholinergic homeostatic regulation by examining common themes, unique features, and outstanding questions associated with these distinct cholinergic synapse types and aims to inform future research in the field.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Antonia M Sames
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
42
|
Arora Y, Dutta A. Perspective: Disentangling the effects of tES on neurovascular unit. Front Neurol 2023; 13:1038700. [PMID: 36698881 PMCID: PMC9868757 DOI: 10.3389/fneur.2022.1038700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/28/2022] [Indexed: 01/11/2023] Open
Abstract
Transcranial electrical stimulation (tES) can modulate the neurovascular unit, including the perivascular space morphology, but the mechanisms are unclear. In this perspective article, we used an open-source "rsHRF toolbox" and an open-source functional magnetic resonance imaging (fMRI) transcranial direct current stimulation (tDCS) data set to show the effects of tDCS on the temporal profile of the haemodynamic response function (HRF). We investigated the effects of tDCS in the gray matter and at three regions of interest in the gray matter, namely, the anodal electrode (FC5), cathodal electrode (FP2), and an independent site remote from the electrodes (PZ). A "canonical HRF" with time and dispersion derivatives and a finite impulse response (FIR) model with three parameters captured the effects of anodal tDCS on the temporal profile of the HRF. The FIR model showed tDCS onset effects on the temporal profile of HRF for verum and sham tDCS conditions that were different from the no tDCS condition, which questions the validity of the sham tDCS (placebo). Here, we postulated that the effects of tDCS onset on the temporal profile of HRF are subserved by the effects on neurovascular coupling. We provide our perspective based on previous work on tES effects on the neurovascular unit, including mechanistic grey-box modeling of the effects of tES on the vasculature that can facilitate model predictive control (MPC). Future studies need to investigate grey-box modeling of online effects of tES on the neurovascular unit, including perivascular space, neurometabolic coupling, and neurovascular coupling, that can facilitate MPC of the tES dose-response to address the momentary ("state") and phenotypic ("trait") factors.
Collapse
Affiliation(s)
- Yashika Arora
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurugram, India
| | - Anirban Dutta
- School of Engineering, University of Lincoln, Lincoln, United Kingdom
| |
Collapse
|
43
|
Armstrong NS, Frank CA. The calcineurin regulator Sarah enables distinct forms of homeostatic plasticity at the Drosophila neuromuscular junction. Front Synaptic Neurosci 2023; 14:1033743. [PMID: 36685082 PMCID: PMC9846150 DOI: 10.3389/fnsyn.2022.1033743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: The ability of synapses to maintain physiological levels of evoked neurotransmission is essential for neuronal stability. A variety of perturbations can disrupt neurotransmission, but synapses often compensate for disruptions and work to stabilize activity levels, using forms of homeostatic synaptic plasticity. Presynaptic homeostatic potentiation (PHP) is one such mechanism. PHP is expressed at the Drosophila melanogaster larval neuromuscular junction (NMJ) synapse, as well as other NMJs. In PHP, presynaptic neurotransmitter release increases to offset the effects of impairing muscle transmitter receptors. Prior Drosophila work has studied PHP using different ways to perturb muscle receptor function-either acutely (using pharmacology) or chronically (using genetics). Some of our prior data suggested that cytoplasmic calcium signaling was important for expression of PHP after genetic impairment of glutamate receptors. Here we followed up on that observation. Methods: We used a combination of transgenic Drosophila RNA interference and overexpression lines, along with NMJ electrophysiology, synapse imaging, and pharmacology to test if regulators of the calcium/calmodulin-dependent protein phosphatase calcineurin are necessary for the normal expression of PHP. Results: We found that either pre- or postsynaptic dysregulation of a Drosophila gene regulating calcineurin, sarah (sra), blocks PHP. Tissue-specific manipulations showed that either increases or decreases in sra expression are detrimental to PHP. Additionally, pharmacologically and genetically induced forms of expression of PHP are functionally separable depending entirely upon which sra genetic manipulation is used. Surprisingly, dual-tissue pre- and postsynaptic sra knockdown or overexpression can ameliorate PHP blocks revealed in single-tissue experiments. Pharmacological and genetic inhibition of calcineurin corroborated this latter finding. Discussion: Our results suggest tight calcineurin regulation is needed across multiple tissue types to stabilize peripheral synaptic outputs.
Collapse
Affiliation(s)
- Noah S. Armstrong
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States
| | - C. Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States,*Correspondence: C. Andrew Frank
| |
Collapse
|
44
|
Creation of Neuronal Ensembles and Cell-Specific Homeostatic Plasticity through Chronic Sparse Optogenetic Stimulation. J Neurosci 2023; 43:82-92. [PMID: 36400529 PMCID: PMC9838708 DOI: 10.1523/jneurosci.1104-22.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/15/2022] [Accepted: 10/16/2022] [Indexed: 11/19/2022] Open
Abstract
Cortical computations emerge from the dynamics of neurons embedded in complex cortical circuits. Within these circuits, neuronal ensembles, which represent subnetworks with shared functional connectivity, emerge in an experience-dependent manner. Here we induced ensembles in ex vivo cortical circuits from mice of either sex by differentially activating subpopulations through chronic optogenetic stimulation. We observed a decrease in voltage correlation, and importantly a synaptic decoupling between the stimulated and nonstimulated populations. We also observed a decrease in firing rate during Up-states in the stimulated population. These ensemble-specific changes were accompanied by decreases in intrinsic excitability in the stimulated population, and a decrease in connectivity between stimulated and nonstimulated pyramidal neurons. By incorporating the empirically observed changes in intrinsic excitability and connectivity into a spiking neural network model, we were able to demonstrate that changes in both intrinsic excitability and connectivity accounted for the decreased firing rate, but only changes in connectivity accounted for the observed decorrelation. Our findings help ascertain the mechanisms underlying the ability of chronic patterned stimulation to create ensembles within cortical circuits and, importantly, show that while Up-states are a global network-wide phenomenon, functionally distinct ensembles can preserve their identity during Up-states through differential firing rates and correlations.SIGNIFICANCE STATEMENT The connectivity and activity patterns of local cortical circuits are shaped by experience. This experience-dependent reorganization of cortical circuits is driven by complex interactions between different local learning rules, external input, and reciprocal feedback between many distinct brain areas. Here we used an ex vivo approach to demonstrate how simple forms of chronic external stimulation can shape local cortical circuits in terms of their correlated activity and functional connectivity. The absence of feedback between different brain areas and full control of external input allowed for a tractable system to study the underlying mechanisms and development of a computational model. Results show that differential stimulation of subpopulations of neurons significantly reshapes cortical circuits and forms subnetworks referred to as neuronal ensembles.
Collapse
|
45
|
Xylaki M, Paiva I, Al-Azzani M, Gerhardt E, Jain G, Islam MR, Vasili E, Wassouf Z, Schulze-Hentrich JM, Fischer A, Outeiro TF. miR-101a-3p Impairs Synaptic Plasticity and Contributes to Synucleinopathy. JOURNAL OF PARKINSON'S DISEASE 2023; 13:179-196. [PMID: 36744345 PMCID: PMC10041420 DOI: 10.3233/jpd-225055] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Synucleinopathies are disorders characterized by the abnormal accumulation of α-synuclein (aSyn). Synaptic compromise is observed in synucleinopathies parallel to aSyn aggregation and is accompanied by transcript deregulation. OBJECTIVE We sought to identify microRNAs associated with synaptic processes that may contribute to synaptic dysfunction and degeneration in synucleinopathies. METHODS We performed small RNA-sequencing of midbrain from 6-month-old transgenic mice expressing A30P mutant aSyn, followed by comparative expression analysis. We then used real-time quantitative polymerase chain reaction (qPCR) for validation. Functional analysis was performed in primary neurons by biochemical assays and imaging. RESULTS We found several deregulated biological processes linked to the synapse. miR-101a-3p was validated as a synaptic miRNA upregulated in aSyn Tg mice and in the cortex of dementia with Lewy bodies patients. Mice and primary cultured neurons overexpressing miR-101a-3p showed downregulation of postsynaptic proteins GABA Ab2 and SAPAP3 and altered dendritic morphology resembling synaptic plasticity impairments and/or synaptic damage. Interestingly, primary cultured neuron exposure to recombinant wild-type aSyn species efficiently increased miR-101a-3p levels. Finally, a dynamic role of miR-101a-3p in synapse plasticity was shown by identifying downregulation of miR-101a-3p in a condition of enhanced synaptic plasticity modelled in Wt animals housed in enriched environment. CONCLUSION To conclude, we correlated pathologic aSyn with high levels of miR-101a-3p and a novel dynamic role of the miRNA in synaptic plasticity.
Collapse
Affiliation(s)
- Mary Xylaki
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Isabel Paiva
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
- Present address: Laboratory of Cognitive and Adaptive Neuroscience, UMR 7364 (CNRS/ Strasbourg University), Strasbourg, France
| | - Mohammed Al-Azzani
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Ellen Gerhardt
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Gaurav Jain
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Md Rezaul Islam
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Eftychia Vasili
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | - Zinah Wassouf
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
| | | | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Centre, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
- Scientific employee with an honorary contract at German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
46
|
Nayak M, Das D, Pradhan J, Ahmed R, Laureano-Melo R, Dandapat J. Epigenetic signature in neural plasticity: the journey so far and journey ahead. Heliyon 2022; 8:e12292. [PMID: 36590572 PMCID: PMC9798197 DOI: 10.1016/j.heliyon.2022.e12292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/31/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Neural plasticity is a remarkable characteristic of the brain which allows neurons to rewire their structure in response to internal and external stimuli. Many external stimuli collectively referred to as 'epigenetic factors' strongly influence structural and functional reorganization of the brain, thereby acting as a potential driver of neural plasticity. DNA methylation and demethylation, histone acetylation, and deacetylation are some of the frontline epigenetic mechanisms behind neural plasticity. Epigenetic signature molecules (mostly proteins) play a pivotal role in epigenetic reprogramming. Though neuro-epigenetics is an incredibly important field of emerging research, the critical role of signature proteins associated with epigenetic alteration and their involvement in neural plasticity needs further attention. This study gives an integrated and systematic overview of the current state of knowledge with a clear idea of types of neural plasticity and the context-dependent role of epigenetic signature molecules and their modulation by some natural bioactive compounds.
Collapse
Affiliation(s)
- Madhusmita Nayak
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Diptimayee Das
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Faculty of Allied Health Science, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai India
| | - Jyotsnarani Pradhan
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Corresponding author.
| | - R.G. Ahmed
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Roberto Laureano-Melo
- Barra Mansa University Center, R. Ver. Pinho de Carvalho, 267, 27330-550, Barra Mansa, Rio de Janeiro, Brazil
| | - Jagneshwar Dandapat
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India,Corresponding author.
| |
Collapse
|
47
|
Perry S, Han Y, Qiu C, Chien C, Goel P, Nishimura S, Sajnani M, Schmid A, Sigrist SJ, Dickman D. A glutamate receptor C-tail recruits CaMKII to suppress retrograde homeostatic signaling. Nat Commun 2022; 13:7656. [PMID: 36496500 PMCID: PMC9741633 DOI: 10.1038/s41467-022-35417-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Presynaptic homeostatic plasticity (PHP) adaptively enhances neurotransmitter release following diminished postsynaptic glutamate receptor (GluR) functionality to maintain synaptic strength. While much is known about PHP expression mechanisms, postsynaptic induction remains enigmatic. For over 20 years, diminished postsynaptic Ca2+ influx was hypothesized to reduce CaMKII activity and enable retrograde PHP signaling at the Drosophila neuromuscular junction. Here, we have interrogated inductive signaling and find that active CaMKII colocalizes with and requires the GluRIIA receptor subunit. Next, we generated Ca2+-impermeable GluRs to reveal that both CaMKII activity and PHP induction are Ca2+-insensitive. Rather, a GluRIIA C-tail domain is necessary and sufficient to recruit active CaMKII. Finally, chimeric receptors demonstrate that the GluRIIA tail constitutively occludes retrograde homeostatic signaling by stabilizing active CaMKII. Thus, the physical loss of the GluRIIA tail is sensed, rather than reduced Ca2+, to enable retrograde PHP signaling, highlighting a unique, Ca2+-independent control mechanism for CaMKII in gating homeostatic plasticity.
Collapse
Affiliation(s)
- Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Chengjie Qiu
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Chun Chien
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Samantha Nishimura
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Manisha Sajnani
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Andreas Schmid
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195, Berlin, Germany
- Faculty of Life Sciences, Albstadt-Sigmaringen University, Sigmaringen, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
48
|
Anderson MA, Squair JW, Gautier M, Hutson TH, Kathe C, Barraud Q, Bloch J, Courtine G. Natural and targeted circuit reorganization after spinal cord injury. Nat Neurosci 2022; 25:1584-1596. [PMID: 36396975 DOI: 10.1038/s41593-022-01196-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/05/2022] [Indexed: 11/18/2022]
Abstract
A spinal cord injury disrupts communication between the brain and the circuits in the spinal cord that regulate neurological functions. The consequences are permanent paralysis, loss of sensation and debilitating dysautonomia. However, the majority of circuits located above and below the injury remain anatomically intact, and these circuits can reorganize naturally to improve function. In addition, various neuromodulation therapies have tapped into these processes to further augment recovery. Emerging research is illuminating the requirements to reconstitute damaged circuits. Here, we summarize these natural and targeted reorganizations of circuits after a spinal cord injury. We also advocate for new concepts of reorganizing circuits informed by multi-omic single-cell atlases of recovery from injury. These atlases will uncover the molecular logic that governs the selection of 'recovery-organizing' neuronal subpopulations, and are poised to herald a new era in spinal cord medicine.
Collapse
Affiliation(s)
- Mark A Anderson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Jordan W Squair
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Matthieu Gautier
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Thomas H Hutson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Claudia Kathe
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Quentin Barraud
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Jocelyne Bloch
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Grégoire Courtine
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland. .,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland. .,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.
| |
Collapse
|
49
|
Turrel O, Ramesh N, Escher MJF, Pooryasin A, Sigrist SJ. Transient active zone remodeling in the Drosophila mushroom body supports memory. Curr Biol 2022; 32:4900-4913.e4. [PMID: 36327980 DOI: 10.1016/j.cub.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 08/15/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
Abstract
Elucidating how the distinct components of synaptic plasticity dynamically orchestrate the distinct stages of memory acquisition and maintenance within neuronal networks remains a major challenge. Specifically, plasticity processes tuning the functional and also structural state of presynaptic active zone (AZ) release sites are widely observed in vertebrates and invertebrates, but their behavioral relevance remains mostly unclear. We here provide evidence that a transient upregulation of presynaptic AZ release site proteins supports aversive olfactory mid-term memory in the Drosophila mushroom body (MB). Upon paired aversive olfactory conditioning, AZ protein levels (ELKS-family BRP/(m)unc13-family release factor Unc13A) increased for a few hours with MB-lobe-specific dynamics. Kenyon cell (KC, intrinsic MB neurons)-specific knockdown (KD) of BRP did not affect aversive olfactory short-term memory (STM) but strongly suppressed aversive mid-term memory (MTM). Different proteins crucial for the transport of AZ biosynthetic precursors (transport adaptor Aplip1/Jip-1; kinesin motor IMAC/Unc104; small GTPase Arl8) were also specifically required for the formation of aversive olfactory MTM. Consistent with the merely transitory increase of AZ proteins, BRP KD did not interfere with the formation of aversive olfactory long-term memory (LTM; i.e., 1 day). Our data suggest that the remodeling of presynaptic AZ refines the MB circuitry after paired aversive conditioning, over a time window of a few hours, to display aversive olfactory memories.
Collapse
Affiliation(s)
- Oriane Turrel
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Niraja Ramesh
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Marc J F Escher
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Atefeh Pooryasin
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
50
|
Flores-Muñoz C, García-Rojas F, Pérez MA, Santander O, Mery E, Ordenes S, Illanes-González J, López-Espíndola D, González-Jamett AM, Fuenzalida M, Martínez AD, Ardiles ÁO. The Long-Term Pannexin 1 Ablation Produces Structural and Functional Modifications in Hippocampal Neurons. Cells 2022; 11:cells11223646. [PMID: 36429074 PMCID: PMC9688914 DOI: 10.3390/cells11223646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/29/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Enhanced activity and overexpression of Pannexin 1 (Panx1) channels contribute to neuronal pathologies such as epilepsy and Alzheimer's disease (AD). The Panx1 channel ablation alters the hippocampus's glutamatergic neurotransmission, synaptic plasticity, and memory flexibility. Nevertheless, Panx1-knockout (Panx1-KO) mice still retain the ability to learn, suggesting that compensatory mechanisms stabilize their neuronal activity. Here, we show that the absence of Panx1 in the adult brain promotes a series of structural and functional modifications in the Panx1-KO hippocampal synapses, preserving spontaneous activity. Compared to the wild-type (WT) condition, the adult hippocampal neurons of Panx1-KO mice exhibit enhanced excitability, a more complex dendritic branching, enhanced spine maturation, and an increased proportion of multiple synaptic contacts. These modifications seem to rely on the actin-cytoskeleton dynamics as an increase in the actin polymerization and an imbalance between the Rac1 and the RhoA GTPase activities were observed in Panx1-KO brain tissues. Our findings highlight a novel interaction between Panx1 channels, actin, and Rho GTPases, which appear to be relevant for synapse stability.
Collapse
Affiliation(s)
- Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Francisca García-Rojas
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Miguel A. Pérez
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Escuela de Ciencias de la Salud, Universidad de Viña del Mar, Viña del Mar 2572007, Chile
| | - Odra Santander
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Elena Mery
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Stefany Ordenes
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2529002, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2529002, Chile
| | - Arlek M. González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología integrativa, CENFI, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| | - Álvaro O. Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de estudios en salud, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2572007, Chile
- Correspondence: (M.F.); (A.D.M.); (Á.O.A.)
| |
Collapse
|