1
|
Koesters AG, Rich MM, Engisch KL. Homeostatic Synaptic Plasticity of Miniature Excitatory Postsynaptic Currents in Mouse Cortical Cultures Requires Neuronal Rab3A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.14.544980. [PMID: 39071374 PMCID: PMC11275788 DOI: 10.1101/2023.06.14.544980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Following prolonged activity blockade, amplitudes of miniature excitatory postsynaptic currents (mEPSCs) increase, a form of plasticity termed "homeostatic synaptic plasticity." We previously showed that a presynaptic protein, the small GTPase Rab3A, is required for full expression of the increase in miniature endplate current amplitudes following prolonged blockade of action potential activity at the mouse neuromuscular junction in vivo (Wang et al., 2011), but it is unknown whether this form of Rab3A-dependent homeostatic plasticity shares any characteristics with central synapses. We show here that homeostatic synaptic plasticity of mEPSCs is impaired in mouse cortical neuron cultures prepared from Rab3A-/- and mutant mice expressing a single point mutation of Rab3A, Rab3A Earlybird mice. To determine if Rab3A is involved in the well-established homeostatic increase in postsynaptic AMPA-type receptors (AMPARs), we performed a series of experiments in which electrophysiological recordings of mEPSCs and confocal imaging of synaptic AMPAR immunofluorescence were assessed within the same cultures. We found that the increase in postsynaptic AMPAR levels was more variable than that of mEPSC amplitudes, suggesting other factors may contribute to the homeostatic increase in synaptic strength. Finally, we demonstrate that Rab3A is acting in neurons because only selective loss of Rab3A in neurons, not glia, disrupted the homeostatic increase in mEPSC amplitudes. This is the first demonstration that a protein thought to function presynaptically is required for homeostatic synaptic plasticity of quantal size.
Collapse
Affiliation(s)
- Andrew G. Koesters
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mark M. Rich
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45345
| | - Kathrin L. Engisch
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine and the College of Science and Mathematics, Wright State University, Dayton, OH 45435
| |
Collapse
|
2
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
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
|
Sun SED, Levenstein D, Li B, Mandelberg N, Chenouard N, Suutari BS, Sanchez S, Tian G, Rinzel J, Buzsáki G, Tsien RW. Synaptic homeostasis transiently leverages Hebbian mechanisms for a multiphasic response to inactivity. Cell Rep 2024; 43:113839. [PMID: 38507409 DOI: 10.1016/j.celrep.2024.113839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/19/2023] [Accepted: 02/05/2024] [Indexed: 03/22/2024] Open
Abstract
Homeostatic regulation of synapses is vital for nervous system function and key to understanding a range of neurological conditions. Synaptic homeostasis is proposed to operate over hours to counteract the destabilizing influence of long-term potentiation (LTP) and long-term depression (LTD). The prevailing view holds that synaptic scaling is a slow first-order process that regulates postsynaptic glutamate receptors and fundamentally differs from LTP or LTD. Surprisingly, we find that the dynamics of scaling induced by neuronal inactivity are not exponential or monotonic, and the mechanism requires calcineurin and CaMKII, molecules dominant in LTD and LTP. Our quantitative model of these enzymes reconstructs the unexpected dynamics of homeostatic scaling and reveals how synapses can efficiently safeguard future capacity for synaptic plasticity. This mechanism of synaptic adaptation supports a broader set of homeostatic changes, including action potential autoregulation, and invites further inquiry into how such a mechanism varies in health and disease.
Collapse
Affiliation(s)
- Simón E D Sun
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Daniel Levenstein
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, 3810 University Street, Montreal, QC, Canada
| | - Boxing Li
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510810, China
| | - Nataniel Mandelberg
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Nicolas Chenouard
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA; Sorbonne Université, INSERM U1127, UMR CNRS 7225, Institut du Cerveau (ICM), 47 bld de l'hôpital, 75013 Paris, France
| | - Benjamin S Suutari
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Sandrine Sanchez
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Guoling Tian
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - John Rinzel
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - György Buzsáki
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA
| | - Richard W Tsien
- Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|
5
|
Zinchenko VP, Dolgacheva LP, Tuleukhanov ST. Calcium-permeable AMPA and kainate receptors of GABAergic neurons. Biophys Rev 2024; 16:165-171. [PMID: 38737208 PMCID: PMC11078900 DOI: 10.1007/s12551-024-01184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/16/2024] [Indexed: 05/14/2024] Open
Abstract
This Commentary presents a brief discussion of the action of glutamate calcium permeable receptors present with neurons on the release of the neurotransmitter gamma-aminobutyric acid (GABA). In particular, Glutamate sensitive Kainic Acid Receptors (KARs) and α-Amino-3-hydroxy-5-Methyl-4-isoxazole Propionic Acid Receptor (AMPARs) are Na+ channels that typically cause neuronal cells to depolarize and release GABA. Some of these receptors are also permeable to Ca2+ and are hence involved in the calcium-dependent release of GABA neurotransmitters. Calcium-permeable kainate and AMPA receptors (CP-KARs and CP-AMPARs) are predominantly located in GABAergic neurons in the mature brain and their primary role is to regulate GABA release. AMPARs which do not contain the GluA2 subunit are mainly localized in the postsynaptic membrane. CP-KAR receptors are located mainly in the presynapse. GABAergic neurons expressing CP-KARs and CP-AMPARs respond to excitation earlier and faster, suppressing hyperexcitation of other neurons by the advanced GABA release due to an early rapid [Ca2+]i increase. CP-AMPARs have demonstrated a more pronounced impact on plasticity compared to NMDARs because of their capacity to elevate intracellular Ca2+ levels independently of voltage. GABAergic neurons that express CP-AMPARs contribute to the disinhibition of glutamatergic neurons by suppressing GABAergic neurons that express CP-KARs. Hence, the presence of glutamate CP-KARs and CP-AMPARs is crucial in governing hyperexcitation and synaptic plasticity in GABAergic neurons.
Collapse
Affiliation(s)
- V. P. Zinchenko
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Institutskaya 3, Pushchino, Russia 142290
| | - L. P. Dolgacheva
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Institutskaya 3, Pushchino, Russia 142290
| | - S. T. Tuleukhanov
- Al-Farabi Kazakh National University, 050040 Al-Farabi Avenue 71, Almaty, Republic of Kazakhstan
| |
Collapse
|
6
|
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: 0] [Impact Index Per Article: 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
|
7
|
Wakazono Y, Midorikawa R, Takamiya K. Temporal and quantitative analysis of the functional expression of Ca 2+-permeable AMPA receptors during LTP. Neurosci Res 2024; 198:21-29. [PMID: 37429464 DOI: 10.1016/j.neures.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
In the present study, we attempted to temporally and quantitatively analyze the functional contributions of Ca2+-permeable (CP) α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) during long-term potentiation (LTP) expression using electrophysiological and pharmacological approaches. In hippocampal CA1 neurons, using 1-naphthyl acetyl spermine (NASPM), a CP-AMPAR antagonist, we began by demonstrating that NASPM-sensitive components, probably including the GluA1 homomer, functionally contributed to about 15% of AMPAR-mediated EPSC amplitude in basal conditions. Then, when NASPM was treated at different time points (3-30 min) after LTP induction, it was found that LTP was almost completely impaired at 3 or 10 min but maintained at 20 or 30 min, although its potentiation was reduced. Further temporal and quantitative analysis revealed that the functional expression of CP-AMPARs began increasing approximately 20 min after LTP induction and reached more than twice the basal level at 30 min. These results suggest that CP-AMPARs in the first 3-10 min of LTP might play an important role in LTP maintenance. Moreover, their decay time was also significantly increased at 30 min, suggesting that CP-AMPARs changed not only quantitatively in LTP but also qualitatively.
Collapse
Affiliation(s)
- Yoshihiko Wakazono
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Japan; Laboratory of Biophysical Research, Frontier Science Research Center, University of Miyazaki, Japan
| | - Ryosuke Midorikawa
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Japan
| | - Kogo Takamiya
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Japan; Laboratory of Biophysical Research, Frontier Science Research Center, University of Miyazaki, Japan.
| |
Collapse
|
8
|
Yuan X, Puvogel S, van Rhijn JR, Ciptasari U, Esteve-Codina A, Meijer M, Rouschop S, van Hugte EJH, Oudakker A, Schoenmaker C, Frega M, Schubert D, Franke B, Nadif Kasri N. A human in vitro neuronal model for studying homeostatic plasticity at the network level. Stem Cell Reports 2023; 18:2222-2239. [PMID: 37863044 PMCID: PMC10679660 DOI: 10.1016/j.stemcr.2023.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/22/2023] Open
Abstract
Mechanisms that underlie homeostatic plasticity have been extensively investigated at single-cell levels in animal models, but are less well understood at the network level. Here, we used microelectrode arrays to characterize neuronal networks following induction of homeostatic plasticity in human induced pluripotent stem cell (hiPSC)-derived glutamatergic neurons co-cultured with rat astrocytes. Chronic suppression of neuronal activity through tetrodotoxin (TTX) elicited a time-dependent network re-arrangement. Increased expression of AMPA receptors and the elongation of axon initial segments were associated with increased network excitability following TTX treatment. Transcriptomic profiling of TTX-treated neurons revealed up-regulated genes related to extracellular matrix organization, while down-regulated genes related to cell communication; also astrocytic gene expression was found altered. Overall, our study shows that hiPSC-derived neuronal networks provide a reliable in vitro platform to measure and characterize homeostatic plasticity at network and single-cell levels; this platform can be extended to investigate altered homeostatic plasticity in brain disorders.
Collapse
Affiliation(s)
- Xiuming Yuan
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Sofía Puvogel
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Jon-Ruben van Rhijn
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Ummi Ciptasari
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Mandy Meijer
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Simon Rouschop
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Eline J H van Hugte
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Astrid Oudakker
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Chantal Schoenmaker
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Monica Frega
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, the Netherlands
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Barbara Franke
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, 6500 HB Nijmegen, the Netherlands.
| |
Collapse
|
9
|
Ekins TG, Brooks I, Kailasa S, Rybicki-Kler C, Jedrasiak-Cape I, Donoho E, Mashour GA, Rech J, Ahmed OJ. Cellular rules underlying psychedelic control of prefrontal pyramidal neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563334. [PMID: 37961554 PMCID: PMC10634703 DOI: 10.1101/2023.10.20.563334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Classical psychedelic drugs are thought to increase excitability of pyramidal cells in prefrontal cortex via activation of serotonin 2A receptors (5-HT2ARs). Here, we instead find that multiple classes of psychedelics dose-dependently suppress intrinsic excitability of pyramidal neurons, and that extracellular delivery of psychedelics decreases excitability significantly more than intracellular delivery. A previously unknown mechanism underlies this psychedelic drug action: enhancement of ubiquitously expressed potassium "M-current" channels that is independent of 5-HT2R activation. Using machine-learning-based data assimilation models, we show that M-current activation interacts with previously described mechanisms to dramatically reduce intrinsic excitability and shorten working memory timespan. Thus, psychedelic drugs suppress intrinsic excitability by modulating ion channels that are expressed throughout the brain, potentially triggering homeostatic adjustments that can contribute to widespread therapeutic benefits.
Collapse
Affiliation(s)
- Tyler G Ekins
- Dept. of Psychology, University of Michigan, Ann Arbor, MI 48109
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI 48109
| | - Isla Brooks
- Dept. of Psychology, University of Michigan, Ann Arbor, MI 48109
| | - Sameer Kailasa
- Dept. of Mathematics, University of Michigan, Ann Arbor, MI 48109
| | - Chloe Rybicki-Kler
- Dept. of Psychology, University of Michigan, Ann Arbor, MI 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109
| | | | - Ethan Donoho
- Dept. of Psychology, University of Michigan, Ann Arbor, MI 48109
| | - George A. Mashour
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI 48109
| | - Jason Rech
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109
| | - Omar J Ahmed
- Dept. of Psychology, University of Michigan, Ann Arbor, MI 48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI 48109
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
10
|
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: 0] [Impact Index Per Article: 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
|
11
|
Sanderson JL, Freund RK, Gorski JA, Dell'Acqua ML. β-Amyloid disruption of LTP/LTD balance is mediated by AKAP150-anchored PKA and Calcineurin regulation of Ca 2+-permeable AMPA receptors. Cell Rep 2021; 37:109786. [PMID: 34610314 PMCID: PMC8530450 DOI: 10.1016/j.celrep.2021.109786] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/02/2021] [Accepted: 09/10/2021] [Indexed: 01/28/2023] Open
Abstract
Regulated insertion and removal of postsynaptic AMPA glutamate receptors (AMPARs) mediates hippocampal long-term potentiation (LTP) and long-term depression (LTD) synaptic plasticity underlying learning and memory. In Alzheimer’s disease β-amyloid (Aβ) oligomers may impair learning and memory by altering AMPAR trafficking and LTP/LTD balance. Importantly, Ca2+-permeable AMPARs (CP-AMPARs) assembled from GluA1 subunits are excluded from hippocampal synapses basally but can be recruited rapidly during LTP and LTD to modify synaptic strength and signaling. By employing mouse knockin mutations that disrupt anchoring of the kinase PKA or phosphatase Calcineurin (CaN) to the postsynaptic scaffold protein AKAP150, we find that local AKAP-PKA signaling is required for CP-AMPAR recruitment, which can facilitate LTP but also, paradoxically, prime synapses for Aβ impairment of LTP mediated by local AKAP-CaN LTD signaling that promotes subsequent CP-AMPAR removal. These findings highlight the importance of PKA/CaN signaling balance and CP-AMPARs in normal plasticity and aberrant plasticity linked to disease. In Alzheimer’s disease, Aβ oligomers disrupt hippocampal neuronal plasticity and cognition. Sanderson et al. show how the postsynaptic scaffold protein AKAP150 coordinates PKA and Calcineurin regulation of Ca2+-permeable AMPA-type glutamate receptors to mediate disruption of synaptic plasticity by Aβ oligomers.
Collapse
Affiliation(s)
- Jennifer L Sanderson
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ronald K Freund
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jessica A Gorski
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA; University of Colorado Alzheimer's and Cognition Center, Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
12
|
Pampaloni NP, Riva I, Carbone AL, Plested AJR. Slow AMPA receptors in hippocampal principal cells. Cell Rep 2021; 36:109496. [PMID: 34348150 PMCID: PMC8356020 DOI: 10.1016/j.celrep.2021.109496] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 06/01/2021] [Accepted: 07/14/2021] [Indexed: 01/22/2023] Open
Abstract
Glutamate receptor ion channels, including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, mediate fast excitatory neurotransmission in the CNS. Previous work suggested that AMPA receptors produce a synaptic current with a millisecond duration. However, we find that about two-thirds of principal cells in the hippocampal CA1 region also express AMPA receptors with reduced desensitization that can stay active for half a second after repetitive stimuli. These slow AMPA receptors are expressed at about half of the synapses, with a flat spatial distribution. The increased charge transfer from slow AMPA receptors allows short-term potentiation from a postsynaptic locus and reliable triggering of action potentials. Biophysical and pharmacological observations imply slow AMPA receptors incorporate auxiliary proteins, and their activation lengthens miniature synaptic currents. These data indicate that AMPA receptors are a major source of synaptic diversity. Synapses harboring slow AMPA receptors could have unique roles in hippocampal function.
Collapse
Affiliation(s)
- Niccolò P Pampaloni
- Molecular Neuroscience and Biophysics, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany; Institute of Biology, Cellular Biophysics, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| | - Irene Riva
- Molecular Neuroscience and Biophysics, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany; Institute of Biology, Cellular Biophysics, Humboldt Universität zu Berlin, 10115 Berlin, Germany
| | - Anna L Carbone
- Molecular Neuroscience and Biophysics, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Andrew J R Plested
- Molecular Neuroscience and Biophysics, Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany; Institute of Biology, Cellular Biophysics, Humboldt Universität zu Berlin, 10115 Berlin, Germany.
| |
Collapse
|
13
|
Bissen D, Kracht MK, Foss F, Hofmann J, Acker-Palmer A. EphrinB2 and GRIP1 stabilize mushroom spines during denervation-induced homeostatic plasticity. Cell Rep 2021; 34:108923. [PMID: 33789115 PMCID: PMC8028307 DOI: 10.1016/j.celrep.2021.108923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 12/20/2020] [Accepted: 03/09/2021] [Indexed: 12/03/2022] Open
Abstract
Despite decades of work, much remains elusive about molecular events at the interplay between physiological and structural changes underlying neuronal plasticity. Here, we combined repetitive live imaging and expansion microscopy in organotypic brain slice cultures to quantitatively characterize the dynamic changes of the intracellular versus surface pools of GluA2-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) across the different dendritic spine types and the shaft during hippocampal homeostatic plasticity. Mechanistically, we identify ephrinB2 and glutamate receptor interacting protein (GRIP) 1 as mediating AMPAR relocation to the mushroom spine surface following lesion-induced denervation. Moreover, stimulation with the ephrinB2 specific receptor EphB4 not only prevents the lesion-induced disappearance of mushroom spines but is also sufficient to shift AMPARs to the surface and rescue spine recovery in a GRIP1 dominant-negative background. Thus, our results unravel a crucial role for ephrinB2 during homeostatic plasticity and identify a potential pharmacological target to improve dendritic spine plasticity upon injury.
Collapse
Affiliation(s)
- Diane Bissen
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany; Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438 Frankfurt am Main, Germany
| | - Maximilian Ken Kracht
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Franziska Foss
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Jan Hofmann
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany; Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438 Frankfurt am Main, Germany; Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
14
|
Kuhlmann N, Wagner Valladolid M, Quesada-Ramírez L, Farrer MJ, Milnerwood AJ. Chronic and Acute Manipulation of Cortical Glutamate Transmission Induces Structural and Synaptic Changes in Co-cultured Striatal Neurons. Front Cell Neurosci 2021; 15:569031. [PMID: 33679324 PMCID: PMC7930618 DOI: 10.3389/fncel.2021.569031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
In contrast to the prenatal topographic development of sensory cortices, striatal circuit organization is slow and requires the functional maturation of cortical and thalamic excitatory inputs throughout the first postnatal month. While mechanisms regulating synapse development and plasticity are quite well described at excitatory synapses of glutamatergic neurons in the neocortex, comparatively little is known of how this translates to glutamate synapses onto GABAergic neurons in the striatum. Here we investigate excitatory striatal synapse plasticity in an in vitro system, where glutamate can be studied in isolation from dopamine and other neuromodulators. We examined pre-and post-synaptic structural and functional plasticity in GABAergic striatal spiny projection neurons (SPNs), co-cultured with glutamatergic cortical neurons. After synapse formation, medium-term (24 h) TTX silencing increased the density of filopodia, and modestly decreased dendritic spine density, when assayed at 21 days in vitro (DIV). Spine reductions appeared to require residual spontaneous activation of ionotropic glutamate receptors. Conversely, chronic (14 days) TTX silencing markedly reduced spine density without any observed increase in filopodia density. Time-dependent, biphasic changes to the presynaptic marker Synapsin-1 were also observed, independent of residual spontaneous activity. Acute silencing (3 h) did not affect presynaptic markers or postsynaptic structures. To induce rapid, activity-dependent plasticity in striatal neurons, a chemical NMDA receptor-dependent “long-term potentiation (LTP)” paradigm was employed. Within 30 min, this increased spine and GluA1 cluster densities, and the percentage of spines containing GluA1 clusters, without altering the presynaptic signal. The results demonstrate that the growth and pruning of dendritic protrusions is an active process, requiring glutamate receptor activity in striatal projection neurons. Furthermore, NMDA receptor activation is sufficient to drive glutamatergic structural plasticity in SPNs, in the absence of dopamine or other neuromodulators.
Collapse
Affiliation(s)
- Naila Kuhlmann
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Lucía Quesada-Ramírez
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Austen J Milnerwood
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Chen S, Wang Y, Wang X, He M, Zhang L, Dong Z. PKA-Dependent Membrane Surface Recruitment of CI-AMPARs Is Crucial for BCP-Mediated Protection Against Post-acute Ischemic Stroke Cognitive Impairment. Front Neurol 2020; 11:566067. [PMID: 33391143 PMCID: PMC7772322 DOI: 10.3389/fneur.2020.566067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/27/2020] [Indexed: 11/21/2022] Open
Abstract
Post-acute ischemic stroke cognitive impairment frequently occurs and seriously affects patients daily activities. Recruitment of GluA2-containing Ca2+-impermeable AMPA receptors (CI-AMPARs) to hippocampal synaptic membrane surfaces was shown to trigger synaptic plasticity. Currently, the effect of CI-AMPAR trafficking on acute ischemic stroke remains poorly understood. β-Caryophyllene (BCP) has been shown to ameliorate cognitive impairment. However, the mechanism has not been characterized. In this study, a 60-min temporary middle cerebral artery occlusion (MCAO) model was established to simulate the pathology of acute ischemic stroke. BCP reduced neurologic deficits, cerebral infarct volume, and pathological damage in MCAO mice and caused CI-AMPARs to translocate to synaptic membranes in the hippocampus; surface expression of CI-AMPARs was also decreased in MCAO mice. Furthermore, this study also showed that BCP treatment significantly activated the cAMP/PKA pathway, which is consistent with the synaptic membrane expression of CI-AMPARs. To better understand the underlying mechanisms, the PKA inhibitor H-89 was used to study the role of BCP in MCAO mice. Interestingly, H-89 treatment significantly disrupted the BCP-mediated facilitation of CI-AMPAR translocation to the synaptic membrane surface and substantially attenuated BCP-induced protection against acute ischemic stroke. Additionally, inhibition the cAMP/PKA pathway not only reduced BCP-induced inhibition of AMPAR-mediated excitatory postsynaptic currents in the hippocampal CA1 region but also decreased the effect of BCP-mediated protection against post-acute ischemic stroke cognitive impairment. Taken together, these data indicate that PKA-dependent synaptic membrane surface recruitment of CI-AMPARs is crucial for the neuroprotective effect of BCP against acute ischemic stroke and protection against post-acute ischemic stroke cognitive impairment.
Collapse
Affiliation(s)
- Sha Chen
- Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacology, Chongqing Medical University, Chongqing, China.,Laboratory Sciences, Department of Clinical Biochemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuchun Wang
- Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xuhui Wang
- Department of Neurosurgery, Research Institute of Surgery Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Meng He
- Laboratory Sciences, Department of Clinical Biochemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lu Zhang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi Dong
- Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Gobert D, Schohl A, Kutsarova E, Ruthazer ES. TORC1 selectively regulates synaptic maturation and input convergence in the developing visual system. Dev Neurobiol 2020; 80:332-350. [DOI: 10.1002/dneu.22782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/07/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Delphine Gobert
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Anne Schohl
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Elena Kutsarova
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| | - Edward S. Ruthazer
- Montreal Neurological Institute‐Hospital McGill University Montreal QC Canada
| |
Collapse
|
17
|
Jacobi E, Engelhardt J. Modulation of information processing by AMPA receptor auxiliary subunits. J Physiol 2020; 599:471-483. [DOI: 10.1113/jp276698] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Eric Jacobi
- Institute of Pathophysiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- Focus Program Translational Neurosciences (FTN) University Medical Center of the Johannes Gutenberg‐University Mainz Mainz Germany
| | - Jakob Engelhardt
- Institute of Pathophysiology University Medical Center of the Johannes Gutenberg University Mainz Mainz Germany
- Focus Program Translational Neurosciences (FTN) University Medical Center of the Johannes Gutenberg‐University Mainz Mainz Germany
| |
Collapse
|
18
|
Tominaga-Yoshino K, Urakubo T, Ueno Y, Kawaai K, Saito S, Tashiro T, Ogura A. Transient appearance of Ca 2+ -permeable AMPA receptors is crucial for the production of repetitive LTP-induced synaptic enhancement (RISE) in cultured hippocampal slices. Hippocampus 2020; 30:763-769. [PMID: 32320117 DOI: 10.1002/hipo.23206] [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: 10/09/2019] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023]
Abstract
We have previously shown that repetitive induction of long-term potentiation (LTP) by glutamate (100 μM, 3 min, three times at 24-hr intervals) provoked long-lasting synaptic enhancement accompanied by synaptogenesis in rat hippocampal slice cultures, a phenomenon termed RISE (repetitive LTP-induced synaptic enhancement). Here, we examined the role of Ca2+ -permeable (CP) AMPA receptors (AMPARs) in the establishment of RISE. We first found a component sensitive to the Joro-spider toxin (JSTX), a blocker of CP-AMPARs, in a field EPSP recorded from CA3-CA1 synapses at 2-3 days after stimulation, but this component was not found for 9-10 days. We also observed that rectification of AMPAR-mediated current appeared only 2-3 days after stimulation, using a whole-cell patch clamp recording from CA1 pyramidal neurons. These findings indicate that CP-AMPAR is transiently expressed in the developing phase of RISE. The blockade of CP-AMPARs by JSTX for 24 hr at this developing phase inhibited RISE establishment, accompanied by the loss of small synapses at the ultrastructural level. These results suggest that transiently induced CP-AMPARs play a critical role in synaptogenesis in the developing phase of long-lasting hippocampal synaptic plasticity, RISE.
Collapse
Affiliation(s)
- Keiko Tominaga-Yoshino
- Department of Neuroscience, Osaka University Graduate School of Frontier Biosciences, Osaka, Japan
| | - Tomoyoshi Urakubo
- Department of Neuroscience, Osaka University Graduate School of Frontier Biosciences, Osaka, Japan
| | - Yukiko Ueno
- Department of Neuroscience, Osaka University Graduate School of Frontier Biosciences, Osaka, Japan
| | - Katsuhiro Kawaai
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama-Gakuin University, Kanagawa, Japan
| | - Shinichi Saito
- Department of Neuroscience, Osaka University Graduate School of Frontier Biosciences, Osaka, Japan
| | - Tomoko Tashiro
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama-Gakuin University, Kanagawa, Japan
| | - Akihiko Ogura
- Department of Neuroscience, Osaka University Graduate School of Frontier Biosciences, Osaka, Japan
| |
Collapse
|
19
|
Purkey AM, Dell’Acqua ML. Phosphorylation-Dependent Regulation of Ca 2+-Permeable AMPA Receptors During Hippocampal Synaptic Plasticity. Front Synaptic Neurosci 2020; 12:8. [PMID: 32292336 PMCID: PMC7119613 DOI: 10.3389/fnsyn.2020.00008] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/18/2020] [Indexed: 01/28/2023] Open
Abstract
Experience-dependent learning and memory require multiple forms of plasticity at hippocampal and cortical synapses that are regulated by N-methyl-D-aspartate receptors (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type ionotropic glutamate receptors (NMDAR, AMPAR). These plasticity mechanisms include long-term potentiation (LTP) and depression (LTD), which are Hebbian input-specific mechanisms that rapidly increase or decrease AMPAR synaptic strength at specific inputs, and homeostatic plasticity that globally scales-up or -down AMPAR synaptic strength across many or even all inputs. Frequently, these changes in synaptic strength are also accompanied by a change in the subunit composition of AMPARs at the synapse due to the trafficking to and from the synapse of receptors lacking GluA2 subunits. These GluA2-lacking receptors are most often GluA1 homomeric receptors that exhibit higher single-channel conductance and are Ca2+-permeable (CP-AMPAR). This review article will focus on the role of protein phosphorylation in regulation of GluA1 CP-AMPAR recruitment and removal from hippocampal synapses during synaptic plasticity with an emphasis on the crucial role of local signaling by the cAMP-dependent protein kinase (PKA) and the Ca2+calmodulin-dependent protein phosphatase 2B/calcineurin (CaN) that is coordinated by the postsynaptic scaffold protein A-kinase anchoring protein 79/150 (AKAP79/150).
Collapse
Affiliation(s)
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
20
|
Croft CL, Futch HS, Moore BD, Golde TE. Organotypic brain slice cultures to model neurodegenerative proteinopathies. Mol Neurodegener 2019; 14:45. [PMID: 31791377 PMCID: PMC6889333 DOI: 10.1186/s13024-019-0346-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/13/2019] [Indexed: 01/30/2023] Open
Abstract
Organotypic slice cultures of brain or spinal cord have been a longstanding tool in neuroscience research but their utility for understanding Alzheimer's disease (AD) and other neurodegenerative proteinopathies has only recently begun to be evaluated. Organotypic brain slice cultures (BSCs) represent a physiologically relevant three-dimensional model of the brain. BSCs support all the central nervous system (CNS) cell types and can be produced from brain areas involved in neurodegenerative disease. BSCs can be used to better understand the induction and significance of proteinopathies underlying the development and progression of AD and other neurodegenerative disorders, and in the future may serve as bridging technologies between cell culture and in vivo experiments for the development and evaluation of novel therapeutic targets and strategies. We review the initial development and general use of BSCs in neuroscience research and highlight the advantages of these cultures as an ex vivo model. Subsequently we focus on i) BSC-based modeling of AD and other neurodegenerative proteinopathies ii) use of BSCs to understand mechanisms underlying these diseases and iii) how BSCs can serve as tools to screen for suitable therapeutics prior to in vivo investigations. Finally, we will examine i) open questions regarding the use of such cultures and ii) how emerging technologies such as recombinant adeno-associated viruses (rAAV) may be combined with these models to advance translational research relevant to neurodegenerative disorders.
Collapse
Affiliation(s)
- C L Croft
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - H S Futch
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - B D Moore
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - T E Golde
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA. .,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA. .,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
21
|
Diering GH, Huganir RL. The AMPA Receptor Code of Synaptic Plasticity. Neuron 2019; 100:314-329. [PMID: 30359599 DOI: 10.1016/j.neuron.2018.10.018] [Citation(s) in RCA: 547] [Impact Index Per Article: 91.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023]
Abstract
Changes in the properties and postsynaptic abundance of AMPA-type glutamate receptors (AMPARs) are major mechanisms underlying various forms of synaptic plasticity, including long-term potentiation (LTP), long-term depression (LTD), and homeostatic scaling. The function and the trafficking of AMPARs to and from synapses is modulated by specific AMPAR GluA1-GluA4 subunits, subunit-specific protein interactors, auxiliary subunits, and posttranslational modifications. Layers of regulation are added to AMPAR tetramers through these different interactions and modifications, increasing the computational power of synapses. Here we review the reliance of synaptic plasticity on AMPAR variants and propose "the AMPAR code" as a conceptual framework. The AMPAR code suggests that AMPAR variants will be predictive of the types and extent of synaptic plasticity that can occur and that a hierarchy exists such that certain AMPARs will be disproportionally recruited to synapses during LTP/homeostatic scaling up, or removed during LTD/homeostatic scaling down.
Collapse
Affiliation(s)
- Graham H Diering
- Department of Cell Biology and Physiology, and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Richard L Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
22
|
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
|
23
|
MicroRNA-186-5p controls GluA2 surface expression and synaptic scaling in hippocampal neurons. Proc Natl Acad Sci U S A 2019; 116:5727-5736. [PMID: 30808806 DOI: 10.1073/pnas.1900338116] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Homeostatic synaptic scaling is a negative feedback response to fluctuations in synaptic strength induced by developmental or learning-related processes, which maintains neuronal activity stable. Although several components of the synaptic scaling apparatus have been characterized, the intrinsic regulatory mechanisms promoting scaling remain largely unknown. MicroRNAs may contribute to posttranscriptional control of mRNAs implicated in different stages of synaptic scaling, but their role in these mechanisms is still undervalued. Here, we report that chronic blockade of glutamate receptors of the AMPA and NMDA types in hippocampal neurons in culture induces changes in the neuronal mRNA and miRNA transcriptomes, leading to synaptic upscaling. Specifically, we show that synaptic activity blockade persistently down-regulates miR-186-5p. Moreover, we describe a conserved miR-186-5p-binding site within the 3'UTR of the mRNA encoding the AMPA receptor GluA2 subunit, and demonstrate that GluA2 is a direct target of miR-186-5p. Overexpression of miR-186 decreased GluA2 surface levels, increased synaptic expression of GluA2-lacking AMPA receptors, and blocked synaptic scaling, whereas inhibition of miR-186-5p increased GluA2 surface levels and the amplitude and frequency of AMPA receptor-mediated currents, and mimicked excitatory synaptic scaling induced by synaptic inactivity. Our findings elucidate an activity-dependent miRNA-mediated mechanism for regulation of AMPA receptor expression.
Collapse
|
24
|
Park P, Kang H, Sanderson TM, Bortolotto ZA, Georgiou J, Zhuo M, Kaang BK, Collingridge GL. The Role of Calcium-Permeable AMPARs in Long-Term Potentiation at Principal Neurons in the Rodent Hippocampus. Front Synaptic Neurosci 2018; 10:42. [PMID: 30524263 PMCID: PMC6262052 DOI: 10.3389/fnsyn.2018.00042] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/05/2018] [Indexed: 11/25/2022] Open
Abstract
Long-term potentiation (LTP) at hippocampal CA1 synapses is classically triggered by the synaptic activation of NMDA receptors (NMDARs). More recently, it has been shown that calcium-permeable (CP) AMPA receptors (AMPARs) can also trigger synaptic plasticity at these synapses. Here, we review this literature with a focus on recent evidence that CP-AMPARs are critical for the induction of the protein kinase A (PKA)- and protein synthesis-dependent component of LTP.
Collapse
Affiliation(s)
- Pojeong Park
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Heather Kang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Thomas M Sanderson
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Zuner A Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - John Georgiou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Min Zhuo
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Bong-Kiun Kaang
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Graham L Collingridge
- Department of Biological Sciences and Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
25
|
Purkey AM, Woolfrey KM, Crosby KC, Stich DG, Chick WS, Aoto J, Dell'Acqua ML. AKAP150 Palmitoylation Regulates Synaptic Incorporation of Ca 2+-Permeable AMPA Receptors to Control LTP. Cell Rep 2018; 25:974-987.e4. [PMID: 30355502 PMCID: PMC6263960 DOI: 10.1016/j.celrep.2018.09.085] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/06/2018] [Accepted: 09/25/2018] [Indexed: 11/22/2022] Open
Abstract
Ca2+-permeable AMPA-type glutamate receptors (CP-AMPARs) containing GluA1 but lacking GluA2 subunits contribute to multiple forms of synaptic plasticity, including long-term potentiation (LTP), but mechanisms regulating CP-AMPARs are poorly understood. A-kinase anchoring protein (AKAP) 150 scaffolds kinases and phosphatases to regulate GluA1 phosphorylation and trafficking, and trafficking of AKAP150 itself is modulated by palmitoylation on two Cys residues. Here, we developed a palmitoylation-deficient knockin mouse to show that AKAP150 palmitoylation regulates CP-AMPAR incorporation at hippocampal synapses. Using biochemical, super-resolution imaging, and electrophysiological approaches, we found that palmitoylation promotes AKAP150 localization to recycling endosomes and the postsynaptic density (PSD) to limit CP-AMPAR basal synaptic incorporation. In addition, we found that AKAP150 palmitoylation is required for LTP induced by weaker stimulation that recruits CP-AMPARs to synapses but not stronger stimulation that recruits GluA2-containing AMPARs. Thus, AKAP150 palmitoylation controls its subcellular localization to maintain proper basal and activity-dependent regulation of synaptic AMPAR subunit composition.
Collapse
Affiliation(s)
- Alicia M Purkey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kevin M Woolfrey
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kevin C Crosby
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Dominik G Stich
- Advanced Light Microscopy Core, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Wallace S Chick
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jason Aoto
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Advanced Light Microscopy Core, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
26
|
Patriarchi T, Buonarati OR, Hell JW. Postsynaptic localization and regulation of AMPA receptors and Cav1.2 by β2 adrenergic receptor/PKA and Ca 2+/CaMKII signaling. EMBO J 2018; 37:e99771. [PMID: 30249603 PMCID: PMC6187224 DOI: 10.15252/embj.201899771] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/25/2018] [Accepted: 08/17/2018] [Indexed: 11/09/2022] Open
Abstract
The synapse transmits, processes, and stores data within its tiny space. Effective and specific signaling requires precise alignment of the relevant components. This review examines current insights into mechanisms of AMPAR and NMDAR localization by PSD-95 and their spatial distribution at postsynaptic sites to illuminate the structural and functional framework of postsynaptic signaling. It subsequently delineates how β2 adrenergic receptor (β2 AR) signaling via adenylyl cyclase and the cAMP-dependent protein kinase PKA is organized within nanodomains. Here, we discuss targeting of β2 AR, adenylyl cyclase, and PKA to defined signaling complexes at postsynaptic sites, i.e., AMPARs and the L-type Ca2+ channel Cav1.2, and other subcellular surface localizations, the role of A kinase anchor proteins, the physiological relevance of the spatial restriction of corresponding signaling, and their interplay with signal transduction by the Ca2+- and calmodulin-dependent kinase CaMKII How localized and specific signaling by cAMP occurs is a central cellular question. The dendritic spine constitutes an ideal paradigm for elucidating the dimensions of spatially restricted signaling because of their small size and defined protein composition.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling/physiology
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cyclic AMP-Dependent Protein Kinases/genetics
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Humans
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Synapses/genetics
- Synapses/metabolism
Collapse
Affiliation(s)
- Tommaso Patriarchi
- Department of Pharmacology, University of California, Davis, CA, USA
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | | | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
27
|
Rozov A, Zakharova Y, Vazetdinova A, Valiullina-Rakhmatullina F. The Role of Polyamine-Dependent Facilitation of Calcium Permeable AMPARs in Short-Term Synaptic Enhancement. Front Cell Neurosci 2018; 12:345. [PMID: 30364146 PMCID: PMC6191566 DOI: 10.3389/fncel.2018.00345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/18/2018] [Indexed: 11/13/2022] Open
Abstract
Depending on subunit composition AMPA receptor channels can be subdivided into two groups: GluA2-containing calcium impermeable AMPARs, and GluA2-lacking calcium permeable, AMPARs. These two groups differ in a number of biophysical properties and, most likely, in their functional role at glutamatergic synapses. GluA2-lacking channels have received a lot of attention over the last two decades mainly due to high calcium permeability, which was suggested to play a significant role in the induction of long-term synaptic plasticity in healthy tissue and neuronal death under neuropathological conditions. However, calcium permeable AMPARs possess another property that can contribute substantially to frequency dependent dynamics of synaptic efficacy. In the closed state calcium permeable AMPARs are blocked by endogenous polyamines, however, repetitive activation leads to progressive relief from the block and to the facilitation of ion flux through these channels. Polyamine-dependent facilitation of AMPARs can contribute to short-term plasticity at synapses that have high initial release probability and express calcium permeable AMPARs. During synaptic transmission activity-dependent relief from polyamine block of postsynaptic calcium-permeable AMPARs either counteracts presynaptic short-term depression in a frequency-dependent manner or, under specific stimulation conditions, induces facilitation of a synaptic response. Taking into account the fact that expression of calcium permeable AMPARs is developmentally regulated, depends on network activity and increases in diseased brain states, polyamine-dependent facilitation of calcium permeable AMPARs is an important, entirely postsynaptic mechanism of synaptic gain regulation.
Collapse
Affiliation(s)
- Andrei Rozov
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Yulia Zakharova
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Alina Vazetdinova
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | | |
Collapse
|
28
|
Haselmann H, Mannara F, Werner C, Planagumà J, Miguez-Cabello F, Schmidl L, Grünewald B, Petit-Pedrol M, Kirmse K, Classen J, Demir F, Klöcker N, Soto D, Doose S, Dalmau J, Hallermann S, Geis C. Human Autoantibodies against the AMPA Receptor Subunit GluA2 Induce Receptor Reorganization and Memory Dysfunction. Neuron 2018; 100:91-105.e9. [PMID: 30146304 DOI: 10.1016/j.neuron.2018.07.048] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 03/14/2018] [Accepted: 07/27/2018] [Indexed: 11/29/2022]
Abstract
AMPA receptors are essential for fast excitatory transmission in the CNS. Autoantibodies to AMPA receptors have been identified in humans with autoimmune encephalitis and severe defects of hippocampal function. Here, combining electrophysiology and high-resolution imaging with neuronal culture preparations and passive-transfer models in wild-type and GluA1-knockout mice, we analyze how specific human autoantibodies against the AMPA receptor subunit GluA2 affect receptor function and composition, synaptic transmission, and plasticity. Anti-GluA2 antibodies induce receptor internalization and a reduction of synaptic GluA2-containing AMPARs followed by compensatory ryanodine receptor-dependent incorporation of synaptic non-GluA2 AMPARs. Furthermore, application of human pathogenic anti-GluA2 antibodies to mice impairs long-term synaptic plasticity in vitro and affects learning and memory in vivo. Our results identify a specific immune-neuronal rearrangement of AMPA receptor subunits, providing a framework to explain disease symptoms.
Collapse
Affiliation(s)
- Holger Haselmann
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Francesco Mannara
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Christian Werner
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Jesús Planagumà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Federico Miguez-Cabello
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Lars Schmidl
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Benedikt Grünewald
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Mar Petit-Pedrol
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Knut Kirmse
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Fatih Demir
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany; Forschungszentrum Jülich, Central Institute for Engineering, Electronics and Analytics (ZEA-3), Wilhelm-Johnen-Strasse, 52425 Jülich, Germany
| | - Nikolaj Klöcker
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - David Soto
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sören Doose
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Josep Dalmau
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys, 23, 08010 Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Raras (CIBERER), (Instituto Carlos III, Madrid), Av. Monforte de Lemos, 3-5 Pabellón 11, 28029 Madrid, Spain
| | - Stefan Hallermann
- Carl-Ludwig-Institute for Physiology, Medical Faculty, University of Leipzig, Liebigstrasse 27, 04103 Leipzig, Germany
| | - Christian Geis
- Hans-Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| |
Collapse
|
29
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|
30
|
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
|
31
|
Bhattacharya S, Khatri A, Swanger SA, DiRaddo JO, Yi F, Hansen KB, Yuan H, Traynelis SF. Triheteromeric GluN1/GluN2A/GluN2C NMDARs with Unique Single-Channel Properties Are the Dominant Receptor Population in Cerebellar Granule Cells. Neuron 2018; 99:315-328.e5. [PMID: 30056832 DOI: 10.1016/j.neuron.2018.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/04/2018] [Accepted: 06/05/2018] [Indexed: 01/12/2023]
Abstract
NMDA-type glutamate receptors (NMDARs) are ligand-gated ion channels that mediate excitatory neurotransmission in the CNS. Here we describe functional and single-channel properties of triheteromeric GluN1/GluN2A/GluN2C receptors, which contain two GluN1, one GluN2A, and one GluN2C subunits. This NMDAR has three conductance levels and opens in bursts similar to GluN1/GluN2A receptors but with a single-channel open time and open probability reminiscent of GluN1/GluN2C receptors. The deactivation time course of GluN1/GluN2A/GluN2C receptors is intermediate to GluN1/GluN2A and GluN1/GluN2C receptors and is not dominated by GluN2A or GluN2C. We show that triheteromeric GluN1/GluN2A/GluN2C receptors are the predominant NMDARs in cerebellar granule cells and propose that co-expression of GluN2A and GluN2C in cerebellar granule cells occludes cell surface expression of diheteromeric GluN1/GluN2C receptors. This new insight into neuronal GluN1/GluN2A/GluN2C receptors highlights the complexity of NMDAR signaling in the CNS.
Collapse
Affiliation(s)
| | - Alpa Khatri
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sharon A Swanger
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John O DiRaddo
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Feng Yi
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Hongjie Yuan
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Control of Homeostatic Synaptic Plasticity by AKAP-Anchored Kinase and Phosphatase Regulation of Ca 2+-Permeable AMPA Receptors. J Neurosci 2018; 38:2863-2876. [PMID: 29440558 DOI: 10.1523/jneurosci.2362-17.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/17/2018] [Accepted: 02/06/2018] [Indexed: 12/31/2022] Open
Abstract
Neuronal information processing requires multiple forms of synaptic plasticity mediated by NMDARs and AMPA-type glutamate receptors (AMPARs). These plasticity mechanisms include long-term potentiation (LTP) and long-term depression (LTD), which are Hebbian, homosynaptic mechanisms locally regulating synaptic strength of specific inputs, and homeostatic synaptic scaling, which is a heterosynaptic mechanism globally regulating synaptic strength across all inputs. In many cases, LTP and homeostatic scaling regulate AMPAR subunit composition to increase synaptic strength via incorporation of Ca2+-permeable receptors (CP-AMPAR) containing GluA1, but lacking GluA2, subunits. Previous work by our group and others demonstrated that anchoring of the kinase PKA and the phosphatase calcineurin (CaN) to A-kinase anchoring protein (AKAP) 150 play opposing roles in regulation of GluA1 Ser845 phosphorylation and CP-AMPAR synaptic incorporation during hippocampal LTP and LTD. Here, using both male and female knock-in mice that are deficient in PKA or CaN anchoring, we show that AKAP150-anchored PKA and CaN also play novel roles in controlling CP-AMPAR synaptic incorporation during homeostatic plasticity in hippocampal neurons. We found that genetic disruption of AKAP-PKA anchoring prevented increases in Ser845 phosphorylation and CP-AMPAR synaptic recruitment during rapid homeostatic synaptic scaling-up induced by combined blockade of action potential firing and NMDAR activity. In contrast, genetic disruption of AKAP-CaN anchoring resulted in basal increases in Ser845 phosphorylation and CP-AMPAR synaptic activity that blocked subsequent scaling-up by preventing additional CP-AMPAR recruitment. Thus, the balanced, opposing phospho-regulation provided by AKAP-anchored PKA and CaN is essential for control of both Hebbian and homeostatic plasticity mechanisms that require CP-AMPARs.SIGNIFICANCE STATEMENT Neuronal circuit function is shaped by multiple forms of activity-dependent plasticity that control excitatory synaptic strength, including LTP/LTD that adjusts strength of individual synapses and homeostatic plasticity that adjusts overall strength of all synapses. Mechanisms controlling LTP/LTD and homeostatic plasticity were originally thought to be distinct; however, recent studies suggest that CP-AMPAR phosphorylation regulation is important during both LTP/LTD and homeostatic plasticity. Here we show that CP-AMPAR regulation by the kinase PKA and phosphatase CaN coanchored to the scaffold protein AKAP150, a mechanism previously implicated in LTP/LTD, is also crucial for controlling synaptic strength during homeostatic plasticity. These novel findings significantly expand our understanding of homeostatic plasticity mechanisms and further emphasize how intertwined they are with LTP and LTD.
Collapse
|
35
|
Synaptic homeostasis requires the membrane-proximal carboxy tail of GluA2. Proc Natl Acad Sci U S A 2017; 114:13266-13271. [PMID: 29180434 DOI: 10.1073/pnas.1716022114] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bidirectional scaling of synaptic transmission, expressed as a compensatory change in quantal size following chronic activity perturbation, is a critical effector mechanism underlying homeostatic plasticity in the brain. An emerging model posits that the GluA2 AMPA receptor (AMPAR) subunit may be important for the bidirectional scaling of excitatory transmission; however, whether this subunit plays an obligatory role in synaptic scaling, and the identity of the precise domain(s) involved, remain controversial. We set out to determine the specific AMPAR subunit required for scaling up in CA1 hippocampal pyramidal neurons, and found that the GluA2 subunit is both necessary and sufficient. In addition, our results point to a critical role for a single amino acid within the membrane-proximal region of the GluA2 cytoplasmic tail, and suggest a distinct model for the regulation of AMPAR trafficking in synaptic homeostasis.
Collapse
|
36
|
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
|
37
|
Wall MJ, Corrêa SAL. The mechanistic link between Arc/Arg3.1 expression and AMPA receptor endocytosis. Semin Cell Dev Biol 2017; 77:17-24. [PMID: 28890421 DOI: 10.1016/j.semcdb.2017.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 02/06/2023]
Abstract
The activity-regulated cytoskeleton associated protein (Arc/Arg3.1) plays a key role in determining synaptic strength through facilitation of AMPA receptor (AMPAR) endocytosis. Although there is considerable data on the mechanism by which Arc induction controls synaptic plasticity and learning behaviours, several key mechanistic questions remain. Here we review data on the link between Arc expression and the clathrin-mediated endocytic pathway which internalises AMPARs and discuss the significance of Arc binding to the clathrin adaptor protein 2 (AP-2) and to endophilin/dynamin. We consider which AMPAR subunits are selected for Arc-mediated internalisation, implications for synaptic function and consider Arc as a therapeutic target.
Collapse
Affiliation(s)
- Mark J Wall
- School of Life Sciences, University of Warwick, United Kingdom.
| | - Sonia A L Corrêa
- School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, United Kingdom.
| |
Collapse
|
38
|
Xu X, Pozzo-Miller L. EEA1 restores homeostatic synaptic plasticity in hippocampal neurons from Rett syndrome mice. J Physiol 2017. [PMID: 28621434 DOI: 10.1113/jp274450] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in MECP2, the gene encoding the transcriptional regulator methyl-CpG-binding protein 2 (MeCP2). Mecp2 deletion in mice results in an imbalance of excitation and inhibition in hippocampal neurons, which affects 'Hebbian' synaptic plasticity. We show that Mecp2-deficient neurons also lack homeostatic synaptic plasticity, likely due to reduced levels of EEA1, a protein involved in AMPA receptor endocytosis. Expression of EEA1 restored homeostatic synaptic plasticity in Mecp2-deficient neurons, providing novel targets of intervention in Rett syndrome. ABSTRACT Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in MECP2, the gene encoding the transcriptional regulator methyl-CpG-binding protein 2 (MeCP2). Deletion of Mecp2 in mice results in an imbalance of synaptic excitation and inhibition in hippocampal pyramidal neurons, which affects 'Hebbian' long-term synaptic plasticity. Since the excitatory-inhibitory balance is maintained by homeostatic mechanisms, we examined the role of MeCP2 in homeostatic synaptic plasticity (HSP) at excitatory synapses. Negative feedback HSP, also known as synaptic scaling, maintains the global synaptic strength of individual neurons in response to sustained alterations in neuronal activity. Hippocampal neurons from Mecp2 knockout (KO) mice do not show the characteristic homeostatic scaling up of the amplitude of miniature excitatory postsynaptic currents (mEPSCs) and of synaptic levels of the GluA1 subunit of AMPA-type glutamate receptors after 48 h silencing with the Na+ channel blocker tetrodotoxin. This deficit in HSP is bidirectional because Mecp2 KO neurons also failed to scale down mEPSC amplitudes and GluA1 synaptic levels after 48 h blockade of type A GABA receptor (GABAA R)-mediated inhibition with bicuculline. Consistent with the role of synaptic trafficking of AMPA-type of glutamate receptors in HSP, Mecp2 KO neurons have lower levels of early endosome antigen 1 (EEA1), a protein involved in AMPA-type glutamate receptor endocytosis. In addition, expression of EEA1 in Mecp2 KO neurons reduced mEPSC amplitudes to wild-type levels, and restored synaptic scaling down of mEPSC amplitudes after 48 h blockade of GABAA R-mediated inhibition with bicuculline. The identification of a molecular deficit in HSP in Mecp2 KO neurons provides potentially novel targets of intervention for improving hippocampal function in Rett syndrome individuals.
Collapse
Affiliation(s)
- Xin Xu
- Department of Neurobiology, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
39
|
Stolyarova A, Izquierdo A. Complementary contributions of basolateral amygdala and orbitofrontal cortex to value learning under uncertainty. eLife 2017; 6. [PMID: 28682238 PMCID: PMC5533586 DOI: 10.7554/elife.27483] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/05/2017] [Indexed: 11/24/2022] Open
Abstract
We make choices based on the values of expected outcomes, informed by previous experience in similar settings. When the outcomes of our decisions consistently violate expectations, new learning is needed to maximize rewards. Yet not every surprising event indicates a meaningful change in the environment. Even when conditions are stable overall, outcomes of a single experience can still be unpredictable due to small fluctuations (i.e., expected uncertainty) in reward or costs. In the present work, we investigate causal contributions of the basolateral amygdala (BLA) and orbitofrontal cortex (OFC) in rats to learning under expected outcome uncertainty in a novel delay-based task that incorporates both predictable fluctuations and directional shifts in outcome values. We demonstrate that OFC is required to accurately represent the distribution of wait times to stabilize choice preferences despite trial-by-trial fluctuations in outcomes, whereas BLA is necessary for the facilitation of learning in response to surprising events. DOI:http://dx.doi.org/10.7554/eLife.27483.001 Nobody likes waiting – we opt for online shopping to avoid standing in lines, grow impatient in traffic, and often prefer restaurants that serve food quickly. When making decisions, humans and other animals try to maximize the benefits by weighing up the costs and rewards associated with a situation. Many regions in the brain help us choose the best options based on quality and size of rewards, and required waiting times. Even before we make decisions, the activity in these brain regions predicts what we will choose. Sometimes, however, unexpected changes can lead to longer waiting times and our preferences suddenly become less desirable. The brain can detect such changes by comparing the outcomes we anticipate to those we experience. When the outcomes are surprising, specific areas in the brain such as the amygdala and the orbitofrontal cortex help us learn to make better choices. However, as surprising events can occur purely by chance, we need to be able to ignore irrelevant surprises and only learn from meaningful ones. Until now, it was not clear whether the amygdala and orbitofrontal cortex play specific roles in successfully learning under such conditions. Stolyarova and Izquierdo trained rats to select between two images and rewarded them with sugar pellets after different delays. If rats chose one of these images they received the rewards after a predictable delay that was about 10 seconds, while choosing the other one produced variable delays – sometimes the time intervals were either very short or very long. Then, the waiting times for one of the alternatives changed unexpectedly. Rats with healthy brains quickly learned to choose the option with the shorter waiting time. Stolyarova and Izquierdo repeated the experiments with rats that had damage in a part of the amygdala. These rats learned more slowly, particularly when the variable option changed for the better. Rats with damage to the orbitofrontal cortex failed to learn at all. Stolyarova and Izquierdo then examined the rats’ behavior during delays. Rats with damage to the orbitofrontal cortex could not distinguish between meaningful and irrelevant surprises and always looked for the food pellet (i.e. anticipated a reward) at the average delay interval. These findings highlight two brain regions that help us distinguish meaningful surprises from irrelevant ones. A next step will be to examine how the amygdala and orbitofrontal cortex interact during learning and see if changes to the activity of these brain regions may affect responses. Advanced methods to non-invasively manipulate brain activity in humans may help people who find it hard to cope with changes; or individuals suffering from substance use disorders, who often struggle to give up drugs that provide them immediate and predictable rewards. DOI:http://dx.doi.org/10.7554/eLife.27483.002
Collapse
Affiliation(s)
- Alexandra Stolyarova
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States
| | - Alicia Izquierdo
- Department of Psychology, University of California, Los Angeles, Los Angeles, United States.,Integrative Center for Learning and Memory, University of California, Los Angeles, Los Angeles, United States.,Integrative Center for Addictions, University of California, Los Angeles, Los Angeles, United States.,The Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
40
|
Christian DT, Wang X, Chen EL, Sehgal LK, Ghassemlou MN, Miao JJ, Estepanian D, Araghi CH, Stutzmann GE, Wolf ME. Dynamic Alterations of Rat Nucleus Accumbens Dendritic Spines over 2 Months of Abstinence from Extended-Access Cocaine Self-Administration. Neuropsychopharmacology 2017; 42:748-756. [PMID: 27555380 PMCID: PMC5240181 DOI: 10.1038/npp.2016.168] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/25/2016] [Accepted: 08/16/2016] [Indexed: 11/09/2022]
Abstract
Chronic cocaine exposure influences the density and morphology of dendritic spines on medium spiny neurons (MSNs) in the nucleus accumbens (NAc), a critical brain region for cocaine craving. However, the relationship between spine plasticity and craving remains unclear. To study this relationship, we trained rats to self-administer cocaine using an extended-access regimen (6 h per day, 10 days); controls self-administered saline. Previously, a time-dependent intensification (incubation) of cue-induced cocaine craving has been demonstrated after withdrawal from this regimen; furthermore, Ca2+-permeable AMPA receptors (CP-AMPARs) increase in the NAc core after ~1 month of withdrawal and thereafter mediate the expression of incubated craving. Although neither craving nor CP-AMPAR levels were measured in the present study, we killed rats at four withdrawal day (WD) time-points (WD14, WD25, WD36, or WD60) selected to span the rising phase of incubation and the transition from low to high CP-AMPAR levels. MSNs were iontophoretically filled with Lucifer yellow and spines were analyzed with NeuronStudio software. Compared with saline controls, cocaine rats showed no changes in spine density or morphology in the NAc core on WD14 or WD25. On WD36, approximately the withdrawal time when stable elevation of CP-AMPAR levels is detected, the cocaine group exhibited increased density of thin spines in the NAc core. By WD60, however, this effect had reversed: the density of thin spines was lower in cocaine rats compared with saline rats. In contrast, craving and CP-AMPAR levels remain high on WD60. We also assessed spine density on WD36 in the dorsolateral striatum, a region that is not implicated in incubation of cocaine craving and does not undergo CP-AMPAR plasticity. Here, the cocaine group exhibited a small leftward shift in the distribution of spine densities plotted as a cumulative distribution, opposite to the effect found in the NAc core. Overall, our results demonstrate changes in NAc core spines over 2 months of withdrawal but no simple relationship between the time dependency of these spine changes and the previously demonstrated time course of incubation of cocaine craving. However, they raise the possibility that CP-AMPAR accumulation in the NAc core occurs in a population of thin spines that emerges after ~1 month of withdrawal.
Collapse
Affiliation(s)
- Daniel T Christian
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Xiaoting Wang
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Eugenia L Chen
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Lakshya K Sehgal
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Michael N Ghassemlou
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Julia J Miao
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Derenik Estepanian
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Cameron H Araghi
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Grace E Stutzmann
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA,Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA, Tel: +1 847 578 8659, Fax: +1 847 578 8515, E-mail:
| |
Collapse
|
41
|
Gillary G, Niebur E. The Edge of Stability: Response Times and Delta Oscillations in Balanced Networks. PLoS Comput Biol 2016; 12:e1005121. [PMID: 27689361 PMCID: PMC5045166 DOI: 10.1371/journal.pcbi.1005121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/26/2016] [Indexed: 11/27/2022] Open
Abstract
The standard architecture of neocortex is a network with excitation and inhibition in closely maintained balance. These networks respond fast and with high precision to their inputs and they allow selective amplification of patterned signals. The stability of such networks is known to depend on balancing the strengths of positive and negative feedback. We here show that a second condition is required for stability which depends on the relative strengths and time courses of fast (AMPA) and slow (NMDA) currents in the excitatory projections. This condition also determines the response time of the network. We show that networks which respond quickly to an input are necessarily close to an oscillatory instability which resonates in the delta range. This instability explains the existence of neocortical delta oscillations and the emergence of absence epilepsy. Although cortical delta oscillations are a network-level phenomenon, we show that in non-pathological networks, individual neurons receive sufficient information to keep the network in the fast-response regime without sliding into the instability. Many networks in the brain are finely balanced, with equal contributions from excitation and inhibition. Deviations from this balance, if for instance the total amount of excitation exceeds that of inhibition, lead to potentially devastating instabilities. Unlike previous work we consider the interaction between fast and slow excitatory connections. We show that not only the amount of excitation needs to be controlled to achieve network stability but also the ratio of slow to fast excitation. Furthermore, optimally fast network performance requires that networks approach instability. However, networks very close to this instability develop oscillations in the delta range (1–4Hz) which potentially cause absence epilepsy. We show that a normal (non-pathological) network can auto-regulate its activity to avoid the instability.
Collapse
Affiliation(s)
- Grant Gillary
- Zanvyl Krieger Mind/Brain Institute, Baltimore, Maryland, United States of America
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Ernst Niebur
- Zanvyl Krieger Mind/Brain Institute, Baltimore, Maryland, United States of America
- Solomon Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
42
|
Sanderson JL, Gorski JA, Dell'Acqua ML. NMDA Receptor-Dependent LTD Requires Transient Synaptic Incorporation of Ca²⁺-Permeable AMPARs Mediated by AKAP150-Anchored PKA and Calcineurin. Neuron 2016; 89:1000-15. [PMID: 26938443 DOI: 10.1016/j.neuron.2016.01.043] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/15/2015] [Accepted: 01/14/2016] [Indexed: 11/17/2022]
Abstract
Information processing in the brain requires multiple forms of synaptic plasticity that converge on regulation of NMDA and AMPA-type glutamate receptors (NMDAR, AMPAR), including long-term potentiation (LTP) and long-term depression (LTD) and homeostatic scaling. In some cases, LTP and homeostatic plasticity regulate synaptic AMPAR subunit composition to increase the contribution of Ca(2+)-permeable receptors (CP-AMPARs) containing GluA1 but lacking GluA2 subunits. Here, we show that PKA anchored to the scaffold protein AKAP150 regulates GluA1 phosphorylation and plays a novel role controlling CP-AMPAR synaptic incorporation during NMDAR-dependent LTD. Using knockin mice that are deficient in AKAP-anchoring of either PKA or the opposing phosphatase calcineurin, we found that CP-AMPARs are recruited to hippocampal synapses by anchored PKA during LTD induction but are then rapidly removed by anchored calcineurin. Importantly, blocking CP-AMPAR recruitment, removal, or activity interferes with LTD. Thus, CP-AMPAR synaptic recruitment is required to transiently augment NMDAR Ca(2+) signaling during LTD induction.
Collapse
Affiliation(s)
- Jennifer L Sanderson
- Department of Pharmacology, University of Colorado School of Medicine, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Jessica A Gorski
- Department of Pharmacology, University of Colorado School of Medicine, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, 12800 East 19th Avenue, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado School of Medicine, 12800 East 19th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
43
|
Seo MK, McIntyre RS, Cho HY, Lee CH, Park SW, Mansur RB, Kim GM, Baek JH, Woo YS, Lee JG, Kim YH. Tianeptine induces mTORC1 activation in rat hippocampal neurons under toxic conditions. Psychopharmacology (Berl) 2016; 233:2617-27. [PMID: 27129862 DOI: 10.1007/s00213-016-4309-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/22/2016] [Indexed: 12/23/2022]
Abstract
RATIONALE Recent studies have demonstrated that mTORC1 activation may be related to antidepressant action. However, the relationship between mTORC1 signaling activation and currently prescribed antidepressants remains unclear. OBJECTIVE The aim of the present study was to determine whether alterations in mTORC1 signaling are observable following treatment with tianeptine under toxic conditions induced by B27 deprivation. Additionally, we investigated whether this drug affects synaptic proteins, neurite outgrowth, and spine density via mTORC1 signaling. METHODS Using Western blotting, we measured the phosphorylation levels of mTORC1, 4E-BP-1, p70S6K, Akt, and ERK in rat primary hippocampal neurons. Changes in BDNF, dendritic outgrowth, spine density, and synaptic proteins (PSD-95, synaptophysin, and GluR1) were measured. RESULTS Tianeptine significantly increased the phosphorylation of mTORC1, 4E-BP-1, p70S6K, Akt, and ERK. The increase in mTOR phosphorylation was blocked by the PI3K, MEK, and mTORC1 inhibitors. Tianeptine increased BDNF, dendritic outgrowth, spine density, and synaptic proteins; all of these effects were blocked by the mTORC1 inhibitor. CONCLUSIONS In this study, we demonstrated that tianeptine activates the mTORC1 signaling pathway and increases dendritic outgrowth, spine density, and synaptic proteins through mTORC1 signaling under toxic conditions in rat primary hippocampal neurons.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Hye Yeon Cho
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Chan Hong Lee
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
- Department of Health Science and Technology, Graduate School, Inje University, Busan, Republic of Korea
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Gyung-Mee Kim
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea
| | - Jun Hyung Baek
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea
| | - Young Sup Woo
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea.
- Mood Disorders Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada.
- Department of Health Science and Technology, Graduate School, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, Inje University Haeundae Paik Hospital, 1435, Jwa-dong, Haeundae-gu, Busan, 612-030, Republic of Korea.
| | - Young Hoon Kim
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea.
- Department of Health Science and Technology, Graduate School, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, Inje University Haeundae Paik Hospital, 1435, Jwa-dong, Haeundae-gu, Busan, 612-030, Republic of Korea.
| |
Collapse
|
44
|
Segal M. Dendritic spines: Morphological building blocks of memory. Neurobiol Learn Mem 2016; 138:3-9. [PMID: 27311757 DOI: 10.1016/j.nlm.2016.06.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/22/2016] [Accepted: 06/12/2016] [Indexed: 12/22/2022]
Abstract
The introduction of novel technologies, including high resolution time lapse imaging in behaving animals, molecular modification of the genome and optogenetic control of neuronal excitability have revolutionized the ability to detect subcellular changes in the brain, associated with learning and memory. The sequence of molecular cascades leading to formation, longevity and erasure of memories are being addressed in growing number of studies. Still, major issues concerning the relationship between the morphology and physiology of dendritic spines and memory mechanisms and the functional, neuronal network relevance of such parameters remain unsettled. The present review will summarize recent studies related to the immediate and long lasting changes in density, morphology and function of dendritic spines and their parent neurons following exposure to plasticity-producing stimulation in vivo and in vitro. Standing issues such as the relations between volume/shape and longevity, with respect to different classes of memories in different brain regions will be addressed. These studies indicate that the rules governing the structure/function relations of dendritic spines and memory in the brain are still not conclusive.
Collapse
Affiliation(s)
- Menahem Segal
- Department of Neurobiology, The Weizmann Institute, Rehovot 76100, Israel.
| |
Collapse
|
45
|
Sweatt JD. Dynamic DNA methylation controls glutamate receptor trafficking and synaptic scaling. J Neurochem 2016; 137:312-30. [PMID: 26849493 DOI: 10.1111/jnc.13564] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/27/2016] [Accepted: 01/30/2016] [Indexed: 12/27/2022]
Abstract
Hebbian plasticity, including long-term potentiation and long-term depression, has long been regarded as important for local circuit refinement in the context of memory formation and stabilization. However, circuit development and stabilization additionally relies on non-Hebbian, homeostatic, forms of plasticity such as synaptic scaling. Synaptic scaling is induced by chronic increases or decreases in neuronal activity. Synaptic scaling is associated with cell-wide adjustments in postsynaptic receptor density, and can occur in a multiplicative manner resulting in preservation of relative synaptic strengths across the entire neuron's population of synapses. Both active DNA methylation and demethylation have been validated as crucial regulators of gene transcription during learning, and synaptic scaling is known to be transcriptionally dependent. However, it has been unclear whether homeostatic forms of plasticity such as synaptic scaling are regulated via epigenetic mechanisms. This review describes exciting recent work that has demonstrated a role for active changes in neuronal DNA methylation and demethylation as a controller of synaptic scaling and glutamate receptor trafficking. These findings bring together three major categories of memory-associated mechanisms that were previously largely considered separately: DNA methylation, homeostatic plasticity, and glutamate receptor trafficking. This review describes exciting recent work that has demonstrated a role for active changes in neuronal DNA methylation and demethylation as a controller of synaptic scaling and glutamate receptor trafficking. These findings bring together three major categories of memory-associated mechanisms that were previously considered separately: glutamate receptor trafficking, DNA methylation, and homeostatic plasticity.
Collapse
Affiliation(s)
- J David Sweatt
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
46
|
Alterations of functional properties of hippocampal networks following repetitive closed-head injury. Exp Neurol 2016; 277:227-243. [DOI: 10.1016/j.expneurol.2015.12.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 12/09/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022]
|
47
|
Lee KFH, Soares C, Thivierge JP, Béïque JC. Correlated Synaptic Inputs Drive Dendritic Calcium Amplification and Cooperative Plasticity during Clustered Synapse Development. Neuron 2016; 89:784-99. [PMID: 26853305 DOI: 10.1016/j.neuron.2016.01.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 09/21/2015] [Accepted: 12/21/2015] [Indexed: 11/29/2022]
Abstract
The mechanisms that instruct the assembly of fine-scale features of synaptic connectivity in neural circuits are only beginning to be understood. Using whole-cell electrophysiology, two-photon calcium imaging, and glutamate uncaging in hippocampal slices, we discovered a functional coupling between NMDA receptor activation and ryanodine-sensitive intracellular calcium release that dominates the spatiotemporal dynamics of activity-dependent calcium signals during synaptogenesis. This developmentally regulated calcium amplification mechanism was tuned to detect and bind spatially clustered and temporally correlated synaptic inputs and enacted a local cooperative plasticity rule between coactive neighboring synapses. Consistent with the hypothesis that synapse maturation is spatially regulated, we observed clustering of synaptic weights in developing dendritic arbors. These results reveal developmental features of NMDA receptor-dependent calcium dynamics and local plasticity rules that are suited to spatially guide synaptic connectivity patterns in emerging neural networks.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Jean-Philippe Thivierge
- Centre for Neural Dynamics, University of Ottawa, Ottawa, ON K1H 8M5, Canada; School of Psychology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jean-Claude Béïque
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Centre for Neural Dynamics, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
48
|
Hussain S, Davanger S. Postsynaptic VAMP/Synaptobrevin Facilitates Differential Vesicle Trafficking of GluA1 and GluA2 AMPA Receptor Subunits. PLoS One 2015; 10:e0140868. [PMID: 26488171 PMCID: PMC4619507 DOI: 10.1371/journal.pone.0140868] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 10/01/2015] [Indexed: 12/03/2022] Open
Abstract
Vertebrate organisms adapt to a continuously changing environment by regulating the strength of synaptic connections between brain cells. Excitatory synapses are believed to increase their strength by vesicular insertion of transmitter glutamate receptors into the postsynaptic plasma membrane. These vesicles, however, have never been demonstrated or characterized. For the first time, we show the presence of small vesicles in postsynaptic spines, often closely adjacent to the plasma membrane and PSD (postsynaptic density). We demonstrate that they harbor vesicle-associated membrane protein 2 (VAMP2/synaptobrevin-2) and glutamate receptor subunit 1 (GluA1). Disrupting VAMP2 by tetanus toxin treatment reduces the concentration of GluA1 in the postsynaptic plasma membrane. GluA1/VAMP2-containing vesicles, but not GluA2/VAMP2-vesicles, are concentrated in postsynaptic spines relative to dendrites. Our results indicate that small postsynaptic vesicles containing GluA1 are inserted directly into the spine plasma membrane through a VAMP2-dependent mechanism.
Collapse
Affiliation(s)
- Suleman Hussain
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, P.O. Box 1105 Blindern, 0317 Oslo, Norway
| | - Svend Davanger
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Science, University of Oslo, P.O. Box 1105 Blindern, 0317 Oslo, Norway
- * E-mail:
| |
Collapse
|
49
|
Astrocyte and Neuronal Plasticity in the Somatosensory System. Neural Plast 2015; 2015:732014. [PMID: 26345481 PMCID: PMC4539490 DOI: 10.1155/2015/732014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/09/2015] [Indexed: 11/17/2022] Open
Abstract
Changing the whisker complement on a rodent's snout can lead to two forms of experience-dependent plasticity (EDP) in the neurons of the barrel cortex, where whiskers are somatotopically represented. One form, termed coding plasticity, concerns changes in synaptic transmission and connectivity between neurons. This is thought to underlie learning and memory processes and so adaptation to a changing environment. The second, called homeostatic plasticity, serves to maintain a restricted dynamic range of neuronal activity thus preventing its saturation or total downregulation. Current explanatory models of cortical EDP are almost exclusively neurocentric. However, in recent years, increasing evidence has emerged on the role of astrocytes in brain function, including plasticity. Indeed, astrocytes appear as necessary partners of neurons at the core of the mechanisms of coding and homeostatic plasticity recorded in neurons. In addition to neuronal plasticity, several different forms of astrocytic plasticity have recently been discovered. They extend from changes in receptor expression and dynamic changes in morphology to alteration in gliotransmitter release. It is however unclear how astrocytic plasticity contributes to the neuronal EDP. Here, we review the known and possible roles for astrocytes in the barrel cortex, including its plasticity.
Collapse
|
50
|
Time-dependent modulation of glutamate synapses onto 5-HT neurons by antidepressant treatment. Neuropharmacology 2015; 95:130-43. [DOI: 10.1016/j.neuropharm.2015.02.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 11/23/2022]
|