1
|
Ghandour K, Haga T, Ohkawa N, Fung CCA, Nomoto M, Fayed MR, Asai H, Sato M, Fukai T, Inokuchi K. Parallel processing of past and future memories through reactivation and synaptic plasticity mechanisms during sleep. Nat Commun 2025; 16:3618. [PMID: 40295514 PMCID: PMC12037800 DOI: 10.1038/s41467-025-58860-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: 06/17/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
Every day, we experience new episodes and store new memories. Although memories are stored in corresponding engram cells, how different sets of engram cells are selected for current and next episodes, and how they create their memories, remains unclear. Here we show that in male mice, hippocampal CA1 neurons show an organized synchronous activity in prelearning home cage sleep that correlates with the learning ensembles only in engram cells, termed preconfigured ensembles. Moreover, after learning, a subset of nonengram cells develops population activity, which is constructed during postlearning offline periods, and then emerges to represent engram cells for new learning. Our model suggests a potential role of synaptic depression and scaling in the reorganization of the activity of nonengram cells. Together, our findings indicate that during offline periods there are two parallel processes occurring: conserving of past memories through reactivation, and preparation for upcoming ones through offline synaptic plasticity mechanisms.
Collapse
Affiliation(s)
- Khaled Ghandour
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Center initiative for training international researchers (CITIR), University of Toyama, Toyama, Japan
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Tatsuya Haga
- Neural Coding and Brain Computing unit, OIST, Okinawa, Japan
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology, Osaka, Japan
| | - Noriaki Ohkawa
- Research Center for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
| | - Chi Chung Alan Fung
- Neural Coding and Brain Computing unit, OIST, Okinawa, Japan
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, Hong Kong
| | - Masanori Nomoto
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Mostafa R Fayed
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Department of Pharmacology and Toxicology, Kafrelsheikh University, Kafr El Sheikh, Egypt
| | - Hirotaka Asai
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaaki Sato
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tomoki Fukai
- Neural Coding and Brain Computing unit, OIST, Okinawa, Japan
| | - Kaoru Inokuchi
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan.
| |
Collapse
|
2
|
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
|
3
|
Caya-Bissonnette L, Béïque JC. Half a century legacy of long-term potentiation. Curr Biol 2024; 34:R640-R662. [PMID: 38981433 DOI: 10.1016/j.cub.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
In 1973, two papers from Bliss and Lømo and from Bliss and Gardner-Medwin reported that high-frequency synaptic stimulation in the dentate gyrus of rabbits resulted in a long-lasting increase in synaptic strength. This form of synaptic plasticity, commonly referred to as long-term potentiation (LTP), was immediately considered as an attractive mechanism accounting for the ability of the brain to store information. In this historical piece looking back over the past 50 years, we discuss how these two landmark contributions directly motivated a colossal research effort and detail some of the resulting milestones that have shaped our evolving understanding of the molecular and cellular underpinnings of LTP. We highlight the main features of LTP, cover key experiments that defined its induction and expression mechanisms, and outline the evidence supporting a potential role of LTP in learning and memory. We also briefly explore some ramifications of LTP on network stability, consider current limitations of LTP as a model of associative memory, and entertain future research orientations.
Collapse
Affiliation(s)
- Léa Caya-Bissonnette
- Graduate Program in Neuroscience, University of Ottawa, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada; Brain and Mind Research Institute's Centre for Neural Dynamics and Artificial Intelligence, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada
| | - Jean-Claude Béïque
- Brain and Mind Research Institute's Centre for Neural Dynamics and Artificial Intelligence, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
4
|
Gonzalez-Islas C, Sabra Z, Fong MF, Yilmam P, Au Yong N, Engisch K, Wenner P. GABAergic synaptic scaling is triggered by changes in spiking activity rather than AMPA receptor activation. eLife 2024; 12:RP87753. [PMID: 38941139 PMCID: PMC11213567 DOI: 10.7554/elife.87753] [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: 06/29/2024] Open
Abstract
Homeostatic plasticity represents a set of mechanisms that are thought to recover some aspect of neural function. One such mechanism called AMPAergic scaling was thought to be a likely candidate to homeostatically control spiking activity. However, recent findings have forced us to reconsider this idea as several studies suggest AMPAergic scaling is not directly triggered by changes in spiking. Moreover, studies examining homeostatic perturbations in vivo have suggested that GABAergic synapses may be more critical in terms of spiking homeostasis. Here, we show results that GABAergic scaling can act to homeostatically control spiking levels. We found that perturbations which increased or decreased spiking in cortical cultures triggered multiplicative GABAergic upscaling and downscaling, respectively. In contrast, we found that changes in AMPA receptor (AMPAR) or GABAR transmission only influence GABAergic scaling through their indirect effect on spiking. We propose that GABAergic scaling represents a stronger candidate for spike rate homeostat than AMPAergic scaling.
Collapse
Affiliation(s)
- Carlos Gonzalez-Islas
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Doctorado en Ciencias Biológicas Universidad Autónoma de TlaxcalaTlaxMexico
| | - Zahraa Sabra
- Department of Neurosurgery, Emory UniversityAtlantaUnited States
| | - Ming-fai Fong
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Department of Biomedical Engineering, Georgia Tech and Emory UniversityAtlantaUnited States
| | - Pernille Yilmam
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Nicholas Au Yong
- Department of Neurosurgery, Emory UniversityAtlantaUnited States
| | - Kathrin Engisch
- Department of Neuroscience, Cell Biology and Physiology, Wright State UniversityDaytonUnited States
| | - Peter Wenner
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| |
Collapse
|
5
|
Koesters AG, Rich MM, Engisch KL. Diverging from the Norm: Reevaluating What Miniature Excitatory Postsynaptic Currents Tell Us about Homeostatic Synaptic Plasticity. Neuroscientist 2024; 30:49-70. [PMID: 35904350 DOI: 10.1177/10738584221112336] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The idea that the nervous system maintains a set point of network activity and homeostatically returns to that set point in the face of dramatic disruption-during development, after injury, in pathologic states, and during sleep/wake cycles-is rapidly becoming accepted as a key plasticity behavior, placing it alongside long-term potentiation and depression. The dramatic growth in studies of homeostatic synaptic plasticity of miniature excitatory synaptic currents (mEPSCs) is attributable, in part, to the simple yet elegant mechanism of uniform multiplicative scaling proposed by Turrigiano and colleagues: that neurons sense their own activity and globally multiply the strength of every synapse by a single factor to return activity to the set point without altering established differences in synaptic weights. We have recently shown that for mEPSCs recorded from control and activity-blocked cultures of mouse cortical neurons, the synaptic scaling factor is not uniform but is close to 1 for the smallest mEPSC amplitudes and progressively increases as mEPSC amplitudes increase, which we term divergent scaling. Using insights gained from simulating uniform multiplicative scaling, we review evidence from published studies and conclude that divergent synaptic scaling is the norm rather than the exception. This conclusion has implications for hypotheses about the molecular mechanisms underlying synaptic scaling.
Collapse
Affiliation(s)
- Andrew G Koesters
- Department of Behavior, Cognition, and Neurophysiology, Environmental Health Effects Laboratory, Naval Medical Research Unit-Dayton, Wright-Patterson AFB, OH, USA
| | - Mark M Rich
- Department of Neuroscience, Cell Biology, and Physiology, College of Science and Mathematics, and Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Kathrin L Engisch
- Department of Neuroscience, Cell Biology, and Physiology, College of Science and Mathematics, and Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| |
Collapse
|
6
|
Li R, Tang J, Wang Y, Wang Y, Yang H, Wei H. Metabolomics and transcriptomics analysis of prefrontal cortex in the Pax2 neuron-specific deletion mice. Prog Neuropsychopharmacol Biol Psychiatry 2024; 128:110858. [PMID: 37660748 DOI: 10.1016/j.pnpbp.2023.110858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
Restricted and repetitive behaviors (RRBs) are one of the characteristics of various neuropsychiatric disorders with complex and diverse molecular mechanisms. Repetitive self-grooming behavior is one of the manifestations of RRBs in humans and rodents. Research on the neural mechanism of repetitive self-grooming behavior is expected to reveal the underlying logic of the occurrence of RRBs. Pax2 is an important member of the paired-box transcription factor family. It is expressed in different regions of the developing central nervous system. Our previous study showed that Pax2 heterozygous gene knockout mice (Pax2+/- KO mice) exhibit significantly increased self-grooming, which suggests that the Pax2 gene is involved in the control of self-grooming behavior, but the molecular mechanism is still unclear. In this study, we further constructed the Pax2 neuron-specific deletion mice (Nestin-Pax2 mice). Targeted metabolomics and transcriptomics techniques was used to analyze. The results showed that there is an excitatory/inhibitory imbalance of the neurotransmitter system and the Arc gene was significantly up-regulated in the prefrontal cortex (PFC) of Nestin-Pax2 mice. This study suggests that the potential regulatory mechanism of the increased repetitive self-grooming behavior in Pax2 gene deletion mice is that the deletion of the Pax2 gene affects the expression of Arc in the PFC, leading to impaired synaptic plasticity and excitatory/inhibitory imbalance, and participating in the occurrence of repetitive self-grooming behavior.
Collapse
Affiliation(s)
- Rui Li
- Department of Neurology, Shanxi Provincial People's Hospital, the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China; Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Jiaming Tang
- School of the Third Clinic, Shanxi University of Chinese Medicine, Taiyuan 030024, China
| | - Yizhuo Wang
- Department of Neurology, Shanxi Provincial People's Hospital, the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China
| | - Ying Wang
- Department of Neurology, Shanxi Provincial People's Hospital, the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China
| | - Hua Yang
- Department of Neurology, Shanxi Provincial People's Hospital, the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China; Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China.
| | - Hongen Wei
- Department of Neurology, Shanxi Provincial People's Hospital, the Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China; Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China.
| |
Collapse
|
7
|
Caya-Bissonnette L, Béïque JC. Low throughput screening in neuroscience: using light to study central synapses one at a time. NEUROPHOTONICS 2023; 10:044407. [PMID: 37881180 PMCID: PMC10594030 DOI: 10.1117/1.nph.10.4.044407] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 10/27/2023]
Abstract
Neurophotonic approaches have fostered substantial progress in our understanding of the brain by providing an assortment of means to either monitor or manipulate neural processes. Among these approaches, the development of two-photon uncaging provides a useful and flexible approach to manipulate the activity of individual synapses. In this short piece, we explore how this technique has emerged at the intersection of chemistry, optics, and electrophysiology to enable spatially and temporally precise photoactivation for studying functional aspects of synaptic transmission and dendritic integration. We discuss advantages and limitations of this approach, focusing on our efforts to study several functional aspects of glutamate receptors using uncaging of glutamate. Among other advancements, this approach has contributed to further our understanding of the subcellular regulation, trafficking, and biophysical features of glutamate receptors (e.g., desensitization and silent synapse regulation), the dynamics of spine calcium, and the integrative features of dendrites, and how these functions are altered by several forms of plasticity.
Collapse
Affiliation(s)
- Léa Caya-Bissonnette
- University of Ottawa’s Brain and Mind Research Institute and Centre of Neural Dynamics, Ottawa, Ontario, Canada
| | - Jean-Claude Béïque
- University of Ottawa’s Brain and Mind Research Institute and Centre of Neural Dynamics, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
Ramsay HJ, Gookin SE, Ramsey AM, Kareemo DJ, Crosby KC, Stich DG, Olah SS, Actor-Engel HS, Smith KR, Kennedy MJ. AMPA and GABAA receptor nanodomains assemble in the absence of synaptic neurotransmitter release. Front Mol Neurosci 2023; 16:1232795. [PMID: 37602191 PMCID: PMC10435253 DOI: 10.3389/fnmol.2023.1232795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Postsynaptic neurotransmitter receptors and their associated scaffolding proteins assemble into discrete, nanometer-scale subsynaptic domains (SSDs) within the postsynaptic membrane at both excitatory and inhibitory synapses. Intriguingly, postsynaptic receptor SSDs are mirrored by closely apposed presynaptic active zones. These trans-synaptic molecular assemblies are thought to be important for efficient neurotransmission because they concentrate postsynaptic receptors near sites of presynaptic neurotransmitter release. While previous studies have characterized the role of synaptic activity in sculpting the number, size, and distribution of postsynaptic SSDs at established synapses, it remains unknown whether neurotransmitter signaling is required for their initial assembly during synapse development. Here, we evaluated synaptic nano-architecture under conditions where presynaptic neurotransmitter release was blocked prior to, and throughout synaptogenesis with tetanus neurotoxin (TeNT). In agreement with previous work, neurotransmitter release was not required for the formation of excitatory or inhibitory synapses. The overall size of the postsynaptic specialization at both excitatory and inhibitory synapses was reduced at chronically silenced synapses. However, both AMPARs and GABAARs still coalesced into SSDs, along with their respective scaffold proteins. Presynaptic active zone assemblies, defined by RIM1, were smaller and more numerous at silenced synapses, but maintained alignment with postsynaptic AMPAR SSDs. Thus, basic features of synaptic nano-architecture, including assembly of receptors and scaffolds into trans-synaptically aligned structures, are intrinsic properties that can be further regulated by subsequent activity-dependent mechanisms.
Collapse
Affiliation(s)
- Harrison J. Ramsay
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Sara E. Gookin
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Austin M. Ramsey
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Dean J. Kareemo
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Kevin C. Crosby
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Dominik G. Stich
- Anschutz Medical Campus, Advanced Light Microscopy Core, University of Colorado, Aurora, CO, United States
| | - Samantha S. Olah
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Hannah S. Actor-Engel
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Katharine R. Smith
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| | - Matthew J. Kennedy
- Anschutz Medical Campus, Department of Pharmacology, University of Colorado, Aurora, CO, United States
| |
Collapse
|
9
|
Ghane MA, Wei W, Yakout DW, Allen ZD, Miller CL, Dong B, Yang JJ, Fang N, Mabb AM. Arc ubiquitination regulates endoplasmic reticulum-mediated Ca 2+ release and CaMKII signaling. Front Cell Neurosci 2023; 17:1091324. [PMID: 36998269 PMCID: PMC10043188 DOI: 10.3389/fncel.2023.1091324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
Synaptic plasticity relies on rapid, yet spatially precise signaling to alter synaptic strength. Arc is a brain enriched protein that is rapidly expressed during learning-related behaviors and is essential for regulating metabotropic glutamate receptor-mediated long-term depression (mGluR-LTD). We previously showed that disrupting the ubiquitination capacity of Arc enhances mGluR-LTD; however, the consequences of Arc ubiquitination on other mGluR-mediated signaling events is poorly characterized. Here we find that pharmacological activation of Group I mGluRs with S-3,5-dihydroxyphenylglycine (DHPG) increases Ca2+ release from the endoplasmic reticulum (ER). Disrupting Arc ubiquitination on key amino acid residues enhances DHPG-induced ER-mediated Ca2+ release. These alterations were observed in all neuronal subregions except secondary branchpoints. Deficits in Arc ubiquitination altered Arc self-assembly and enhanced its interaction with calcium/calmodulin-dependent protein kinase IIb (CaMKIIb) and constitutively active forms of CaMKII in HEK293 cells. Colocalization of Arc and CaMKII was altered in cultured hippocampal neurons, with the notable exception of secondary branchpoints. Finally, disruptions in Arc ubiquitination were found to increase Arc interaction with the integral ER protein Calnexin. These results suggest a previously unknown role for Arc ubiquitination in the fine tuning of ER-mediated Ca2+ signaling that may support mGluR-LTD, which in turn, may regulate CaMKII and its interactions with Arc.
Collapse
Affiliation(s)
- Mohammad A. Ghane
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Wei Wei
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Dina W. Yakout
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Zachary D. Allen
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| | - Cassandra L. Miller
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Bin Dong
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Jenny J. Yang
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, United States
| | - Ning Fang
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
10
|
Drion C. Homeostatic Control of Neuronal Activity. Physiology (Bethesda) 2022. [DOI: 10.5772/intechopen.108577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
For healthy brain functioning, it is crucial that neuronal networks do not become hyperactive, but also, that they remain excitable. Homeostatic mechanisms ensure that neuronal activity remains within a functional range. How does that work? In this chapter, we will explore homeostatic control of neuronal activity. We will start by introducing the basics of neuronal communication to establish what makes a neuron excitable. Then, we will learn how neurons are able to tune their own excitability, which is called homeostatic intrinsic plasticity. Next, we will discuss the ability of neurons to tune the strength of their connections to other neurons. This is called homeostatic synaptic plasticity and involves synaptic scaling, the up- and downregulation of receptors, and the control of neurotransmitter release. Finally, we will review the role of glia in neuronal network homeostasis and discuss disorders where the homeostatic control of neuronal activity is compromised.
Collapse
|
11
|
Chen X, Jia B, Araki Y, Liu B, Ye F, Huganir R, Zhang M. Arc weakens synapses by dispersing AMPA receptors from postsynaptic density via modulating PSD phase separation. Cell Res 2022; 32:914-930. [PMID: 35856091 PMCID: PMC9525282 DOI: 10.1038/s41422-022-00697-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/29/2022] [Indexed: 01/16/2023] Open
Abstract
In response to stimuli, the immediate early gene product Arc can acutely down-regulate synaptic strength by removing AMPA receptors (AMPARs) from synapses and thus regulate synaptic plasticity. How Arc, a scaffold protein, can specifically facilitate synaptic removal of AMPARs is unknown. We found that Arc directly antagonizes with PSD-95 in binding to TARPs, which are the auxiliary subunits of AMPARs. Arc, in a highly concentration-sensitive manner, acutely disperses TARPs from the postsynaptic density (PSD) condensate formed via phase separation. TARPs with the Ser residue in the "P-S-Y"-motif of its tail phosphorylated are completely refractory from being dispersed by Arc, suggesting that Arc cannot displace AMPARs from PSDs in active synapses. Conversely, strengthening the interaction between Arc and TARPs enhances Arc's capacity in weakening synapses. Thus, Arc can specifically and effectively modulate synaptic AMPAR clustering via modulating PSD phase separation. Our study further suggests that activity-dependent, bi-directional modulation of PSD condensate formation/dispersion represents a general regulatory mechanism for synaptic plasticity.
Collapse
Affiliation(s)
- Xudong Chen
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Bowen Jia
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yoichi Araki
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bian Liu
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fei Ye
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Richard Huganir
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mingjie Zhang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
12
|
Leung HW, Foo G, VanDongen A. Arc Regulates Transcription of Genes for Plasticity, Excitability and Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081946. [PMID: 36009494 PMCID: PMC9405677 DOI: 10.3390/biomedicines10081946] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
The immediate early gene Arc is a master regulator of synaptic function and a critical determinant of memory consolidation. Here, we show that Arc interacts with dynamic chromatin and closely associates with histone markers for active enhancers and transcription in cultured rat hippocampal neurons. Both these histone modifications, H3K27Ac and H3K9Ac, have recently been shown to be upregulated in late-onset Alzheimer’s disease (AD). When Arc induction by pharmacological network activation was prevented using a short hairpin RNA, the expression profile was altered for over 1900 genes, which included genes associated with synaptic function, neuronal plasticity, intrinsic excitability, and signalling pathways. Interestingly, about 100 Arc-dependent genes are associated with the pathophysiology of AD. When endogenous Arc expression was induced in HEK293T cells, the transcription of many neuronal genes was increased, suggesting that Arc can control expression in the absence of activated signalling pathways. Taken together, these data establish Arc as a master regulator of neuronal activity-dependent gene expression and suggest that it plays a significant role in the pathophysiology of AD.
Collapse
Affiliation(s)
| | - Gabriel Foo
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Antonius VanDongen
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Correspondence:
| |
Collapse
|
13
|
Dubes S, Soula A, Benquet S, Tessier B, Poujol C, Favereaux A, Thoumine O, Letellier M. miR
‐124‐dependent tagging of synapses by synaptopodin enables input‐specific homeostatic plasticity. EMBO J 2022; 41:e109012. [PMID: 35875872 PMCID: PMC9574720 DOI: 10.15252/embj.2021109012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 06/11/2022] [Accepted: 06/27/2022] [Indexed: 12/26/2022] Open
Abstract
Homeostatic synaptic plasticity is a process by which neurons adjust their synaptic strength to compensate for perturbations in neuronal activity. Whether the highly diverse synapses on a neuron respond uniformly to the same perturbation remains unclear. Moreover, the molecular determinants that underlie synapse‐specific homeostatic synaptic plasticity are unknown. Here, we report a synaptic tagging mechanism in which the ability of individual synapses to increase their strength in response to activity deprivation depends on the local expression of the spine‐apparatus protein synaptopodin under the regulation of miR‐124. Using genetic manipulations to alter synaptopodin expression or regulation by miR‐124, we show that synaptopodin behaves as a “postsynaptic tag” whose translation is derepressed in a subpopulation of synapses and allows for nonuniform homeostatic strengthening and synaptic AMPA receptor stabilization. By genetically silencing individual connections in pairs of neurons, we demonstrate that this process operates in an input‐specific manner. Overall, our study shifts the current view that homeostatic synaptic plasticity affects all synapses uniformly to a more complex paradigm where the ability of individual synapses to undergo homeostatic changes depends on their own functional and biochemical state.
Collapse
Affiliation(s)
- Sandra Dubes
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Anaïs Soula
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Sébastien Benquet
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Béatrice Tessier
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Christel Poujol
- University of Bordeaux CNRS INSERM Bordeaux Imaging Center BIC UMS 3420, US 4 Bordeaux France
| | - Alexandre Favereaux
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Olivier Thoumine
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Mathieu Letellier
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| |
Collapse
|
14
|
Vergara P, Pino G, Vera J, Arancibia F, Sanhueza M. Heterogeneous CaMKII-Dependent Synaptic Compensations in CA1 Pyramidal Neurons From Acute Hippocampal Slices. Front Cell Neurosci 2022; 16:821088. [PMID: 35431809 PMCID: PMC9005847 DOI: 10.3389/fncel.2022.821088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Prolonged changes in neural activity trigger homeostatic synaptic plasticity (HSP) allowing neuronal networks to operate within functional ranges. Cell-wide or input-specific adaptations can be induced by pharmacological or genetic manipulations of activity, and by sensory deprivation. Reactive functional changes caused by deafferentation may partially share mechanisms with HSP. Acute hippocampal slices are a suitable model to investigate relatively rapid (hours) modifications occurring after denervation and explore the underlying mechanisms. As during slicing many afferents are cut, we conducted whole-cell recordings of miniature excitatory postsynaptic currents (mEPSCs) in CA1 pyramidal neurons to evaluate changes over the following 12 h. As Schaffer collaterals constitute a major glutamatergic input to these neurons, we also dissected CA3. We observed an average increment in mEPSCs amplitude and a decrease in decay time, suggesting synaptic AMPA receptor upregulation and subunit content modifications. Sorting mEPSC by rise time, a correlate of synapse location along dendrites, revealed amplitude raises at two separate domains. A specific frequency increase was observed in the same domains and was accompanied by a global, unspecific raise. Amplitude and frequency increments were lower at sites initially more active, consistent with local compensatory processes. Transient preincubation with a specific Ca2+/calmodulin-dependent kinase II (CaMKII) inhibitor either blocked or occluded amplitude and frequency upregulation in different synapse populations. Results are consistent with the concurrent development of different known CaMKII-dependent HSP processes. Our observations support that deafferentation causes rapid and diverse compensations resembling classical slow forms of adaptation to inactivity. These results may contribute to understand fast-developing homeostatic or pathological events after brain injury.
Collapse
|
15
|
Chater TE, Goda Y. The Shaping of AMPA Receptor Surface Distribution by Neuronal Activity. Front Synaptic Neurosci 2022; 14:833782. [PMID: 35387308 PMCID: PMC8979068 DOI: 10.3389/fnsyn.2022.833782] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/25/2022] [Indexed: 12/29/2022] Open
Abstract
Neurotransmission is critically dependent on the number, position, and composition of receptor proteins on the postsynaptic neuron. Of these, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) are responsible for the majority of postsynaptic depolarization at excitatory mammalian synapses following glutamate release. AMPARs are continually trafficked to and from the cell surface, and once at the surface, AMPARs laterally diffuse in and out of synaptic domains. Moreover, the subcellular distribution of AMPARs is shaped by patterns of activity, as classically demonstrated by the synaptic insertion or removal of AMPARs following the induction of long-term potentiation (LTP) and long-term depression (LTD), respectively. Crucially, there are many subtleties in the regulation of AMPARs, and exactly how local and global synaptic activity drives the trafficking and retention of synaptic AMPARs of different subtypes continues to attract attention. Here we will review how activity can have differential effects on AMPAR distribution and trafficking along with its subunit composition and phosphorylation state, and we highlight some of the controversies and remaining questions. As the AMPAR field is extensive, to say the least, this review will focus primarily on cellular and molecular studies in the hippocampus. We apologise to authors whose work could not be cited directly owing to space limitations.
Collapse
|
16
|
Enzymatic Degradation of Cortical Perineuronal Nets Reverses GABAergic Interneuron Maturation. Mol Neurobiol 2022; 59:2874-2893. [PMID: 35233718 PMCID: PMC9016038 DOI: 10.1007/s12035-022-02772-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/16/2022] [Indexed: 12/03/2022]
Abstract
Perineuronal nets (PNNs) are specialised extracellular matrix structures which preferentially enwrap fast-spiking (FS) parvalbumin interneurons and have diverse roles in the cortex. PNN maturation coincides with closure of the critical period of cortical plasticity. We have previously demonstrated that BDNF accelerates interneuron development in a c-Jun-NH2-terminal kinase (JNK)–dependent manner, which may involve upstream thousand-and-one amino acid kinase 2 (TAOK2). Chondroitinase-ABC (ChABC) enzymatic digestion of PNNs reportedly reactivates ‘juvenile-like’ plasticity in the adult CNS. However, the mechanisms involved are unclear. We show that ChABC produces an immature molecular phenotype in cultured cortical neurons, corresponding to the phenotype prior to critical period closure. ChABC produced different patterns of PNN-related, GABAergic and immediate early (IE) gene expression than well-characterised modulators of mature plasticity and network activity (GABAA-R antagonist, bicuculline, and sodium-channel blocker, tetrodotoxin (TTX)). ChABC downregulated JNK activity, while this was upregulated by bicuculline. Bicuculline, but not ChABC, upregulated Bdnf expression and ERK activity. Furthermore, we found that BDNF upregulation of semaphorin-3A and IE genes was TAOK mediated. Our data suggest that ChABC heightens structural flexibility and network disinhibition, potentially contributing to ‘juvenile-like’ plasticity. The molecular phenotype appears to be distinct from heightened mature synaptic plasticity and could relate to JNK signalling. Finally, we highlight that BDNF regulation of plasticity and PNNs involves TAOK signalling.
Collapse
|
17
|
Liu DC, Lee KY, Lizarazo S, Cook JK, Tsai NP. ER stress-induced modulation of neural activity and seizure susceptibility is impaired in a fragile X syndrome mouse model. Neurobiol Dis 2021; 158:105450. [PMID: 34303799 DOI: 10.1016/j.nbd.2021.105450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/18/2021] [Indexed: 01/29/2023] Open
Abstract
Imbalanced neuronal excitability homeostasis is commonly observed in patients with fragile X syndrome (FXS) and the animal model of FXS, the Fmr1 KO. While alterations of neuronal intrinsic excitability and synaptic activity at the steady state in FXS have been suggested to contribute to such a deficit and ultimately the increased susceptibility to seizures in FXS, it remains largely unclear whether and how the homeostatic response of neuronal excitability following extrinsic challenges is disrupted in FXS. Our previous work has shown that the acute response following induction of endoplasmic reticulum (ER) stress can reduce neural activity and seizure susceptibility. Because many signaling pathways associated with ER stress response are mediated by Fmr1, we asked whether acute ER stress-induced reduction of neural activity and seizure susceptibility are altered in FXS. Our results first revealed that acute ER stress can trigger a protein synthesis-dependent prevention of neural network synchronization in vitro and a reduction of susceptibility to kainic acid-induced seizures in vivo in wild-type but not in Fmr1 KO mice. Mechanistically, we found that acute ER stress-induced activation of murine double minute-2 (Mdm2), ubiquitination of p53, and the subsequent transient protein synthesis are all impaired in Fmr1 KO neurons. Employing a p53 inhibitor, Pifithrin-α, to mimic p53 inactivation, we were able to blunt the increase in neural network synchronization and reduce the seizure susceptibility in Fmr1 KO mice following ER stress induction. In summary, our data revealed a novel cellular defect in Fmr1 KO mice and suggest that an impaired response to common extrinsic challenges may contribute to imbalanced neuronal excitability homeostasis in FXS.
Collapse
Affiliation(s)
- Dai-Chi Liu
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Simon Lizarazo
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jessie K Cook
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Nien-Pei Tsai
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
18
|
Zhong M, Cywiak C, Metto AC, Liu X, Qian C, Pelled G. Multi-session delivery of synchronous rTMS and sensory stimulation induces long-term plasticity. Brain Stimul 2021; 14:884-894. [PMID: 34029768 DOI: 10.1016/j.brs.2021.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/17/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Combining training or sensory stimulation with non-invasive brain stimulation has shown to improve performance in healthy subjects and improve brain function in patients after brain injury. However, the plasticity mechanisms and the optimal parameters to induce long-term and sustainable enhanced performance remain unknown. OBJECTIVE This work was designed to identify the protocols of which combining sensory stimulation with repetitive transcranial magnetic stimulation (rTMS) will facilitate the greatest changes in fMRI activation maps in the rat's primary somatosensory cortex (S1). METHODS Several protocols of combining forepaw electrical stimulation with rTMS were tested, including a single stimulation session compared to multiple, daily stimulation sessions, as well as synchronous and asynchronous delivery of both modalities. High-resolution fMRI was used to determine how pairing sensory stimulation with rTMS induced short and long-term plasticity in the rat S1. RESULTS All groups that received a single session of rTMS showed short-term increases in S1 activity, but these increases did not last three days after the session. The group that received a stimulation protocol of 10 Hz forepaw stimulation that was delivered simultaneously with 10 Hz rTMS for five consecutive days demonstrated the greatest increases in the extent of the evoked fMRI responses compared to groups that received other stimulation protocols. CONCLUSIONS Our results provide direct indication that pairing peripheral stimulation with rTMS induces long-term plasticity, and this phenomenon appears to follow a time-dependent plasticity mechanism. These results will be important to lead the design of new training and rehabilitation paradigms and training towards achieving maximal performance in healthy subjects.
Collapse
Affiliation(s)
- Ming Zhong
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Carolina Cywiak
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Abigael C Metto
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Xiang Liu
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Galit Pelled
- Neuroengineering Division, The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA; Department of Radiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
19
|
Chokshi V, Grier BD, Dykman A, Lantz CL, Niebur E, Quinlan EM, Lee HK. Naturalistic Spike Trains Drive State-Dependent Homeostatic Plasticity in Superficial Layers of Visual Cortex. Front Synaptic Neurosci 2021; 13:663282. [PMID: 33935679 PMCID: PMC8081846 DOI: 10.3389/fnsyn.2021.663282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
The history of neural activity determines the synaptic plasticity mechanisms employed in the brain. Previous studies report a rapid reduction in the strength of excitatory synapses onto layer 2/3 (L2/3) pyramidal neurons of the primary visual cortex (V1) following two days of dark exposure and subsequent re-exposure to light. The abrupt increase in visually driven activity is predicted to drive homeostatic plasticity, however, the parameters of neural activity that trigger these changes are unknown. To determine this, we first recorded spike trains in vivo from V1 layer 4 (L4) of dark exposed (DE) mice of both sexes that were re-exposed to light through homogeneous or patterned visual stimulation. We found that delivering the spike patterns recorded in vivo to L4 of V1 slices was sufficient to reduce the amplitude of miniature excitatory postsynaptic currents (mEPSCs) of V1 L2/3 neurons in DE mice, but not in slices obtained from normal reared (NR) controls. Unexpectedly, the same stimulation pattern produced an up-regulation of mEPSC amplitudes in V1 L2/3 neurons from mice that received 2 h of light re-exposure (LE). A Poisson spike train exhibiting the same average frequency as the patterns recorded in vivo was equally effective at depressing mEPSC amplitudes in L2/3 neurons in V1 slices prepared from DE mice. Collectively, our results suggest that the history of visual experience modifies the responses of V1 neurons to stimulation and that rapid homeostatic depression of excitatory synapses can be driven by non-patterned input activity.
Collapse
Affiliation(s)
- Varun Chokshi
- The Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, United States
- Cell Molecular Developmental Biology and Biophysics (CMDB) Graduate Program, Johns Hopkins University, Baltimore, MD, United States
| | - Bryce D. Grier
- The Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Andrew Dykman
- The Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Crystal L. Lantz
- Department of Biology, University of Maryland, College Park, MD, United States
| | - Ernst Niebur
- The Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Elizabeth M. Quinlan
- Department of Biology, University of Maryland, College Park, MD, United States
- Neuroscience and Cognitive Science Program, Brain and Behavior Institute, University of Maryland, College Park, MD, United States
| | - Hey-Kyoung Lee
- The Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, United States
- Cell Molecular Developmental Biology and Biophysics (CMDB) Graduate Program, Johns Hopkins University, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
20
|
Scarpa JR, Jiang P, Gao VD, Vitaterna MH, Turek FW, Kasarskis A. NREM delta power and AD-relevant tauopathy are associated with shared cortical gene networks. Sci Rep 2021; 11:7797. [PMID: 33833255 PMCID: PMC8032807 DOI: 10.1038/s41598-021-86255-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/27/2020] [Indexed: 02/01/2023] Open
Abstract
Reduced NREM sleep in humans is associated with AD neuropathology. Recent work has demonstrated a reduction in NREM sleep in preclinical AD, pointing to its potential utility as an early marker of dementia. We test the hypothesis that reduced NREM delta power and increased tauopathy are associated with shared underlying cortical molecular networks in preclinical AD. We integrate multi-omics data from two extensive public resources, a human Alzheimer's disease cohort from the Mount Sinai Brain Bank (N = 125) reflecting AD progression and a (C57BL/6J × 129S1/SvImJ) F2 mouse population in which NREM delta power was measured (N = 98). Two cortical gene networks, including a CLOCK-dependent circadian network, are associated with NREM delta power and AD tauopathy progression. These networks were validated in independent mouse and human cohorts. Identifying gene networks related to preclinical AD elucidate possible mechanisms associated with the early disease phase and potential targets to alter the disease course.
Collapse
Affiliation(s)
- Joseph R Scarpa
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Peng Jiang
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Vance D Gao
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Martha H Vitaterna
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Fred W Turek
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Andrew Kasarskis
- Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
21
|
Haider HF, Hoare DJ, Ribeiro SF, Ribeiro D, Caria H, Trigueiros N, Borrego LM, Szczepek AJ, Papoila AL, Elarbed A, da Luz Martins M, Paço J, Sereda M. Evidence for biological markers of tinnitus: A systematic review. PROGRESS IN BRAIN RESEARCH 2021; 262:345-398. [PMID: 33931188 DOI: 10.1016/bs.pbr.2021.01.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Subjective tinnitus is a phantom sound heard only by the affected person and may be a symptom of various diseases. Tinnitus diagnosis and monitoring is based on subjective audiometric and psychometric methods. This review aimed to synthesize evidence for tinnitus presence or its severity. We searched several electronic databases, citation searches of the included primary studies through Web of Science, and further hand searches. At least two authors performed all systematic review steps. Sixty-two records were included and were categorized according the biological variable. Evidence for possible tinnitus biomarkers come from oxidative stress, interleukins, steroids and neurotransmitters categories. We found conflicting evidence for full blood count, vitamins, lipid profile, neurotrophic factors, or inorganic ions. There was no evidence for an association between tinnitus and the remaining categories. The current review evidences that larger studies, with stricter exclusion criteria and powerful harmonized methodological design are needed. Protocol published on PROSPERO (CRD42017070998).
Collapse
Affiliation(s)
- Haúla F Haider
- ENT Department, Hospital Cuf Tejo-Nova Medical School, Lisbon, Portugal; CUF Academic and Research Medical Center, Lisbon, Portugal; Comprehensive Health Research Centre (CHRC), Lisbon, Portugal.
| | - Derek J Hoare
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Sara F Ribeiro
- ENT Department, Hospital Cuf Tejo-Nova Medical School, Lisbon, Portugal; CUF Academic and Research Medical Center, Lisbon, Portugal
| | - Diogo Ribeiro
- ENT Department, Hospital Cuf Tejo-Nova Medical School, Lisbon, Portugal; CUF Academic and Research Medical Center, Lisbon, Portugal
| | - Helena Caria
- Deafness Research Group, BTR Unit, BioISI, Faculty of Sciences, University of Lisbon (FCUL), Portugal; ESS/IPS-Biomedical Sciences Department, School of Health, Polytechnic Institute of Setubal, Portugal
| | - Nuno Trigueiros
- ENT Department, Hospital Pedro Hispano, Matosinhos, Portugal
| | - Luís Miguel Borrego
- Department of Immunology, Chronic Diseases Research Center (CEDOC), Faculty of Medical Sciences, NOVA Medical School, Lisbon, Portugal; Department of Immunoallergy, LUZ SAUDE, Hospital da Luz, Lisbon, Portugal
| | - Agnieszka J Szczepek
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Ana Luísa Papoila
- Bioestatistics Department, Faculty of Medical Sciences, NOVA Medical School, Lisbon, Portugal
| | - Asma Elarbed
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Maria da Luz Martins
- ENT Department, Hospital Cuf Tejo-Nova Medical School, Lisbon, Portugal; CUF Academic and Research Medical Center, Lisbon, Portugal
| | - João Paço
- ENT Department, Hospital Cuf Tejo-Nova Medical School, Lisbon, Portugal; CUF Academic and Research Medical Center, Lisbon, Portugal
| | - Magdalena Sereda
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
22
|
Müller-Schiffmann A, Trossbach SV, Lingappa VR, Korth C. Viruses as 'Truffle Hounds': Molecular Tools for Untangling Brain Cellular Pathology. Trends Neurosci 2020; 44:352-365. [PMID: 33317827 DOI: 10.1016/j.tins.2020.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022]
Abstract
The ability of viruses to evolve several orders of magnitude faster than their host cells has enabled them to exploit host cellular machinery by selectively recruiting multiprotein complexes (MPCs) for their catalyzed assembly and replication. This hijacking may depend on alternative, 'moonlighting' functions of host proteins that deviate from their canonical functions thereby inducing cellular pathology. Here, we posit that if virus-induced cellular pathology is similar to that of other, unknown (non-viral) causes, the identification and molecular characterization of the host proteins involved in virus-mediated cellular pathology can be leveraged to decipher the non-viral disease-relevant mechanisms. We focus on how virus-induced aberrant proteostasis and protein aggregation resemble the cellular pathology of sporadic neurodegenerative diseases (NDs) and how this can be exploited for drug discovery.
Collapse
Affiliation(s)
- Andreas Müller-Schiffmann
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Svenja V Trossbach
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | - Carsten Korth
- Department of Neuropathology, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
23
|
Wang JKT. Uniting homeostatic plasticity and exosome biology: A revision of the conceptual framework for drug discovery in neurodegenerative diseases? ADVANCES IN PHARMACOLOGY 2020; 90:277-306. [PMID: 33706937 DOI: 10.1016/bs.apha.2020.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Neurodegenerative diseases (NDDs) are in need of new drug discovery approaches. Our previous systematic analyses of Huntington's Disease (HD) literature for protein-protein interactors (PPIs) and modifiers of mutant Huntingtin-driven phenotypes revealed enrichment for PPIs of genes required for homeostatic synaptic plasticity (HSP) and exosome (EV) function and exosomal proteins, which in turn highly overlapped each other and with PPIs of genes associated with other NDDs. We proposed that HSP and EVs are linked to each other and are also involved in NDD pathophysiology. Recent studies showed that HSP is indeed altered in HD and AD, and that presynaptic homeostatic plasticity in motoneurons compensates for ALS pathology. Eliminating it causes earlier degeneration and death. If this holds true in other NDDs, drug discovery in animal models should then include elucidation of homeostatic compensation that either masks phenotypes of physiologically expressed mutant genes or are overridden by their overexpression. In this new conceptual framework, enhancing such underlying homeostatic compensation forms the basis for novel therapeutic strategies to slow progression of NDDs. Moreover, if EVs are linked to HSP, then their ability to penetrate the brain, target cell types, deliver miRNA and other molecules can be leveraged to develop attractive drug modalities. Testing this new framework is posed as four questions on model development and mechanistic studies progressing from higher throughput platforms to mouse models. Similar approaches may apply to other CNS disorders including schizophrenia, autism, Rett and Fragile X syndromes due to potential links of their susceptibility genes to HSP and EVs.
Collapse
|
24
|
Khlghatyan J, Evstratova A, Bozoyan L, Chamberland S, Chatterjee D, Marakhovskaia A, Soares Silva T, Toth K, Mongrain V, Beaulieu J. Fxr1 regulates sleep and synaptic homeostasis. EMBO J 2020; 39:e103864. [PMID: 32893934 PMCID: PMC7604579 DOI: 10.15252/embj.2019103864] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
The fragile X autosomal homolog 1 (Fxr1) is regulated by lithium and has been GWAS-associated with schizophrenia and insomnia. Homeostatic regulation of synaptic strength is essential for the maintenance of brain functions and involves both cell-autonomous and system-level processes such as sleep. We examined the contribution of Fxr1 to cell-autonomous homeostatic synaptic scaling and neuronal responses to sleep loss, using a combination of gene overexpression and Crispr/Cas9-mediated somatic knockouts to modulate gene expression. Our findings indicate that Fxr1 is downregulated during both scaling and sleep deprivation via a glycogen synthase kinase 3 beta (GSK3β)-dependent mechanism. In both conditions, downregulation of Fxr1 is essential for the homeostatic modulation of surface AMPA receptors and synaptic strength. Preventing the downregulation of Fxr1 during sleep deprivation results in altered EEG signatures. Furthermore, sequencing of neuronal translatomes revealed the contribution of Fxr1 to changes induced by sleep deprivation. These findings uncover a role of Fxr1 as a shared signaling hub between cell-autonomous homeostatic plasticity and system-level responses to sleep loss, with potential implications for neuropsychiatric illnesses and treatments.
Collapse
Affiliation(s)
- Jivan Khlghatyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
Department of NeuroscienceNovartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Alesya Evstratova
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Lusine Bozoyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Simon Chamberland
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
NYU Neuroscience InstituteLangone Medical CenterNew York UniversityNew YorkNYUSA
| | | | | | - Tiago Soares Silva
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Katalin Toth
- Department of Cellular and Molecular MedicineFaculty of MedicineUniversity of OttawaOttawaONCanada
| | - Valerie Mongrain
- Department of NeuroscienceUniversité de Montréal and Center for Advanced Research in Sleep MedicineHôpital du Sacré‐Coeur de Montréal (CIUSSS‐NIM)MontrealQCCanada
| | | |
Collapse
|
25
|
Heir R, Stellwagen D. TNF-Mediated Homeostatic Synaptic Plasticity: From in vitro to in vivo Models. Front Cell Neurosci 2020; 14:565841. [PMID: 33192311 PMCID: PMC7556297 DOI: 10.3389/fncel.2020.565841] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Since it was first described almost 30 years ago, homeostatic synaptic plasticity (HSP) has been hypothesized to play a key role in maintaining neuronal circuit function in both developing and adult animals. While well characterized in vitro, determining the in vivo roles of this form of plasticity remains challenging. Since the discovery that the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) mediates some forms of HSP, it has been possible to probe some of the in vivo contribution of TNF-mediated HSP. Work from our lab and others has found roles for TNF-HSP in a variety of functions, including the developmental plasticity of sensory systems, models of drug addiction, and the response to psychiatric drugs.
Collapse
Affiliation(s)
- Renu Heir
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC, Canada
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, QC, Canada
| |
Collapse
|
26
|
Zhang H, Bramham CR. Arc/Arg3.1 function in long-term synaptic plasticity: Emerging mechanisms and unresolved issues. Eur J Neurosci 2020; 54:6696-6712. [PMID: 32888346 DOI: 10.1111/ejn.14958] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/18/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Arc (activity-regulated cytoskeleton-associated protein) is posited as a critical regulator of long-term synaptic plasticity at excitatory synapses, including long-term potentiation, long-term depression, inverse synaptic tagging and homoeostatic scaling, with pivotal roles in memory and postnatal cortical development. However, the mechanisms underlying the bidirectional regulation of synaptic strength are poorly understood. Here we review evidence from different plasticity paradigms, highlight outstanding issues and discuss stimulus-specific mechanisms that dictate Arc function. We propose a model in which Arc bidirectionally controls synaptic strength by coordinate regulation of AMPA-type glutamate receptor (AMPAR) trafficking and actin cytoskeletal dynamics in dendritic spines. Key to this model, Arc is proposed to function as an activity-dependent regulator of AMPAR lateral membrane diffusion and trapping at synapses.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
27
|
Goel P, Nishimura S, Chetlapalli K, Li X, Chen C, Dickman D. Distinct Target-Specific Mechanisms Homeostatically Stabilize Transmission at Pre- and Post-synaptic Compartments. Front Cell Neurosci 2020; 14:196. [PMID: 32676010 PMCID: PMC7333441 DOI: 10.3389/fncel.2020.00196] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/05/2020] [Indexed: 12/28/2022] Open
Abstract
Neurons must establish and stabilize connections made with diverse targets, each with distinct demands and functional characteristics. At Drosophila neuromuscular junctions (NMJs), synaptic strength remains stable in a manipulation that simultaneously induces hypo-innervation on one target and hyper-innervation on the other. However, the expression mechanisms that achieve this exquisite target-specific homeostatic control remain enigmatic. Here, we identify the distinct target-specific homeostatic expression mechanisms. On the hypo-innervated target, an increase in postsynaptic glutamate receptor (GluR) abundance is sufficient to compensate for reduced innervation, without any apparent presynaptic adaptations. In contrast, a target-specific reduction in presynaptic neurotransmitter release probability is reflected by a decrease in active zone components restricted to terminals of hyper-innervated targets. Finally, loss of postsynaptic GluRs on one target induces a compartmentalized, homeostatic enhancement of presynaptic neurotransmitter release called presynaptic homeostatic potentiation (PHP) that can be precisely balanced with the adaptations required for both hypo- and hyper-innervation to maintain stable synaptic strength. Thus, distinct anterograde and retrograde signaling systems operate at pre- and post-synaptic compartments to enable target-specific, homeostatic control of neurotransmission.
Collapse
|
28
|
Effects of daily L-dopa administration on learning and brain structure in older adults undergoing cognitive training: a randomised clinical trial. Sci Rep 2020; 10:5227. [PMID: 32251360 PMCID: PMC7090037 DOI: 10.1038/s41598-020-62172-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 03/04/2020] [Indexed: 11/08/2022] Open
Abstract
Cognitive aging creates major individual and societal burden, motivating search for treatment and preventive care strategies. Behavioural interventions can improve cognitive performance in older age, but effects are small. Basic research has implicated dopaminergic signalling in plasticity. We investigated whether supplementation with the dopamine-precursor L-dopa improves effects of cognitive training on performance. Sixty-three participants for this randomised, parallel-group, double-blind, placebo-controlled trial were recruited via newspaper advertisements. Inclusion criteria were: age of 65–75 years, Mini-Mental State Examination score >25, absence of serious medical conditions. Eligible subjects were randomly allocated to either receive 100/25 mg L-dopa/benserazide (n = 32) or placebo (n = 31) prior to each of twenty cognitive training sessions administered during a four-week period. Participants and staff were blinded to group assignment. Primary outcomes were latent variables of spatial and verbal fluid intelligence. Compared to the placebo group, subjects receiving L-dopa improved less in spatial intelligence (−0.267 SDs; 95%CI [−0.498, −0.036]; p = 0.024). Change in verbal intelligence did not significantly differ between the groups (−0.081 SDs, 95%CI [−0.242, 0.080]; p = 0.323). Subjects receiving L-dopa also progressed slower through the training and the groups displayed differential volumetric changes in the midbrain. No statistically significant differences were found for the secondary cognitive outcomes. Adverse events occurred for 10 (31%) and 7 (23%) participants in the active and control groups, correspondingly. The results speak against early pharmacological interventions in older healthy adults to improve broader cognitive functions by targeting the dopaminergic system and provide no support for learning-enhancing properties of L-dopa supplements in the healthy elderly. The findings warrant closer investigation about the cognitive effects of early dopamine-replacement therapy in neurological disorders. This trial was preregistered at the European Clinical Trial Registry, EudraCT#2016-000891-54 (2016-10-05).
Collapse
|
29
|
Tan HL, Roth RH, Graves AR, Cudmore RH, Huganir RL. Lamina-specific AMPA receptor dynamics following visual deprivation in vivo. eLife 2020; 9:e52420. [PMID: 32125273 PMCID: PMC7053996 DOI: 10.7554/elife.52420] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/10/2020] [Indexed: 11/19/2022] Open
Abstract
Regulation of AMPA receptor (AMPAR) expression is central to synaptic plasticity and brain function, but how these changes occur in vivo remains elusive. Here, we developed a method to longitudinally monitor the expression of synaptic AMPARs across multiple cortical layers in awake mice using two-photon imaging. We observed that baseline AMPAR expression in individual spines is highly dynamic with more dynamics in primary visual cortex (V1) layer 2/3 (L2/3) neurons than V1 L5 neurons. Visual deprivation through binocular enucleation induces a synapse-specific and depth-dependent change of synaptic AMPARs in V1 L2/3 neurons, wherein deep synapses are potentiated more than superficial synapses. The increase is specific to L2/3 neurons and absent on apical dendrites of L5 neurons, and is dependent on expression of the AMPAR-binding protein GRIP1. Our study demonstrates that specific neuronal connections, across cortical layers and even within individual neurons, respond uniquely to changes in sensory experience.
Collapse
Affiliation(s)
- Han L Tan
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Richard H Roth
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Austin R Graves
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Robert H Cudmore
- Department of Physiology and Membrane Biology, University of California School of MedicineDavisUnited States
| | - Richard L Huganir
- Solomon H Snyder Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
30
|
Dubes S, Favereaux A, Thoumine O, Letellier M. miRNA-Dependent Control of Homeostatic Plasticity in Neurons. Front Cell Neurosci 2019; 13:536. [PMID: 31866828 PMCID: PMC6906196 DOI: 10.3389/fncel.2019.00536] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/19/2019] [Indexed: 11/13/2022] Open
Abstract
Homeostatic plasticity is a form of plasticity in which neurons compensate for changes in neuronal activity through the control of key physiological parameters such as the number and the strength of their synaptic inputs and intrinsic excitability. Recent studies revealed that miRNAs, which are small non-coding RNAs repressing mRNA translation, participate in this process by controlling the translation of multiple effectors such as glutamate transporters, receptors, signaling molecules and voltage-gated ion channels. In this review, we present and discuss the role of miRNAs in both cell-wide and compartmentalized forms of homeostatic plasticity as well as their implication in pathological processes associated with homeostatic failure.
Collapse
Affiliation(s)
- Sandra Dubes
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Alexandre Favereaux
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Olivier Thoumine
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Mathieu Letellier
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| |
Collapse
|
31
|
Frank CA, James TD, Müller M. Homeostatic control of Drosophila neuromuscular junction function. Synapse 2019; 74:e22133. [PMID: 31556149 PMCID: PMC6817395 DOI: 10.1002/syn.22133] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/05/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023]
Abstract
The ability to adapt to changing internal and external conditions is a key feature of biological systems. Homeostasis refers to a regulatory process that stabilizes dynamic systems to counteract perturbations. In the nervous system, homeostatic mechanisms control neuronal excitability, neurotransmitter release, neurotransmitter receptors, and neural circuit function. The neuromuscular junction (NMJ) of Drosophila melanogaster has provided a wealth of molecular information about how synapses implement homeostatic forms of synaptic plasticity, with a focus on the transsynaptic, homeostatic modulation of neurotransmitter release. This review examines some of the recent findings from the Drosophila NMJ and highlights questions the field will ponder in coming years.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Programs in Neuroscience, Genetics, and Molecular Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Thomas D James
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
| | - Martin Müller
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Chokshi V, Gao M, Grier BD, Owens A, Wang H, Worley PF, Lee HK. Input-Specific Metaplasticity in the Visual Cortex Requires Homer1a-Mediated mGluR5 Signaling. Neuron 2019; 104:736-748.e6. [PMID: 31563294 DOI: 10.1016/j.neuron.2019.08.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 06/24/2019] [Accepted: 08/09/2019] [Indexed: 11/17/2022]
Abstract
Effective sensory processing depends on sensory experience-dependent metaplasticity, which allows homeostatic maintenance of neural network activity and preserves feature selectivity. Following a strong increase in sensory drive, plasticity mechanisms that decrease the strength of excitatory synapses are preferentially engaged to maintain stability in neural networks. Such adaptation has been demonstrated in various model systems, including mouse primary visual cortex (V1), where excitatory synapses on layer 2/3 (L2/3) neurons undergo rapid reduction in strength when visually deprived mice are reexposed to light. Here, we report that this form of plasticity is specific to intracortical inputs to V1 L2/3 neurons and depends on the activity of NMDA receptors (NMDARs) and group I metabotropic glutamate receptor 5 (mGluR5). Furthermore, we found that expression of the immediate early gene (IEG) Homer1a (H1a) and its subsequent interaction with mGluR5s are necessary for this input-specific metaplasticity.
Collapse
Affiliation(s)
- Varun Chokshi
- The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA; Cell Molecular Developmental Biology and Biophysics (CMDB) Graduate Program, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ming Gao
- The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bryce D Grier
- The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ashley Owens
- The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hui Wang
- The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Paul F Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Hey-Kyoung Lee
- The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA; Cell Molecular Developmental Biology and Biophysics (CMDB) Graduate Program, Johns Hopkins University, Baltimore, MD 21218, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
33
|
Penrod RD, Kumar J, Smith LN, McCalley D, Nentwig T, Hughes B, Barry G, Glover K, Taniguchi M, Cowan CW. Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) regulates anxiety- and novelty-related behaviors. GENES, BRAIN, AND BEHAVIOR 2019; 18:e12561. [PMID: 30761730 PMCID: PMC6692244 DOI: 10.1111/gbb.12561] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 01/19/2019] [Accepted: 02/12/2019] [Indexed: 02/06/2023]
Abstract
The activity-regulated cytoskeleton-associated protein (Arc, also known as Arg3.1) regulates glutamatergic synapse plasticity and has been linked to neuropsychiatric illness; however, its role in behaviors associated with mood and anxiety disorders remains unclear. We find that stress upregulates Arc expression in the adult mouse nucleus accumbens (NAc)-a brain region implicated in mood and anxiety behaviors. Global Arc knockout mice have altered AMPAR-subunit surface levels in the adult NAc, and the Arc-deficient mice show reductions in anxiety-like behavior, deficits in social novelty preference, and antidepressive-like behavior. Viral-mediated expression of Arc in the adult NAc of male, global Arc KO mice restores normal levels of anxiety-like behavior in the elevated plus maze (EPM). Consistent with this finding, viral-mediated reduction of Arc in the adult NAc reduces anxiety-like behavior in male, but not female, mice in the EPM. NAc-specific reduction of Arc also produced significant deficits in both object and social novelty preference tasks. Together our findings indicate that Arc is essential for regulating normal mood- and anxiety-related behaviors and novelty discrimination, and that Arc's function within the adult NAc contributes to these behavioral effects.
Collapse
Affiliation(s)
- Rachel D. Penrod
- Departments of Neuroscience and Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston SC 29425
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA 02478
| | - Jaswinder Kumar
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA 02478
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas TX 75390-9070
| | - Laura N. Smith
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA 02478
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas TX 75390-9070
| | - Daniel McCalley
- Departments of Neuroscience and Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston SC 29425
| | - Todd Nentwig
- Departments of Neuroscience and Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston SC 29425
| | - Brandon Hughes
- Departments of Neuroscience and Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston SC 29425
| | - Gabriella Barry
- Department of Science and Mathematics, Honors College, College of Charleston, Charleston SC 29424
| | - Kelsey Glover
- Department of Science and Mathematics, Honors College, College of Charleston, Charleston SC 29424
| | - Makoto Taniguchi
- Departments of Neuroscience and Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston SC 29425
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA 02478
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas TX 75390-9070
| | - Christopher W. Cowan
- Departments of Neuroscience and Psychiatry & Behavioral Sciences, Medical University of South Carolina, Charleston SC 29425
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont MA 02478
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas TX 75390-9070
| |
Collapse
|
34
|
Disruption of NMDAR Function Prevents Normal Experience-Dependent Homeostatic Synaptic Plasticity in Mouse Primary Visual Cortex. J Neurosci 2019; 39:7664-7673. [PMID: 31413075 DOI: 10.1523/jneurosci.2117-18.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/21/2022] Open
Abstract
Homeostatic regulation of synaptic strength allows for maintenance of neural activity within a dynamic range for proper circuit function. There are largely two distinct modes of synaptic plasticity that allow for homeostatic adaptation of cortical circuits: synaptic scaling and sliding threshold (BCM theory). Previous findings suggest that the induction of synaptic scaling is not prevented by blocking NMDARs, whereas the sliding threshold model posits that the synaptic modification threshold of LTP and LTD readjusts with activity and thus the outcome of synaptic plasticity is NMDAR dependent. Although synaptic scaling and sliding threshold have been considered two distinct mechanisms, there are indications from recent studies that these two modes of homeostatic plasticity may interact or that they may operate under two distinct activity regimes. Here, we report using both sexes of mouse that acute genetic knock-out of the obligatory subunit of NMDAR or acute pharmacological block of NMDAR prevents experience-dependent homeostatic regulation of AMPAR-mediated miniature EPSCs in layer 2/3 of visual cortex. This was not due to gross changes in postsynaptic neuronal activity with inhibiting NMDAR function as determine by c-Fos expression and two-photon Ca2+ imaging in awake mice. Our results suggest that experience-dependent homeostatic regulation of intact cortical circuits is mediated by NMDAR-dependent plasticity mechanisms, which supports a sliding threshold model of homeostatic adaptation.SIGNIFICANCE STATEMENT Prolonged changes in sensory experience lead to homeostatic adaptation of excitatory synaptic strength in sensory cortices. Both sliding threshold and synaptic scaling models can account for the observed homeostatic synaptic plasticity. Here we report that visual experience-dependent homeostatic plasticity of excitatory synapses observed in superficial layers of visual cortex is dependent on NMDAR function. In particular, both strengthening of synapses induced by visual deprivation and the subsequent weakening by reinstatement of visual experience were prevented in the absence of functional NMDARs. Our results suggest that sensory experience-dependent homeostatic adaptation depends on NMDARs, which supports the sliding threshold model of plasticity and input-specific homeostatic control observed in vivo.
Collapse
|
35
|
Bell M, Bartol T, Sejnowski T, Rangamani P. Dendritic spine geometry and spine apparatus organization govern the spatiotemporal dynamics of calcium. J Gen Physiol 2019; 151:1017-1034. [PMID: 31324651 PMCID: PMC6683673 DOI: 10.1085/jgp.201812261] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 05/10/2019] [Accepted: 06/17/2019] [Indexed: 01/27/2023] Open
Abstract
Dendritic spines are small subcompartments that protrude from the dendrites of neurons and are important for signaling activity and synaptic communication. These subcompartments have been characterized to have different shapes. While it is known that these shapes are associated with spine function, the specific nature of these shape-function relationships is not well understood. In this work, we systematically investigated the relationship between the shape and size of both the spine head and spine apparatus, a specialized endoplasmic reticulum compartment within the spine head, in modulating rapid calcium dynamics using mathematical modeling. We developed a spatial multicompartment reaction-diffusion model of calcium dynamics in three dimensions with various flux sources, including N-methyl-D-aspartate receptors (NMDARs), voltage-sensitive calcium channels (VSCCs), and different ion pumps on the plasma membrane. Using this model, we make several important predictions. First, the volume to surface area ratio of the spine regulates calcium dynamics. Second, membrane fluxes impact calcium dynamics temporally and spatially in a nonlinear fashion. Finally, the spine apparatus can act as a physical buffer for calcium by acting as a sink and rescaling the calcium concentration. These predictions set the stage for future experimental investigations of calcium dynamics in dendritic spines.
Collapse
Affiliation(s)
- Miriam Bell
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA
| | - Tom Bartol
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA
| | - Terrence Sejnowski
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA
- Division of Biological Sciences, University of California, San Diego, San Diego, CA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA
| |
Collapse
|
36
|
Wang G, Zhong J, Guttieres D, Man HY. Non-scaling regulation of AMPA receptors in homeostatic synaptic plasticity. Neuropharmacology 2019; 158:107700. [PMID: 31283924 DOI: 10.1016/j.neuropharm.2019.107700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/10/2019] [Accepted: 07/04/2019] [Indexed: 11/28/2022]
Abstract
Homeostatic synaptic plasticity (HSP) as an activity-dependent negative feedback regulation of synaptic strength plays important roles in the maintenance of neuronal and neural circuitry stability. A primary cellular substrate for HSP expression is alterations in synaptic accumulation of glutamatergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR). It is widely believed that during HSP, AMPAR accumulation changes with the same proportion at each synapse of a neuron, a process known as synaptic scaling. However, direct evidence on AMPAR synaptic scaling remains largely lacking. Here we report a direct examination of inactivity-induced homeostatic scaling of AMPAR at individual synapse by live-imaging. Surprisingly, instead of uniform up-scaling, a scattered pattern of changes in synaptic AMPAR was observed in cultured rat hippocampal neurons. While the majority of synapses showed up-regulation after activity inhibition, a reduction of AMPAR could be detected in certain synapses. More importantly, among the up-regulated synapses, a wide range of AMPAR changes was observed in synapses of the same neuron. We also found that synapses with higher levels of pre-existing AMPAR tend to be up-regulated by lesser extents, whereas the locations of synapses relative to the soma seem not affecting AMPAR scaling strengths. In addition, we observed strong competition between neighboring synapses during HSP. These results reveal that synaptic AMPAR may not be scaled during HSP, suggesting novel molecular mechanisms for information processing and storage at synapses.
Collapse
Affiliation(s)
- Guan Wang
- Department of Biology, Boston University, Boston, MA, USA; School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jia Zhong
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA; Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
37
|
Lesuis SL, Hoeijmakers L, Korosi A, de Rooij SR, Swaab DF, Kessels HW, Lucassen PJ, Krugers HJ. Vulnerability and resilience to Alzheimer's disease: early life conditions modulate neuropathology and determine cognitive reserve. Alzheimers Res Ther 2018; 10:95. [PMID: 30227888 PMCID: PMC6145191 DOI: 10.1186/s13195-018-0422-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with a high prevalence among the elderly and a huge personal and societal impact. Recent epidemiological studies have indicated that the incidence and age of onset of sporadic AD can be modified by lifestyle factors such as education, exercise, and (early) stress exposure. Early life adversity is known to promote cognitive decline at a later age and to accelerate aging, which are both primary risk factors for AD. In rodent models, exposure to 'negative' or 'positive' early life experiences was recently found to modulate various measures of AD neuropathology, such as amyloid-beta levels and cognition at later ages. Although there is emerging interest in understanding whether experiences during early postnatal life also modulate AD risk in humans, the mechanisms and possible substrates underlying these long-lasting effects remain elusive. METHODS We review literature and discuss the role of early life experiences in determining later age and AD-related processes from a brain and cognitive 'reserve' perspective. We focus on rodent studies and the identification of possible early determinants of later AD vulnerability or resilience in relation to early life adversity/enrichment. RESULTS Potential substrates and mediators of early life experiences that may influence the development of AD pathology and cognitive decline are: programming of the hypothalamic-pituitary-adrenal axis, priming of the neuroinflammatory response, dendritic and synaptic complexity and function, overall brain plasticity, and proteins such as early growth response protein 1 (EGR1), activity regulated cytoskeleton-associated protein (Arc), and repressor element-1 silencing transcription factor (REST). CONCLUSIONS We conclude from these rodent studies that the early postnatal period is an important and sensitive phase that influences the vulnerability to develop AD pathology. Yet translational studies are required to investigate whether early life experiences also modify AD development in human studies, and whether similar molecular mediators can be identified in the sensitivity to develop AD in humans.
Collapse
Affiliation(s)
- Sylvie L. Lesuis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Susanne R. de Rooij
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics & Bio informatics, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Dick F. Swaab
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Helmut W. Kessels
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Department of Cellular and Computational Neuroscience, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
38
|
El-Boustani S, Ip JPK, Breton-Provencher V, Knott GW, Okuno H, Bito H, Sur M. Locally coordinated synaptic plasticity of visual cortex neurons in vivo. Science 2018; 360:1349-1354. [PMID: 29930137 DOI: 10.1126/science.aao0862] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 03/07/2018] [Accepted: 05/08/2018] [Indexed: 11/02/2022]
Abstract
Plasticity of cortical responses in vivo involves activity-dependent changes at synapses, but the manner in which different forms of synaptic plasticity act together to create functional changes in neurons remains unknown. We found that spike timing-induced receptive field plasticity of visual cortex neurons in mice is anchored by increases in the synaptic strength of identified spines. This is accompanied by a decrease in the strength of adjacent spines on a slower time scale. The locally coordinated potentiation and depression of spines involves prominent AMPA receptor redistribution via targeted expression of the immediate early gene product Arc. Hebbian strengthening of activated synapses and heterosynaptic weakening of adjacent synapses thus cooperatively orchestrate cell-wide plasticity of functional neuronal responses.
Collapse
Affiliation(s)
- Sami El-Boustani
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Jacque P K Ip
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vincent Breton-Provencher
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Graham W Knott
- Bio Electron Microscopy Laboratory, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Hiroyuki Okuno
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto 606-8507, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
39
|
Hirai S, Hotta K, Okado H. Developmental Roles and Evolutionary Significance of AMPA-Type Glutamate Receptors. Bioessays 2018; 40:e1800028. [PMID: 30058076 DOI: 10.1002/bies.201800028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/02/2018] [Indexed: 11/07/2022]
Abstract
Organogenesis and metamorphosis require the intricate orchestration of multiple types of cellular interactions and signaling pathways. Glutamate (Glu) is an excitatory extracellular signaling molecule in the nervous system, while Ca2+ is a major intracellular signaling molecule. The first Glu receptors to be cloned are Ca2+ -permeable receptors in mammalian brains. Although recent studies have focused on Glu signaling in synaptic mechanisms of the mammalian central nervous system, it is unclear how this signaling functions in development. Our recent article demonstrated that Ca2+ -permeable AMPA-type Glu receptors (GluAs) are essential for formation of a photosensitive organ, development of some neurons, and metamorphosis, including tail absorption and body axis rotation, in ascidian embryos. Based on findings in these embryos and mammalian brains, we formed several hypotheses regarding the evolution of GluAs, the non-synaptic function of Glu, the origin of GluA-positive neurons, and the neuronal network that controls metamorphosis in ascidians.
Collapse
Affiliation(s)
- Shinobu Hirai
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-0057, Japan
| | - Kohji Hotta
- Faculty of Science and Technology, Department of Biosciences and Informatics, Keio University, Kohoku, Yokohama, 223-8522, Japan
| | - Haruo Okado
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-0057, Japan
| |
Collapse
|
40
|
Soares C, Lee KFH, Béïque JC. Metaplasticity at CA1 Synapses by Homeostatic Control of Presynaptic Release Dynamics. Cell Rep 2018; 21:1293-1303. [PMID: 29091767 DOI: 10.1016/j.celrep.2017.10.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/16/2017] [Accepted: 10/05/2017] [Indexed: 10/18/2022] Open
Abstract
Hebbian and homeostatic forms of plasticity operate on different timescales to regulate synaptic strength. The degree of mechanistic overlap between these processes and their mutual influence are still incompletely understood. Here, we report that homeostatic synaptic strengthening induced by prolonged network inactivity compromised the ability of CA1 synapses to exhibit LTP. This effect could not be accounted for by an obvious deficit in the postsynaptic capacity for LTP expression, since neither the fraction of silent synapses nor the ability to induce LTP by two-photon glutamate uncaging were reduced by the homeostatic process. Rather, optical quantal analysis reveals that homeostatically strengthened synapses display a reduced capacity to maintain glutamate release fidelity during repetitive stimulation, ultimately impeding the induction, and thus expression, of LTP. By regulating the short-term dynamics of glutamate release, the homeostatic process thus influences key aspects of dynamic network function and exhibits features of metaplasticity.
Collapse
Affiliation(s)
- Cary Soares
- Neuroscience Graduate Program, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Kevin F H Lee
- Neuroscience Graduate Program, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jean-Claude Béïque
- Neuroscience Graduate Program, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Canadian Partnership for Stroke Recovery, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Brain and Mind Research Institute's Centre for Neural Dynamics, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
41
|
Grassi D, Franz H, Vezzali R, Bovio P, Heidrich S, Dehghanian F, Lagunas N, Belzung C, Krieglstein K, Vogel T. Neuronal Activity, TGFβ-Signaling and Unpredictable Chronic Stress Modulate Transcription of Gadd45 Family Members and DNA Methylation in the Hippocampus. Cereb Cortex 2018; 27:4166-4181. [PMID: 28444170 DOI: 10.1093/cercor/bhx095] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/04/2017] [Indexed: 02/06/2023] Open
Abstract
Neuronal activity is altered in several neurological and psychiatric diseases. Upon depolarization not only neurotransmitters are released but also cytokines and other activators of signaling cascades. Unraveling their complex implication in transcriptional control in receiving cells will contribute to understand specific central nervous system (CNS) pathologies and will be of therapeutically interest. In this study we depolarized mature hippocampal neurons in vitro using KCl and revealed increased release not only of brain-derived neurotrophic factor (BDNF) but also of transforming growth factor beta (TGFB). Neuronal activity together with BDNF and TGFB controls transcription of DNA modifying enzymes specifically members of the DNA-damage-inducible (Gadd) family, Gadd45a, Gadd45b, and Gadd45g. MeDIP followed by massive parallel sequencing and transcriptome analyses revealed less DNA methylation upon KCl treatment. Psychiatric disorder-related genes, namely Tshz1, Foxn3, Jarid2, Per1, Map3k5, and Arc are transcriptionally activated and demethylated upon neuronal activation. To analyze whether misexpression of Gadd45 family members are associated with psychiatric diseases, we applied unpredictable chronic mild stress (UCMS) as established model for depression to mice. UCMS led to reduced expression of Gadd45 family members. Taken together, our data demonstrate that Gadd45 family members are new putative targets for UCMS treatments.
Collapse
Affiliation(s)
- Daniela Grassi
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany.,Department of Basic Biomedical Sciences, Faculty of Biomedical Science and Health, Universidad Europea de Madrid, Madrid, Spain
| | - Henriette Franz
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Riccardo Vezzali
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Patrick Bovio
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Stefanie Heidrich
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Fariba Dehghanian
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Natalia Lagunas
- Inserm U 930, Université François Rabelais, 37200 Tours, France
| | | | - Kerstin Krieglstein
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Tanja Vogel
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
42
|
Bourke AM, Bowen AB, Kennedy MJ. New approaches for solving old problems in neuronal protein trafficking. Mol Cell Neurosci 2018; 91:48-66. [PMID: 29649542 DOI: 10.1016/j.mcn.2018.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 11/16/2022] Open
Abstract
Fundamental cellular properties are determined by the repertoire and abundance of proteins displayed on the cell surface. As such, the trafficking mechanisms for establishing and maintaining the surface proteome must be tightly regulated for cells to respond appropriately to extracellular cues, yet plastic enough to adapt to ever-changing environments. Not only are the identity and abundance of surface proteins critical, but in many cases, their regulated spatial positioning within surface nanodomains can greatly impact their function. In the context of neuronal cell biology, surface levels and positioning of ion channels and neurotransmitter receptors play essential roles in establishing important properties, including cellular excitability and synaptic strength. Here we review our current understanding of the trafficking pathways that control the abundance and localization of proteins important for synaptic function and plasticity, as well as recent technological advances that are allowing the field to investigate protein trafficking with increasing spatiotemporal precision.
Collapse
Affiliation(s)
- Ashley M Bourke
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Aaron B Bowen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States.
| |
Collapse
|
43
|
Synapse development organized by neuronal activity-regulated immediate-early genes. Exp Mol Med 2018; 50:1-7. [PMID: 29628504 PMCID: PMC5938016 DOI: 10.1038/s12276-018-0025-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
Classical studies have shown that neuronal immediate-early genes (IEGs) play important roles in synaptic processes critical for key brain functions. IEGs are transiently activated and rapidly upregulated in discrete neurons in response to a wide variety of cellular stimuli, and they are uniquely involved in various aspects of synapse development. In this review, we summarize recent studies of a subset of neuronal IEGs in regulating synapse formation, transmission, and plasticity. We also discuss how the dysregulation of neuronal IEGs is associated with the onset of various brain disorders and pinpoint key outstanding questions that should be addressed in this field. Immediate-early genes (IEGs), genes that are rapidly and transiently activated by cellular stimuli, regulate the interactions between neurons and key brain functions. Ji Won Um and colleagues at Daegu Gyeongbuk Institute of Science and Technology in South Korea review recent studies on three IEGs that are activated by neuronal activity and highlight their contribution to neuronal excitability and cognitive behaviors. These genes rely on different molecular mechanisms to regulate neuronal receptors and the structure of synapses. Research in mice lacking any one of these IEGs reveals their contribution to learning and memory as well as to some behavioral abnormalities associated with neuropsychiatric disorders. Further research into the activity of IEGs will advance our understanding of how a neuron’s environment influences brain development and disease.
Collapse
|
44
|
Chowdhury D, Hell JW. Homeostatic synaptic scaling: molecular regulators of synaptic AMPA-type glutamate receptors. F1000Res 2018; 7:234. [PMID: 29560257 PMCID: PMC5832907 DOI: 10.12688/f1000research.13561.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2018] [Indexed: 01/31/2023] Open
Abstract
The ability of neurons and circuits to maintain their excitability and activity levels within the appropriate dynamic range by homeostatic mechanisms is fundamental for brain function. Neuronal hyperactivity, for instance, could cause seizures. One such homeostatic process is synaptic scaling, also known as synaptic homeostasis. It involves a negative feedback process by which neurons adjust (scale) their postsynaptic strength over their whole synapse population to compensate for increased or decreased overall input thereby preventing neuronal hyper- or hypoactivity that could otherwise result in neuronal network dysfunction. While synaptic scaling is well-established and critical, our understanding of the underlying molecular mechanisms is still in its infancy. Homeostatic adaptation of synaptic strength is achieved through upregulation (upscaling) or downregulation (downscaling) of the functional availability of AMPA-type glutamate receptors (AMPARs) at postsynaptic sites. Understanding how synaptic AMPARs are modulated in response to alterations in overall neuronal activity is essential to gain valuable insights into how neuronal networks adapt to changes in their environment, as well as the genesis of an array of neurological disorders. Here we discuss the key molecular mechanisms that have been implicated in tuning the synaptic abundance of postsynaptic AMPARs in order to maintain synaptic homeostasis.
Collapse
Affiliation(s)
| | - Johannes W Hell
- Department of Pharmacology, University of California Davis, Davis, California, USA
| |
Collapse
|
45
|
Gestational exposure to inorganic arsenic (iAs3+) alters glutamate disposition in the mouse hippocampus and ionotropic glutamate receptor expression leading to memory impairment. Arch Toxicol 2017; 92:1037-1048. [DOI: 10.1007/s00204-017-2111-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 11/08/2017] [Indexed: 12/17/2022]
|
46
|
Turrigiano GG. The dialectic of Hebb and homeostasis. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0258. [PMID: 28093556 DOI: 10.1098/rstb.2016.0258] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2016] [Indexed: 01/12/2023] Open
Abstract
It has become widely accepted that homeostatic and Hebbian plasticity mechanisms work hand in glove to refine neural circuit function. Nonetheless, our understanding of how these fundamentally distinct forms of plasticity compliment (and under some circumstances interfere with) each other remains rudimentary. Here, I describe some of the recent progress of the field, as well as some of the deep puzzles that remain. These include unravelling the spatial and temporal scales of different homeostatic and Hebbian mechanisms, determining which aspects of network function are under homeostatic control, and understanding when and how homeostatic and Hebbian mechanisms must be segregated within neural circuits to prevent interference.This article is part of the themed issue 'Integrating Hebbian and homeostatic plasticity'.
Collapse
|
47
|
Yee AX, Hsu YT, Chen L. A metaplasticity view of the interaction between homeostatic and Hebbian plasticity. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0155. [PMID: 28093549 DOI: 10.1098/rstb.2016.0155] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2016] [Indexed: 01/25/2023] Open
Abstract
Hebbian and homeostatic plasticity are two major forms of plasticity in the nervous system: Hebbian plasticity provides a synaptic basis for associative learning, whereas homeostatic plasticity serves to stabilize network activity. While achieving seemingly very different goals, these two types of plasticity interact functionally through overlapping elements in their respective mechanisms. Here, we review studies conducted in the mammalian central nervous system, summarize known circuit and molecular mechanisms of homeostatic plasticity, and compare these mechanisms with those that mediate Hebbian plasticity. We end with a discussion of 'local' homeostatic plasticity and the potential role of local homeostatic plasticity as a form of metaplasticity that modulates a neuron's future capacity for Hebbian plasticity.This article is part of the themed issue 'Integrating Hebbian and homeostatic plasticity'.
Collapse
Affiliation(s)
- Ada X Yee
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Yu-Tien Hsu
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Lu Chen
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| |
Collapse
|
48
|
Chowdhury D, Turner M, Patriarchi T, Hergarden AC, Anderson D, Zhang Y, Sun J, Chen CY, Ames JB, Hell JW. Ca 2+/calmodulin binding to PSD-95 mediates homeostatic synaptic scaling down. EMBO J 2017; 37:122-138. [PMID: 29118000 DOI: 10.15252/embj.201695829] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 09/07/2017] [Accepted: 10/02/2017] [Indexed: 11/09/2022] Open
Abstract
Postsynaptic density protein-95 (PSD-95) localizes AMPA-type glutamate receptors (AMPARs) to postsynaptic sites of glutamatergic synapses. Its postsynaptic displacement is necessary for loss of AMPARs during homeostatic scaling down of synapses. Here, we demonstrate that upon Ca2+ influx, Ca2+/calmodulin (Ca2+/CaM) binding to the N-terminus of PSD-95 mediates postsynaptic loss of PSD-95 and AMPARs during homeostatic scaling down. Our NMR structural analysis identified E17 within the PSD-95 N-terminus as important for binding to Ca2+/CaM by interacting with R126 on CaM. Mutating E17 to R prevented homeostatic scaling down in primary hippocampal neurons, which is rescued via charge inversion by ectopic expression of CaMR126E, as determined by analysis of miniature excitatory postsynaptic currents. Accordingly, increased binding of Ca2+/CaM to PSD-95 induced by a chronic increase in Ca2+ influx is a critical molecular event in homeostatic downscaling of glutamatergic synaptic transmission.
Collapse
Affiliation(s)
| | - Matthew Turner
- Department of Chemistry, University of California, Davis, CA, USA
| | | | - Anne C Hergarden
- Department of Pharmacology, University of California, Davis, CA, USA
| | - David Anderson
- Department of Chemistry, University of California, Davis, CA, USA
| | - Yonghong Zhang
- Department of Chemistry, University of Texas, Edinburgh, TX, USA
| | - Junqing Sun
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Chao-Yin Chen
- Department of Pharmacology, University of California, Davis, CA, USA
| | - James B Ames
- Department of Chemistry, University of California, Davis, CA, USA
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
49
|
Somatic Arc protein expression in hippocampal granule cells is increased in response to environmental change but independent of task-specific learning. Sci Rep 2017; 7:12477. [PMID: 28963515 PMCID: PMC5622137 DOI: 10.1038/s41598-017-12583-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 09/08/2017] [Indexed: 01/04/2023] Open
Abstract
Activated neurons express immediate-early genes, such as Arc. Expression of Arc in the hippocampal granule cell layer, an area crucial for spatial learning and memory, is increased during acquisition of spatial learning; however, it is unclear whether this effect is related to the task-specific learning process or to nonspecific aspects of the testing procedure (e.g. exposure to the testing apparatus and exploration of the environment). Herein, we show that Arc-positive cells numbers are increased to the same extent in the granule cell layer after both acquisition of a single spatial learning event in the active place avoidance task and exploration of the testing environment, as compared to naïve (i.e. caged) mice. Repeated exposure the testing apparatus and environment did not reduce Arc expression. Furthermore, Arc expression did not correlate with performance in both adult and aged animals, suggesting that exploration of the testing environment, rather than the specific acquisition of the active place avoidance task, induces Arc expression in the dentate granule cell layer. These findings thus suggest that Arc is an experience-induced immediate-early gene.
Collapse
|
50
|
Okuno H, Minatohara K, Bito H. Inverse synaptic tagging: An inactive synapse-specific mechanism to capture activity-induced Arc/arg3.1 and to locally regulate spatial distribution of synaptic weights. Semin Cell Dev Biol 2017; 77:43-50. [PMID: 28939038 DOI: 10.1016/j.semcdb.2017.09.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/15/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022]
Abstract
Long-lasting forms of synaptic plasticity such as long-term potentiation (LTP) and long-term depression (LTD) are fundamental cellular mechanisms underlying learning and memory. The synaptic tagging and capture (STC) hypothesis has provided a theoretical framework on how products of activity-dependent genes may interact with potentiated synapses to facilitate and maintain such long-lasting synaptic plasticity. Although Arc/arg3.1 was initially assumed to participate in STC processes during LTP, accumulating evidence indicated that Arc/arg3.1 might rather contribute in weakening of synaptic weights than in their strengthening. In particular, analyses of Arc/Arg3.1 protein dynamics and function in the dendrites after plasticity-inducing stimuli have revealed a new type of inactivity-dependent redistribution of synaptic weights, termed "inverse synaptic tagging". The original synaptic tagging and inverse synaptic tagging likely co-exist and are mutually non-exclusive mechanisms, which together may help orchestrate the redistribution of synaptic weights and promote the enhancement and maintenance of their contrast between potentiated and non-potentiated synapses during the late phase of long-term synaptic plasticity. In this review, we describe the inverse synaptic tagging mechanism that controls synaptic dynamics of Arc/Arg3.1, an immediate early gene product which is captured and preferentially targeted to non-potentiated synapses, and discuss its impact on neuronal circuit refinement and cognitive function.
Collapse
Affiliation(s)
- Hiroyuki Okuno
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Keiichiro Minatohara
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|