1
|
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
|
2
|
Nagase M, Nagashima T, Hamada S, Morishima M, Tohyama S, Arima-Yoshida F, Hiyoshi K, Hirano T, Ohtsuka T, Watabe AM. All-optical presynaptic plasticity induction by photoactivated adenylyl cyclase targeted to axon terminals. CELL REPORTS METHODS 2024; 4:100740. [PMID: 38521059 PMCID: PMC11045876 DOI: 10.1016/j.crmeth.2024.100740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/08/2023] [Accepted: 02/28/2024] [Indexed: 03/25/2024]
Abstract
Intracellular signaling plays essential roles in various cell types. In the central nervous system, signaling cascades are strictly regulated in a spatiotemporally specific manner to govern brain function; for example, presynaptic cyclic adenosine monophosphate (cAMP) can enhance the probability of neurotransmitter release. In the last decade, channelrhodopsin-2 has been engineered for subcellular targeting using localization tags, but optogenetic tools for intracellular signaling are not well developed. Therefore, we engineered a selective presynaptic fusion tag for photoactivated adenylyl cyclase (bPAC-Syn1a) and found its high localization at presynaptic terminals. Furthermore, an all-optical electrophysiological method revealed rapid and robust short-term potentiation by bPAC-Syn1a at brain stem-amygdala synapses in acute brain slices. Additionally, bPAC-Syn1a modulated mouse immobility behavior. These results indicate that bPAC-Syn1a can manipulate presynaptic cAMP signaling in vitro and in vivo. The all-optical manipulation technique developed in this study can help further elucidate the dynamic regulation of various cellular functions.
Collapse
Affiliation(s)
- Masashi Nagase
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Takashi Nagashima
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Shun Hamada
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Mieko Morishima
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Suguru Tohyama
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Fumiko Arima-Yoshida
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Kanae Hiyoshi
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Tomoha Hirano
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan.
| | - Ayako M Watabe
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan.
| |
Collapse
|
3
|
Wang HY, Takagi H, Stoney PN, Echeverria A, Kuhn B, Hsu KS, Takahashi T. Anoxia-induced hippocampal LTP is regeneratively produced by glutamate and nitric oxide from the neuro-glial-endothelial axis. iScience 2024; 27:109515. [PMID: 38591010 PMCID: PMC11000013 DOI: 10.1016/j.isci.2024.109515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/17/2024] [Accepted: 03/14/2024] [Indexed: 04/10/2024] Open
Abstract
Transient anoxia causes amnesia and neuronal death. This is attributed to enhanced glutamate release and modeled as anoxia-induced long-term potentiation (aLTP). aLTP is mediated by glutamate receptors and nitric oxide (·NO) and occludes stimulation-induced LTP. We identified a signaling cascade downstream of ·NO leading to glutamate release and a glutamate-·NO loop regeneratively boosting aLTP. aLTP in entothelial ·NO synthase (eNOS)-knockout mice and blocking neuronal NOS (nNOS) activity suggested that both nNOS and eNOS contribute to aLTP. Immunostaining result showed that eNOS is predominantly expressed in vascular endothelia. Transient anoxia induced a long-lasting Ca2+ elevation in astrocytes that mirrored aLTP. Blocking astrocyte metabolism or depletion of the NMDA receptor ligand D-serine abolished eNOS-dependent aLTP, suggesting that astrocytic Ca2+ elevation stimulates D-serine release from endfeet to endothelia, thereby releasing ·NO synthesized by eNOS. Thus, the neuro-glial-endothelial axis is involved in long-term enhancement of glutamate release after transient anoxia.
Collapse
Affiliation(s)
- Han-Ying Wang
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
- Academia Sinica, Institute of Biomedical Sciences, Taipei 115, Taiwan
| | - Hiroshi Takagi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
- Department of Neurosurgery, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Patrick N. Stoney
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Anai Echeverria
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Bernd Kuhn
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Kuei-Sen Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tomoyuki Takahashi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| |
Collapse
|
4
|
Dai D, Zhao T, Li Z, Li W, Chen A, Tang Y, Gao XF, Xiong L. The plasticity of neuropeptide Y-Y1 receptor system on Tac2 neurons contributes to mechanical hyperknesis during chronic itch. Theranostics 2024; 14:363-378. [PMID: 38164144 PMCID: PMC10750199 DOI: 10.7150/thno.89433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/26/2023] [Indexed: 01/03/2024] Open
Abstract
Rationale: In the physiological states, the act of scratching protects the person from harmful substances, while in certain pathological conditions, the patient suffers from chronic itch, both physically and mentally. Chronic itch sufferers are more sensitive to mechanical stimuli, and mechanical hyperknesis relief is essential for chronic itch treatment. While neuropeptide Y-Y1 receptor (NPY-Y1R) system is known to play a crucial role in modulating mechanical itch in physiological conditions, it is elusive how they are altered during chronic itch. We hypothesize that the negative regulatory effect of Y1Rs on Tac2 neurons, the key neurons that transmit mechanical itch, declines during chronic itch. Methods: We combined transgenic mice, chemogenetic manipulation, immunofluorescence, rabies virus circuit tracing, and electrophysiology to investigate the plasticity of Y1Rs on Tac2 neurons during chronic itch. Results: We found that Tac2 neurons receive direct input from Npy neurons and that inhibition of Npy neurons induces activation of Tac2 neurons. Moreover, the expression of Y1Rs on Tac2 neurons is reduced, and the regulatory effect is also reduced during chronic itch. Conclusion: Our study clarifies the plasticity of Y1Rs on Tac2 neurons during chronic itch and further elucidates the mechanism by which NPY-Y1R system is responsible for modulating mechanical itch. We highlight Y1Rs as a promising therapeutic target for mechanical hyperknesis during chronic itch.
Collapse
Affiliation(s)
- Danqing Dai
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Tiantian Zhao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Zhen Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Wanrong Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Aiwen Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Yali Tang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Xiao-Fei Gao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No.1481, Xinshi North Road, Shanghai 200434, China
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, No. 1279, Sanmen Road, Shanghai 200434, China
| |
Collapse
|
5
|
Mrestani A, Dannhäuser S, Pauli M, Kollmannsberger P, Hübsch M, Morris L, Langenhan T, Heckmann M, Paul MM. Nanoscaled RIM clustering at presynaptic active zones revealed by endogenous tagging. Life Sci Alliance 2023; 6:e202302021. [PMID: 37696575 PMCID: PMC10494931 DOI: 10.26508/lsa.202302021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
Chemical synaptic transmission involves neurotransmitter release from presynaptic active zones (AZs). The AZ protein Rab-3-interacting molecule (RIM) is important for normal Ca2+-triggered release. However, its precise localization within AZs of the glutamatergic neuromuscular junctions of Drosophila melanogaster remains elusive. We used CRISPR/Cas9-assisted genome engineering of the rim locus to incorporate small epitope tags for targeted super-resolution imaging. A V5-tag, derived from simian virus 5, and an HA-tag, derived from human influenza virus, were N-terminally fused to the RIM Zinc finger. Whereas both variants are expressed in co-localization with the core AZ scaffold Bruchpilot, electrophysiological characterization reveals that AP-evoked synaptic release is disturbed in rimV5-Znf but not in rimHA-Znf In addition, rimHA-Znf synapses show intact presynaptic homeostatic potentiation. Combining super-resolution localization microscopy and hierarchical clustering, we detect ∼10 RIMHA-Znf subclusters with ∼13 nm diameter per AZ that are compacted and increased in numbers in presynaptic homeostatic potentiation.
Collapse
Affiliation(s)
- Achmed Mrestani
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Neurology, Leipzig University Medical Center, Leipzig, Germany
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Martin Pauli
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Martha Hübsch
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Lydia Morris
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Tobias Langenhan
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Manfred Heckmann
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Mila M Paul
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
6
|
Weichard I, Taschenberger H, Gsell F, Bornschein G, Ritzau-Jost A, Schmidt H, Kittel RJ, Eilers J, Neher E, Hallermann S, Nerlich J. Fully-primed slowly-recovering vesicles mediate presynaptic LTP at neocortical neurons. Proc Natl Acad Sci U S A 2023; 120:e2305460120. [PMID: 37856547 PMCID: PMC10614622 DOI: 10.1073/pnas.2305460120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/26/2023] [Indexed: 10/21/2023] Open
Abstract
Pre- and postsynaptic forms of long-term potentiation (LTP) are candidate synaptic mechanisms underlying learning and memory. At layer 5 pyramidal neurons, LTP increases the initial synaptic strength but also short-term depression during high-frequency transmission. This classical form of presynaptic LTP has been referred to as redistribution of synaptic efficacy. However, the underlying mechanisms remain unclear. We therefore performed whole-cell recordings from layer 5 pyramidal neurons in acute cortical slices of rats and analyzed presynaptic function before and after LTP induction by paired pre- and postsynaptic neuronal activity. LTP was successfully induced in about half of the synaptic connections tested and resulted in increased synaptic short-term depression during high-frequency transmission and a decelerated recovery from short-term depression due to an increased fraction of a slow recovery component. Analysis with a recently established sequential two-step vesicle priming model indicates an increase in the abundance of fully-primed and slowly-recovering vesicles. A systematic analysis of short-term plasticity and synapse-to-synapse variability of synaptic strength at various types of synapses revealed that stronger synapses generally recover more slowly from synaptic short-term depression. Finally, pharmacological stimulation of the cyclic adenosine monophosphate and diacylglycerol signaling pathways, which are both known to promote synaptic vesicle priming, mimicked LTP and slowed the recovery from short-term depression. Our data thus demonstrate that LTP at layer 5 pyramidal neurons increases synaptic strength primarily by enlarging a subpool of fully-primed slowly-recovering vesicles.
Collapse
Affiliation(s)
- Iron Weichard
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37075, Germany
| | - Felix Gsell
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| | - Grit Bornschein
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| | - Andreas Ritzau-Jost
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| | - Hartmut Schmidt
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| | - Robert J. Kittel
- Department of Animal Physiology, Institute of Biology, Leipzig University, Leipzig04103, Germany
| | - Jens Eilers
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| | - Erwin Neher
- Emeritus Laboratory of Membrane Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen37070, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells”, University of Göttingen, Göttingen37073, Germany
| | - Stefan Hallermann
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| | - Jana Nerlich
- Faculty of Medicine, Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig04103, Germany
| |
Collapse
|
7
|
Götz J, Wieters F, Fritz VJ, Käsgen O, Kalantari A, Fink GR, Aswendt M. Temporal and Spatial Gene Expression Profile of Stroke Recovery Genes in Mice. Genes (Basel) 2023; 14:454. [PMID: 36833381 PMCID: PMC9956317 DOI: 10.3390/genes14020454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Stroke patients show some degree of spontaneous functional recovery, but this is not sufficient to prevent long-term disability. One promising approach is to characterize the dynamics of stroke recovery genes in the lesion and distant areas. We induced sensorimotor cortex lesions in adult C57BL/6J mice using photothrombosis and performed qPCR on selected brain areas at 14, 28, and 56 days post-stroke (P14-56). Based on the grid walk and rotating beam test, the mice were classified into two groups. The expression of cAMP pathway genes Adora2a, Pde10a, and Drd2, was higher in poor- compared to well-recovered mice in contralesional primary motor cortex (cl-MOp) at P14&56 and cl-thalamus (cl-TH), but lower in cl-striatum (cl-Str) at P14 and cl-primary somatosensory cortex (cl-SSp) at P28. Plasticity and axonal sprouting genes, Lingo1 and BDNF, were decreased in cl-MOp at P14 and cl-Str at P28 and increased in cl-SSp at P28 and cl-Str at P14, respectively. In the cl-TH, Lingo1 was increased, and BDNF decreased at P14. Atrx, also involved in axonal sprouting, was only increased in poor-recovered mice in cl-MOp at P28. The results underline the gene expression dynamics and spatial variability and challenge existing theories of restricted neural plasticity.
Collapse
Affiliation(s)
- Jan Götz
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Frederique Wieters
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Veronika J. Fritz
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Olivia Käsgen
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Aref Kalantari
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| | - Gereon R. Fink
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, 52425 Juelich, Germany
| | - Markus Aswendt
- Faculty of Medicine, University of Cologne, 50923 Cologne, Germany
- Department of Neurology, University Hospital Cologne, 50931 Cologne, Germany
| |
Collapse
|
8
|
Wong NF, Xu-Friedman MA. Induction of Activity-Dependent Plasticity at Auditory Nerve Synapses. J Neurosci 2022; 42:6211-6220. [PMID: 35790402 PMCID: PMC9374128 DOI: 10.1523/jneurosci.0666-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 06/25/2022] [Indexed: 11/21/2022] Open
Abstract
Exposure to nontraumatic noise in vivo drives long-lasting changes in auditory nerve synapses, which may influence hearing, but the induction mechanisms are not known. We mimicked activity in acute slices of the cochlear nucleus from mice of both sexes by treating them with high potassium, after which voltage-clamp recordings from bushy cells indicated that auditory nerve synapses had reduced EPSC amplitude, quantal size, and vesicle release probability (P r). The effects of high potassium were prevented by blockers of nitric oxide (NO) synthase and protein kinase A. Treatment with the NO donor, PAPA-NONOate, also decreased P r, suggesting NO plays a central role in inducing synaptic changes. To identify the source of NO, we activated auditory nerve fibers specifically using optogenetics. Strobing for 2 h led to decreased EPSC amplitude and P r, which was prevented by antagonists against ionotropic glutamate receptors and NO synthase. This suggests that the activation of AMPA and NMDA receptors in postsynaptic targets of auditory nerve fibers drives release of NO, which acts retrogradely to cause long-term changes in synaptic function in auditory nerve synapses. This may provide insight into preventing or treating disorders caused by noise exposure.SIGNIFICANCE STATEMENT Auditory nerve fibers undergo long-lasting changes in synaptic properties in response to noise exposure in vivo, which may contribute to changes in hearing. Here, we investigated the cellular mechanisms underlying induction of synaptic changes using high potassium and optogenetic stimulation in vitro and identified important signaling pathways using pharmacology. Our results suggest that auditory nerve activity drives postsynaptic depolarization through AMPA and NMDA receptors, leading to the release of nitric oxide, which acts retrogradely to regulate presynaptic neurotransmitter release. These experiments revealed that auditory nerve synapses are unexpectedly sensitive to activity and can show dramatic, long-lasting changes in a few hours that could affect hearing.
Collapse
Affiliation(s)
- Nicole F Wong
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| | - Matthew A Xu-Friedman
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| |
Collapse
|
9
|
Müller JA, Betzin J, Santos-Tejedor J, Mayer A, Oprişoreanu AM, Engholm-Keller K, Paulußen I, Gulakova P, McGovern TD, Gschossman LJ, Schönhense E, Wark JR, Lamprecht A, Becker AJ, Waardenberg AJ, Graham ME, Dietrich D, Schoch S. A presynaptic phosphosignaling hub for lasting homeostatic plasticity. Cell Rep 2022; 39:110696. [PMID: 35443170 DOI: 10.1016/j.celrep.2022.110696] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/26/2021] [Accepted: 03/29/2022] [Indexed: 11/29/2022] Open
Abstract
Stable function of networks requires that synapses adapt their strength to levels of neuronal activity, and failure to do so results in cognitive disorders. How such homeostatic regulation may be implemented in mammalian synapses remains poorly understood. Here we show that the phosphorylation status of several positions of the active-zone (AZ) protein RIM1 are relevant for synaptic glutamate release. Position RIMS1045 is necessary and sufficient for expression of silencing-induced homeostatic plasticity and is kept phosphorylated by serine arginine protein kinase 2 (SRPK2). SRPK2-induced upscaling of synaptic release leads to additional RIM1 nanoclusters and docked vesicles at the AZ and is not observed in the absence of RIM1 and occluded by RIMS1045E. Our data suggest that SRPK2 and RIM1 represent a presynaptic phosphosignaling hub that is involved in the homeostatic balance of synaptic coupling of neuronal networks.
Collapse
Affiliation(s)
- Johannes Alexander Müller
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany; Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Julia Betzin
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Jorge Santos-Tejedor
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Annika Mayer
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Ana-Maria Oprişoreanu
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Kasper Engholm-Keller
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark; Synapse Proteomics, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | | | - Polina Gulakova
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany; Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | | | - Lena Johanna Gschossman
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany; Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Eva Schönhense
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Jesse R Wark
- Synapse Proteomics, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Alf Lamprecht
- Department of Pharmaceutics, Bonn University, Bonn, Germany
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | - Ashley J Waardenberg
- Australian Institute for Tropical Health and Medicine, James Cook University, Smithfield, QLD 4878, Australia; i-Synapse, Cairns, QLD, Australia
| | - Mark E Graham
- Synapse Proteomics, Children's Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| | - Dirk Dietrich
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany.
| | - Susanne Schoch
- Section for Translational Epilepsy Research, Department of Neuropathology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
10
|
Shahoha M, Cohen R, Ben-Simon Y, Ashery U. cAMP-Dependent Synaptic Plasticity at the Hippocampal Mossy Fiber Terminal. Front Synaptic Neurosci 2022; 14:861215. [PMID: 35444523 PMCID: PMC9013808 DOI: 10.3389/fnsyn.2022.861215] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/23/2022] [Indexed: 11/24/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a crucial second messenger involved in both pre- and postsynaptic plasticity in many neuronal types across species. In the hippocampal mossy fiber (MF) synapse, cAMP mediates presynaptic long-term potentiation and depression. The main cAMP-dependent signaling pathway linked to MF synaptic plasticity acts via the activation of the protein kinase A (PKA) molecular cascade. Accordingly, various downstream putative synaptic PKA target proteins have been linked to cAMP-dependent MF synaptic plasticity, such as synapsin, rabphilin, synaptotagmin-12, RIM1a, tomosyn, and P/Q-type calcium channels. Regulating the expression of some of these proteins alters synaptic release probability and calcium channel clustering, resulting in short- and long-term changes to synaptic efficacy. However, despite decades of research, the exact molecular mechanisms by which cAMP and PKA exert their influences in MF terminals remain largely unknown. Here, we review current knowledge of different cAMP catalysts and potential downstream PKA-dependent molecular cascades, in addition to non-canonical cAMP-dependent but PKA-independent cascades, which might serve as alternative, compensatory or competing pathways to the canonical PKA cascade. Since several other central synapses share a similar form of presynaptic plasticity with the MF, a better description of the molecular mechanisms governing MF plasticity could be key to understanding the relationship between the transcriptional and computational levels across brain regions.
Collapse
Affiliation(s)
- Meishar Shahoha
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ronni Cohen
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yoav Ben-Simon
- Department of Neurophysiology, Vienna Medical University, Vienna, Austria
- *Correspondence: Yoav Ben-Simon,
| | - Uri Ashery
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Uri Ashery,
| |
Collapse
|
11
|
Gil-Miravet I, Mañas-Ojeda A, Ros-Bernal F, Castillo-Gómez E, Albert-Gascó H, Gundlach AL, Olucha-Bordonau FE. Involvement of the Nucleus Incertus and Relaxin-3/RXFP3 Signaling System in Explicit and Implicit Memory. Front Neuroanat 2021; 15:637922. [PMID: 33867946 PMCID: PMC8044989 DOI: 10.3389/fnana.2021.637922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
Telencephalic cognitive and emotional circuits/functions are strongly modulated by subcortical inputs. The main focus of past research on the nature of this modulation has been on the widespread monoamine projections to the telencephalon. However, the nucleus incertus (NI) of the pontine tegmentum provides a strong GABAergic and peptidergic innervation of the hippocampus, basal forebrain, amygdala, prefrontal cortex, and related regions; and represents a parallel source of ascending modulation of cognitive and emotional domains. NI GABAergic neurons express multiple peptides, including neuromedin-B, cholecystokinin, and relaxin-3, and receptors for stress and arousal transmitters, including corticotrophin-releasing factor and orexins/hypocretins. A functional relationship exists between NI neurons and their associated peptides, relaxin-3 and neuromedin-B, and hippocampal theta rhythm, which in turn, has a key role in the acquisition and extinction of declarative and emotional memories. Furthermore, RXFP3, the cognate receptor for relaxin-3, is a Gi/o protein-coupled receptor, and its activation inhibits the cellular accumulation of cAMP and induces phosphorylation of ERK, processes associated with memory formation in the hippocampus and amygdala. Therefore, this review summarizes the role of NI transmitter systems in relaying stress- and arousal-related signals to the higher neural circuits and processes associated with memory formation and retrieval.
Collapse
Affiliation(s)
- Isis Gil-Miravet
- Unitat Predepartamental de Medicina, Facultat de Ciències de la Salut, Universitat Jaume I, Castelló de la Plana, Spain
| | - Aroa Mañas-Ojeda
- Unitat Predepartamental de Medicina, Facultat de Ciències de la Salut, Universitat Jaume I, Castelló de la Plana, Spain
| | - Francisco Ros-Bernal
- Unitat Predepartamental de Medicina, Facultat de Ciències de la Salut, Universitat Jaume I, Castelló de la Plana, Spain
| | - Esther Castillo-Gómez
- Unitat Predepartamental de Medicina, Facultat de Ciències de la Salut, Universitat Jaume I, Castelló de la Plana, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Hector Albert-Gascó
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge, United Kingdom
| | - Andrew L Gundlach
- The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Francisco E Olucha-Bordonau
- Unitat Predepartamental de Medicina, Facultat de Ciències de la Salut, Universitat Jaume I, Castelló de la Plana, Spain.,Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
| |
Collapse
|
12
|
Oldani S, Moreno-Velasquez L, Faiss L, Stumpf A, Rosenmund C, Schmitz D, Rost BR. SynaptoPAC, an optogenetic tool for induction of presynaptic plasticity. J Neurochem 2020; 156:324-336. [PMID: 33037623 DOI: 10.1111/jnc.15210] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/21/2020] [Accepted: 10/01/2020] [Indexed: 11/26/2022]
Abstract
Optogenetic manipulations have transformed neuroscience in recent years. While sophisticated tools now exist for controlling the firing patterns of neurons, it remains challenging to optogenetically define the plasticity state of individual synapses. A variety of synapses in the mammalian brain express presynaptic long-term potentiation (LTP) upon elevation of presynaptic cyclic adenosine monophosphate (cAMP), but the molecular expression mechanisms as well as the impact of presynaptic LTP on network activity and behavior are not fully understood. In order to establish optogenetic control of presynaptic cAMP levels and thereby presynaptic potentiation, we developed synaptoPAC, a presynaptically targeted version of the photoactivated adenylyl cyclase bPAC. In cultures of hippocampal granule cells of Wistar rats, activation of synaptoPAC with blue light increased action potential-evoked transmission, an effect not seen in hippocampal cultures of non-granule cells. In acute brain slices of C57BL/6N mice, synaptoPAC activation immediately triggered a strong presynaptic potentiation at mossy fiber synapses in CA3, but not at Schaffer collateral synapses in CA1. Following light-triggered potentiation, mossy fiber transmission decreased within 20 min, but remained enhanced still after 30 min. The optogenetic potentiation altered the short-term plasticity dynamics of release, reminiscent of presynaptic LTP. Our work establishes synaptoPAC as an optogenetic tool that enables acute light-controlled potentiation of transmitter release at specific synapses in the brain, facilitating studies of the role of presynaptic potentiation in network function and animal behavior in an unprecedented manner. Read the Editorial Highlight for this article on page 270.
Collapse
Affiliation(s)
- Silvia Oldani
- German Center for Neurodegenerative Diseases (DZNE), 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, Berlin, Germany
| | - Laura Moreno-Velasquez
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Berlin, Germany
| | - Lukas Faiss
- German Center for Neurodegenerative Diseases (DZNE), 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, Berlin, Germany
| | - Alexander Stumpf
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Berlin, Germany
| | - Christian Rosenmund
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE), 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, Berlin, Germany.,Max-Delbruck-Centrum (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Bernstein Center for Computational Neuroscience, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Benjamin R Rost
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| |
Collapse
|
13
|
Abstract
Synaptic plasticity is a fundamental property of neurons referring to the activity-dependent changes in the strength and efficacy of synaptic transmission at preexisting synapses. Such changes can last from milliseconds to hours, days, or even longer and are involved in learning and memory as well as in development and response of the brain to injuries. Several types of synaptic plasticity have been described across neuronal types, brain regions, and species, but all of them share in one way or another capital importance of Ca2+-mediated processes. In this chapter, we will focus on the Ca2+-dependent events necessary for the induction and expression of multiple forms of synaptic plasticity.
Collapse
|
14
|
Ohadi D, Schmitt DL, Calabrese B, Halpain S, Zhang J, Rangamani P. Computational Modeling Reveals Frequency Modulation of Calcium-cAMP/PKA Pathway in Dendritic Spines. Biophys J 2019; 117:1963-1980. [PMID: 31668749 PMCID: PMC7031750 DOI: 10.1016/j.bpj.2019.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Dendritic spines are the primary excitatory postsynaptic sites that act as subcompartments of signaling. Ca2+ is often the first and most rapid signal in spines. Downstream of calcium, the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway plays a critical role in the regulation of spine formation, morphological modifications, and ultimately, learning and memory. Although the dynamics of calcium are reasonably well-studied, calcium-induced cAMP/PKA dynamics, particularly with respect to frequency modulation, are not fully explored. In this study, we present a well-mixed model for the dynamics of calcium-induced cAMP/PKA dynamics in dendritic spines. The model is constrained using experimental observations in the literature. Further, we measured the calcium oscillation frequency in dendritic spines of cultured hippocampal CA1 neurons and used these dynamics as model inputs. Our model predicts that the various steps in this pathway act as frequency modulators for calcium, and the high frequency of calcium input is filtered by adenylyl cyclase 1 and phosphodiesterases in this pathway such that cAMP/PKA only responds to lower frequencies. This prediction has important implications for noise filtering and long-timescale signal transduction in dendritic spines. A companion manuscript presents a three-dimensional spatial model for the same pathway.
Collapse
Affiliation(s)
- Donya Ohadi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California
| | - Danielle L Schmitt
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Barbara Calabrese
- Division of Biological Sciences and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Shelley Halpain
- Division of Biological Sciences and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Jin Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California.
| |
Collapse
|
15
|
Ito W, Morozov A. Prefrontal-amygdala plasticity enabled by observational fear. Neuropsychopharmacology 2019; 44:1778-1787. [PMID: 30759453 PMCID: PMC6785088 DOI: 10.1038/s41386-019-0342-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 01/07/2019] [Accepted: 02/06/2019] [Indexed: 01/10/2023]
Abstract
Observing fear in others (OF) is a form of social stress. In mice, it enhances inhibitory avoidance learning and causes the formation of silent synapses in the prefrontal-amygdala pathway. Here, we report that OF made that pathway prone to facilitation both ex vivo and in vivo. Ex vivo, OF enabled induction of long-term potentiation (LTP), expressed mostly postsynaptically and occluded by inhibitory avoidance training. In vivo, OF enabled facilitation of the dmPFC-BLA pathway by inhibitory avoidance training. The facilitation persisted during the first 4 h after the training when the prefrontal cortex and amygdala are involved in memory consolidation. Thus, the OF-generated silent synapses likely enable plasticity that may enhance the consolidation of inhibitory avoidance memories.
Collapse
Affiliation(s)
- Wataru Ito
- Virginia Tech Carilion Research Institute, Roanoke, Virginia, USA.
| | - Alexei Morozov
- Virginia Tech Carilion Research Institute, Roanoke, Virginia, USA. .,School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia, USA. .,Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA.
| |
Collapse
|
16
|
Mechanisms of fear learning and extinction: synaptic plasticity-fear memory connection. Psychopharmacology (Berl) 2019; 236:163-182. [PMID: 30415278 PMCID: PMC6374177 DOI: 10.1007/s00213-018-5104-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/02/2018] [Indexed: 12/21/2022]
Abstract
RATIONALE The ability to memorize threat-associated cues and subsequently react to them, exhibiting escape or avoidance responses, is an essential, often life-saving behavioral mechanism that can be experimentally studied using the fear (threat) conditioning training paradigm. Presently, there is substantial evidence supporting the Synaptic Plasticity-Memory (SPM) hypothesis in relation to the mechanisms underlying the acquisition, retention, and extinction of conditioned fear memory. OBJECTIVES The purpose of this review article is to summarize findings supporting the SPM hypothesis in the context of conditioned fear control, applying the set of criteria and tests which were proposed as necessary to causally link lasting changes in synaptic transmission in corresponding neural circuits to fear memory acquisition and extinction with an emphasis on their pharmacological diversity. RESULTS The mechanisms of synaptic plasticity in fear circuits exhibit complex pharmacological profiles and satisfy all four SPM criteria-detectability, anterograde alteration, retrograde alteration, and mimicry. CONCLUSION The reviewed findings, accumulated over the last two decades, provide support for both necessity and sufficiency of synaptic plasticity in fear circuits for fear memory acquisition and retention, and, in part, for fear extinction, with the latter requiring additional experimental work.
Collapse
|
17
|
Active Zone Proteins RIM1αβ Are Required for Normal Corticostriatal Transmission and Action Control. J Neurosci 2018; 39:1457-1470. [PMID: 30559150 DOI: 10.1523/jneurosci.1940-18.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/13/2018] [Accepted: 12/04/2018] [Indexed: 11/21/2022] Open
Abstract
Dynamic regulation of synaptic transmission at cortical inputs to the dorsal striatum is considered critical for flexible and efficient action learning and control. Presynaptic mechanisms governing the properties and plasticity of glutamate release from these inputs are not fully understood, and the corticostriatal synaptic processes that support normal action learning and control remain unclear. Here we show in male and female mice that conditional deletion of presynaptic proteins RIM1αβ (RIM1) from excitatory cortical neurons impairs corticostriatal synaptic transmission in the dorsolateral striatum. Key forms of presynaptic G-protein-coupled receptor-mediated short- and long-term striatal plasticity are spared following RIM1 deletion. Conditional RIM1 KO mice show heightened novelty-induced locomotion and impaired motor learning on the accelerating rotarod. They further show heightened self-paced instrumental responding for food and impaired learning of a habitual instrumental response strategy. Together, these findings reveal a selective role for presynaptic RIM1 in neurotransmitter release at prominent basal ganglia synapses, and provide evidence that RIM1-dependent processes help to promote the refinement of skilled actions, constrain goal-directed behaviors, and support the learning and use of habits.SIGNIFICANCE STATEMENT Our daily functioning hinges on the ability to flexibly and efficiently learn and control our actions. How the brain encodes these capacities is unclear. Here we identified a selective role for presynaptic proteins RIM1αβ in controlling glutamate release from cortical inputs to the dorsolateral striatum, a brain structure critical for action learning and control. Behavioral analysis of mice with restricted genetic deletion of RIM1αβ further revealed roles for RIM1αβ-dependent processes in the learning and refinement of motor skills and the balanced expression of goal-directed and habitual actions.
Collapse
|
18
|
Persistent but Labile Synaptic Plasticity at Excitatory Synapses. J Neurosci 2018; 38:5750-5758. [PMID: 29802202 DOI: 10.1523/jneurosci.2772-17.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 04/11/2018] [Accepted: 04/19/2018] [Indexed: 11/21/2022] Open
Abstract
Short-term synaptic plasticity contributes to many computations in the brain and allows synapses to keep a finite record of recent activity. Here we have investigated the mechanisms underlying an intriguing form of short-term plasticity termed labile LTP, at hippocampal and PFC synapses in male rats and male and female mice. In the hippocampus, labile LTP is triggered by high-frequency activation of presynaptic axons and is rapidly discharged with further activation of those axons. However, if the synapses are quiescent, they remain potentiated until further presynaptic activation. To distinguish labile LTP from NMDAR-dependent forms of potentiation, we blocked NMDARs in all experiments. Labile LTP was synapse-specific and was accompanied by a decreased paired pulse ratio, consistent with an increased release probability. Presynaptic Ca2+ and protein kinase activation during the tetanus appeared to be required for its initiation. Labile LTP was not reversed by a PKC inhibitor and did not require either RIM1α or synaptotagmin-7, proteins implicated in other forms of presynaptic short-term plasticity. Similar NMDAR-independent potentiation could be elicited at synapses in mPFC. Labile LTP allows for rapid information storage that is erased under controlled circumstances and could have a role in a variety of hippocampal and prefrontal cortical computations related to short-term memory.SIGNIFICANCE STATEMENT Changes in synaptic strength are thought to represent information storage relevant to particular nervous system tasks. A single synapse can exhibit multiple overlapping forms of plasticity that shape information transfer from presynaptic to postsynaptic neurons. Here we investigate the mechanisms underlying labile LTP, an NMDAR-independent form of plasticity induced at hippocampal synapses. The potentiation is maintained for long periods as long as the synapses are infrequently active, but with regular activation, the synapses are depotentiated. Similar NMDAR-independent potentiation can also be induced at L2/3-to-L5 synapses in mPFC. Labile LTP requires a rise in presynaptic Ca2+ and protein kinase activation but is unaffected in RIM1α or synaptotagmin-7 mutant mice. Labile LTP may contribute to short-term or working memory in hippocampus and mPFC.
Collapse
|
19
|
Matikainen-Ankney BA, Kravitz AV. Persistent effects of obesity: a neuroplasticity hypothesis. Ann N Y Acad Sci 2018; 1428:221-239. [PMID: 29741270 DOI: 10.1111/nyas.13665] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 12/21/2022]
Abstract
The obesity epidemic is a leading cause of health problems in the United States, increasing the risk of cardiovascular, endocrine, and psychiatric diseases. Although many people lose weight through changes in diet and lifestyle, keeping the weight off remains a challenge. Here, we discuss a hypothesis that seeks to explain why obesity is so persistent. There is a great degree of overlap in the circuits implicated in substance use disorder and obesity, and neural plasticity of these circuits in response to drugs of abuse is well documented. We hypothesize that obesity is also associated with neural plasticity in these circuits, and this may underlie persistent changes in behavior, energy balance, and body weight. Here, we discuss how obesity-associated reductions in motivation and physical activity may be rooted in neurophysiological alterations in these circuits. Such plasticity may alter how humans and animals use, expend, and store energy, even after weight loss.
Collapse
Affiliation(s)
- Bridget A Matikainen-Ankney
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Alexxai V Kravitz
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland.,National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
20
|
Lømo T. Discovering long-term potentiation (LTP) - recollections and reflections on what came after. Acta Physiol (Oxf) 2018; 222. [PMID: 28719040 DOI: 10.1111/apha.12921] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/17/2017] [Accepted: 07/11/2017] [Indexed: 01/31/2023]
Abstract
Chance events led me to a lifelong career in scientific research. They paved the way for being the first to see long-term potentiation of synaptic efficiency (LTP) in Per Andersen's laboratory in Oslo in 1966. Here I describe my way to this discovery and the experiments with Tim Bliss in 1968-1969 that led to Bliss and Lømo, 1973. Surprisingly, we later failed to reproduce these results. I discuss possible reasons for this failure, which made us both leave LTP research, in my case for good, in Tim's case for several years. After 30 years of work in a different field, I renewed my interest in the hippocampus and LTP in the early 2000s and published, for the first time, results that I had obtained 40 years earlier. Here I present my take on how interest in and research on LTP evolved after the early years. This includes a discussion of the functions of hippocampus as seen in those early days, the case of patient H.M., Donald Hebb's place in the story, the search for 'memory molecules' such as PKMζ, and the primary site for LTP expression (pre- and/or post-synaptic?). Throughout, I reflect on my life in science, how science is done and what drives it. The reflections are quite personal and I admit to mixed feelings about broadcasting them.
Collapse
Affiliation(s)
- T. Lømo
- Institute of Basic Medical Sciences; University of Oslo; Oslo Norway
| |
Collapse
|
21
|
Differential Regulation of Evoked and Spontaneous Release by Presynaptic NMDA Receptors. Neuron 2017; 96:839-855.e5. [DOI: 10.1016/j.neuron.2017.09.030] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/03/2017] [Accepted: 09/19/2017] [Indexed: 01/04/2023]
|
22
|
Yasmin F, Saxena K, McEwen BS, Chattarji S. The delayed strengthening of synaptic connectivity in the amygdala depends on NMDA receptor activation during acute stress. Physiol Rep 2017; 4:4/20/e13002. [PMID: 27798355 PMCID: PMC5099964 DOI: 10.14814/phy2.13002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/20/2016] [Indexed: 01/21/2023] Open
Abstract
There is growing evidence that stress leads to contrasting patterns of structural plasticity in the hippocampus and amygdala, two brain areas implicated in the cognitive and affective symptoms of stress‐related psychiatric disorders. Acute stress has been shown to trigger a delayed increase in the density of dendritic spines in the basolateral amygdala (BLA) of rodents. However, the physiological correlates of this delayed spinogenesis in the BLA remain unexplored. Furthermore, NMDA receptors (NMDARs) have been known to underlie chronic stress‐induced structural plasticity in the hippocampus, but nothing is known about the role of these receptors in the delayed spinogenesis, and its physiological consequences, in the BLA following acute stress. Here, using whole‐cell recordings in rat brain slices, we find that a single exposure to 2‐h immobilization stress enhances the frequency, but not amplitude, of miniature excitatory postsynaptic currents (mEPSCs) recorded from principal neurons in the BLA 10 days later. This was also accompanied by faster use‐dependent block of NMDA receptor currents during repeated stimulation of thalamic inputs to the BLA, which is indicative of higher presynaptic release probability at these inputs 10 days later. Furthermore, targeted in vivo infusion of the NMDAR‐antagonist APV into the BLA during the acute stress prevents the increase in mEPSC frequency and spine density 10 days later. Together, these results identify a role for NMDARs during acute stress in both the physiological and morphological strengthening of synaptic connectivity in the BLA in a delayed fashion. These findings also raise the possibility that activation of NMDA receptors during stress may serve as a common molecular mechanism despite the divergent patterns of plasticity that eventually emerge after stress in the amygdala and hippocampus.
Collapse
Affiliation(s)
- Farhana Yasmin
- National Centre for Biological Sciences, Bangalore, India
| | - Kapil Saxena
- National Centre for Biological Sciences, Bangalore, India.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, USA
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Bangalore, India .,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,Centre for Integrative Physiology, Deanery of Biomedical Sciences, The University of Edinburgh, George Square, Edinburgh, UK
| |
Collapse
|
23
|
Zhang T, Chen T, Chen P, Zhang B, Hong J, Chen L. MPTP-Induced Dopamine Depletion in Basolateral Amygdala via Decrease of D2R Activation Suppresses GABA A Receptors Expression and LTD Induction Leading to Anxiety-Like Behaviors. Front Mol Neurosci 2017; 10:247. [PMID: 28824377 PMCID: PMC5545577 DOI: 10.3389/fnmol.2017.00247] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/24/2017] [Indexed: 11/13/2022] Open
Abstract
Anxiety disorders commonly occur in Parkinson’s disease. Using field potential recording and patch-clamp recording, we evaluated influence of MPTP-reduced dopaminergic afferent in basolateral amygdala (BLA), a main region for affective regulation, on excitatory–inhibitory circuits and synaptic plasticity. Field excitatory post-synaptic potential (fEPSP) slopes at external capsule-BLA synapses were increased in MPTP-mice with decreases in paired-pulse facilitation and long-term potentiation amplitude, which were corrected by bath-application of D2R agonist quinpirole or cannabinoid type 1 receptors agonist WIN55,212-2, but not D1R agonist SKF38393. Compared to single waveform fEPSP in control mice, a multi-spike waveform fEPSP was observed in MPTP-mice with prolongation of duration and an increase in paired-pulse inhibition, which were recovered by BLA-injection of quinpirole for 2 days rather than bath-application. Density of GABA-evoked current (IGABA) in BLA principal neurons and GABAAR-α2 subunit expression were reduced in MPTP-mice, which were recovered by administration of quinpirole. Decline of PKC phosphorylation in BLA of MPTP-mice was corrected by bath-application of quinpirole, but not SKF38393. In MPTP-mice, BLA-injection of quinpirole or PKC activator PMA could recover GABAAR expression, which was sensitive to PKC inhibitor GF109203X. The impairment of long-term depression (LTD) in MPTP-mice was rescued by bath-application of GABAAR agonist muscimol or BLA-injection of quinpirole and PMA. Finally, BLA-injection of muscimol, quinpirole or PMA relieved anxiety-like behaviors in MPTP-mice. The results indicate that the MPTP-induced dopamine depletion in BLA principal neurons through reducing D2R-mediated PKC phosphorylation suppresses GABAAR expression and activity, which impairs GABAAR-mediated inhibition and LTD induction leading to anxiety-like behaviors.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Tingting Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Peipei Chen
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Baofeng Zhang
- Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Juan Hong
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China.,Department of Physiology, Nanjing Medical UniversityNanjing, China
| |
Collapse
|
24
|
Park D, Na M, Kim JA, Lee U, Cho E, Jang M, Chang S. Activation of CaMKIV by soluble amyloid-β 1-42 impedes trafficking of axonal vesicles and impairs activity-dependent synaptogenesis. Sci Signal 2017; 10:10/487/eaam8661. [PMID: 28698220 DOI: 10.1126/scisignal.aam8661] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The prefibrillar form of soluble amyloid-β (sAβ1-42) impairs synaptic function and is associated with the early phase of Alzheimer's disease (AD). We investigated how sAβ1-42 led to presynaptic defects using a quantum dot-based, single particle-tracking method to monitor synaptic vesicle (SV) trafficking along axons. We found that sAβ1-42 prevented new synapse formation induced by chemical long-term potentiation (cLTP). In cultured rat hippocampal neurons, nanomolar amounts of sAβ1-42 impaired Ca2+ clearance from presynaptic terminals and increased the basal Ca2+ concentration. This caused an increase in the phosphorylation of Ca2+/calmodulin-dependent protein kinase IV (CaMKIV) and its substrate synapsin, which markedly inhibited SV trafficking along axons between synapses. Neurons derived from a transgenic AD mouse model had similar defects, which were prevented by an inhibitor of CaMK kinase (CaMKK; which activates CaMKIV), by antibodies against Aβ1-42, or by expression a phosphodeficient synapsin mutant. The CaMKK inhibitor also abolished the defects in activity-dependent synaptogenesis caused by sAβ1-42 Our results suggest that by disrupting SV reallocation between synapses, sAβ1-42 prevents neurons from forming new synapses or adjusting strength and activity among neighboring synapses. Targeting this mechanism might prevent synaptic dysfunction in AD patients.
Collapse
Affiliation(s)
- Daehun Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Myeongsu Na
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Jung Ah Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea.,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Unghwi Lee
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Eunji Cho
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Mirye Jang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea.,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, South Korea. .,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea.,Biomembrane Plasticity Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| |
Collapse
|
25
|
RIM1/2 in retinal ganglion cells are required for the refinement of ipsilateral axons and eye-specific segregation. Sci Rep 2017; 7:3236. [PMID: 28607399 PMCID: PMC5468276 DOI: 10.1038/s41598-017-03361-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 04/24/2017] [Indexed: 12/22/2022] Open
Abstract
Neural activity is crucial for the refinement of neuronal connections during development, but the contribution of synaptic release mechanisms is not known. In the mammalian retina, spontaneous neural activity controls the refinement of retinal projections to the dorsal lateral geniculate nucleus (dLGN) and the superior colliculus (SC) to form appropriate topographic and eye-specific maps. To evaluate the role of synaptic release, the rab-interacting molecules (RIMs), a family of active zone proteins that play a central role in calcium-triggered release, were conditionally ablated in a subset of retinal ganglion cells (RGCs). We found that this deletion is sufficient to reduce presynaptic release probability onto dLGN neurons. Furthermore, eye-specific segregation in the dLGN and topographic refinement of ipsilateral axons in the SC and the dLGN, are impaired in RIM1/2 conditional knock-out (Rim-cDKO) mice. These defects are similar to those found when retinal activity is globally disturbed. However, reduction in synaptic release had no effect on eye-specific lamination in the SC nor on the retinotopic refinement of contralateral axons in the SC. This study highlights a potential distinction between synaptic and non-synaptic roles of neuronal activity for different mapping rules operating in visual system development.
Collapse
|
26
|
SCRAPPER Selectively Contributes to Spontaneous Release and Presynaptic Long-Term Potentiation in the Anterior Cingulate Cortex. J Neurosci 2017; 37:3887-3895. [PMID: 28292828 DOI: 10.1523/jneurosci.0023-16.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/10/2017] [Accepted: 03/06/2017] [Indexed: 12/27/2022] Open
Abstract
SCRAPPER is an E3 ubiquitin ligase expressed in presynaptic terminals, neural cell body, and dendrites of the hippocampus and cortex, which is coded by the FBXL20 gene. SCRAPPER is known to regulate synaptic transmissions and long-term potentiation (LTP) in the hippocampus, but no report is available for the cortex. Here we show genetic evidence for critical roles of SCRAPPER in excitatory transmission and presynaptic LTP (pre-LTP) of the anterior cingulate cortex (ACC), a critical cortical region for pain, anxiety, and fear. Miniature and spontaneous releases, but not evoked release, of glutamate were significantly increased in SCRAPPER knock-out (SCR-KO) mice. Interestingly, SCRAPPER selectively contributes to the increases of frequency and amplitude. The pre-LTP in the ACC was completely blocked in SCR-KO mice. Our results thus provide direct evidence for SCRAPPER in both spontaneous release and pre-LTP in the ACC and reveal a potential novel target for treating anxiety-related disease.SIGNIFICANCE STATEMENT The anterior cingulate cortex (ACC) plays critical roles in pain, anxiety, and fear. Peripheral injury induces long-term changes in synaptic transmission in the ACC. Our recent study found that a presynaptic form of LTP (pre-LTP) in the ACC contributes to chronic pain-induced anxiety. Here, we show that SCRAPPER plays a critical role in ACC pre-LTP as well as synaptic transmission.
Collapse
|
27
|
Phosphodiesterase-10A Inverse Changes in Striatopallidal and Striatoentopeduncular Pathways of a Transgenic Mouse Model of DYT1 Dystonia. J Neurosci 2017; 37:2112-2124. [PMID: 28115486 DOI: 10.1523/jneurosci.3207-15.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/12/2016] [Accepted: 12/14/2016] [Indexed: 12/15/2022] Open
Abstract
We report that changes of phosphodiesterase-10A (PDE10A) can map widespread functional imbalance of basal ganglia circuits in a mouse model of DYT1 dystonia overexpressing mutant torsinA. PDE10A is a key enzyme in the catabolism of second messenger cAMP and cGMP, whose synthesis is stimulated by D1 receptors and inhibited by D2 receptors preferentially expressed in striatoentopeducuncular/substantia nigra or striatopallidal pathways, respectively. PDE10A was studied in control mice (NT) and in mice carrying human wild-type torsinA (hWT) or mutant torsinA (hMT). Quantitative analysis of PDE10A expression was assessed in different brain areas by rabbit anti-PDE10A antibody immunohistochemistry and Western blotting. PDE10A-dependent cAMP hydrolyzing activity and PDE10A mRNA were also assessed. Striatopallidal neurons were identified by rabbit anti-enkephalin antibody.In NT mice, PDE10A is equally expressed in medium spiny striatal neurons and in their projections to entopeduncular nucleus/substantia nigra and to external globus pallidus. In hMT mice, PDE10A content selectively increases in enkephalin-positive striatal neuronal bodies; moreover, PDE10A expression and activity in hMT mice, compared with NT mice, significantly increase in globus pallidus but decrease in entopeduncular nucleus/substantia nigra. Similar changes of PDE10A occur in hWT mice, but such changes are not always significant. However, PDE10A mRNA expression appears comparable among NT, hWT, and hMT mice.In DYT1 transgenic mice, the inverse changes of PDE10A in striatoentopeduncular and striatopallidal projections might result over time in an imbalance between direct and indirect pathways for properly focusing movement. The decrease of PDE10A in the striatoentopeduncular/nigral projections might lead to increased intensity and duration of D1-stimulated cAMP/cGMP signaling; conversely, the increase of PDE10A in the striatopallidal projections might lead to increased intensity and duration of D2-inhibited cAMP/cGMP signaling.SIGNIFICANCE STATEMENT In DYT1 transgenic mouse model of dystonia, PDE10A, a key enzyme in cAMP and cGMP catabolism, is downregulated in striatal projections to entopeduncular nucleus/substantia nigra, preferentially expressing D1 receptors that stimulate cAMP/cGMP synthesis. Conversely, in DYT1 mice, PDE10A is upregulated in striatal projections to globus pallidus, preferentially expressing D2 receptors that inhibit cAMP/cGMP synthesis. The inverse changes to PDE10A in striatoentopeduncular/substantia nigra and striatopallidal pathways might tightly interact downstream to dopamine receptors, likely resulting over time to increased intensity and duration respectively of D1-stimulated and D2-inhibited cAMP/cGMP signals. Therefore, PDE10A changes in the DYT1 model of dystonia can upset the functional balance of basal ganglia circuits, affecting direct and indirect pathways simultaneously.
Collapse
|
28
|
Chand KK, Lee KM, Lavidis NA, Noakes PG. Loss of laminin‐a4 results in pre‐ and postsynaptic modifications at the neuromuscular junction. FASEB J 2016; 31:1323-1336. [DOI: 10.1096/fj.201600899r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/06/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Kirat K. Chand
- School of Biomedical Sciences St. Lucia Queensland Australia
| | - Kah Meng Lee
- School of Biomedical Sciences St. Lucia Queensland Australia
| | | | - Peter G. Noakes
- School of Biomedical Sciences St. Lucia Queensland Australia
- Queensland Brain InstituteThe University of Queensland St. Lucia Queensland Australia
| |
Collapse
|
29
|
Lanore F, Silver RA. Extracting quantal properties of transmission at central synapses. NEUROMETHODS 2016; 113:193-211. [PMID: 30245548 DOI: 10.1007/978-1-4939-3411-9_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chemical synapses enable neurons to communicate rapidly, process and filter signals and to store information. However, studying their functional properties is difficult because synaptic connections typically consist of multiple synaptic contacts that release vesicles stochastically and exhibit time-dependent behavior. Moreover, most central synapses are small and inaccessible to direct measurements. Estimation of synaptic properties from responses recorded at the soma is complicated by the presence of nonuniform release probability and nonuniform quantal properties. The presence of multivesicular release and postsynaptic receptor saturation at some synapses can also complicate the interpretation of quantal parameters. Multiple-probability fluctuation analysis (MPFA; also known as variance-mean analysis) is a method that has been developed for estimating synaptic parameters from the variance and mean amplitude of synaptic responses recorded at different release probabilities. This statistical approach, which incorporates nonuniform synaptic properties, has become widely used for studying synaptic transmission. In this chapter, we describe the statistical models used to extract quantal parameters and discuss their interpretation when applying MPFA.
Collapse
Affiliation(s)
- Frederic Lanore
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - R Angus Silver
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
30
|
Keil MF, Briassoulis G, Stratakis CA. The Role of Protein Kinase A in Anxiety Behaviors. Neuroendocrinology 2016; 103:625-39. [PMID: 26939049 DOI: 10.1159/000444880] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/19/2016] [Indexed: 11/19/2022]
Abstract
This review focuses on the genetic and other evidence supporting the notion that the cyclic AMP (cAMP) signaling pathway and its mediator, the protein kinase A (PKA) enzyme, which respond to environmental stressors and regulate stress responses, are central to the pathogenesis of disorders related to anxiety. We describe the PKA pathway and review in vitro animal studies (mouse) and other evidence that support the importance of PKA in regulating behaviors that lead to anxiety. Since cAMP signaling and PKA have been pharmacologically exploited since the 1940s (even before the identification of cAMP as a second messenger with PKA as its mediator) for a number of disorders from asthma to cardiovascular diseases, there is ample opportunity to develop therapies using this new knowledge about cAMP, PKA, and anxiety disorders.
Collapse
Affiliation(s)
- Margaret F Keil
- Section on Endocrinology and Genetics, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Md., USA
| | | | | |
Collapse
|
31
|
Banerjee A, Larsen RS, Philpot BD, Paulsen O. Roles of Presynaptic NMDA Receptors in Neurotransmission and Plasticity. Trends Neurosci 2015; 39:26-39. [PMID: 26726120 DOI: 10.1016/j.tins.2015.11.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/30/2015] [Accepted: 11/10/2015] [Indexed: 01/01/2023]
Abstract
Presynaptic NMDA receptors (preNMDARs) play pivotal roles in excitatory neurotransmission and synaptic plasticity. They facilitate presynaptic neurotransmitter release and modulate mechanisms controlling synaptic maturation and plasticity during formative periods of brain development. There is an increasing understanding of the roles of preNMDARs in experience-dependent synaptic and circuit-specific computation. In this review we summarize the latest understanding of compartment-specific expression and function of preNMDARs, and how they contribute to synapse-specific and circuit-level information processing.
Collapse
Affiliation(s)
- Abhishek Banerjee
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Benjamin D Philpot
- Department of Cell Biology and Physiology, Neuroscience Center, and Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA.
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
32
|
Jones GL, Soden ME, Knakal CR, Lee H, Chung AS, Merriam EB, Zweifel LS. A genetic link between discriminative fear coding by the lateral amygdala, dopamine, and fear generalization. eLife 2015; 4. [PMID: 26402461 PMCID: PMC4621744 DOI: 10.7554/elife.08969] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 09/23/2015] [Indexed: 01/01/2023] Open
Abstract
The lateral amygdala (LA) acquires differential coding of predictive and non-predictive fear stimuli that is critical for proper fear memory assignment. The neurotransmitter dopamine is an important modulator of LA activity and facilitates fear memory formation, but whether dopamine neurons aid in the establishment of discriminative fear coding by the LA is unknown. NMDA-type glutamate receptors in dopamine neurons are critical for the prevention of generalized fear following an aversive experience, suggesting a potential link between a cell autonomous function of NMDAR in dopamine neurons and fear coding by the LA. Here, we utilized mice with a selective genetic inactivation functional NMDARs in dopamine neurons (DAT-NR1 KO mice) combined with behavior, in vivo electrophysiology, and ex vivo electrophysiology in LA neurons to demonstrate that plasticity underlying differential fear coding in the LA is regulated by NMDAR signaling in dopamine neurons and alterations in this plasticity is associated non-discriminative cued-fear responses. DOI:http://dx.doi.org/10.7554/eLife.08969.001 When we experience a situation that causes us to feel fearful, the brain processes information about the events that led up to it. This information is encoded by groups of nerve cells called neurons in a region of the brain called the lateral amygdala. The nerve cells communicate with each other through chemicals called neurotransmitters. At a junction between two neurons—called a synapse—neurotransmitters are released from one cell and influence the activity of the other cell. Long-term changes in the strength of these communications in response to specific cues underlie the formation of memories about fearful events. When these changes occur incorrectly they can lead to memories about particular events becoming inaccurate, which can lead to fear being associated with related, but non-threatening, situations. This ‘generalization’ of fear can lead to generalized anxiety disorder and post-traumatic stress disorder. Dopamine is a neurotransmitter that plays an important role in forming memories of fearful events. However, it is not clear whether neurons that release dopamine are also involved in correctly discriminating fearful events from non-fearful ones. ‘Receptor’ proteins called NMDARs on the surface of neurons that release dopamine are critical for preventing generalized fear. These receptors detect another neurotransmitter called glutamate. Jones et al. used genetics and ‘electrophysiology’ techniques to study these receptors in mice. The experiments show that a gene that encodes part of an NMDAR in dopamine neurons plays a key role in how fear memories are formed. When this gene is selectively switched off in the dopamine neurons, mice are more likely to develop generalized fear and anxiety behaviors after a threatening experience. The experiments also demonstrate that these generalized threat responses are associated with differences in the way the synaptic connections in the lateral amygdala are strengthened. The next major challenge will be to find out which specific synaptic connections are strengthened and to establish how dopamine neuron activity patterns influences this connectivity. DOI:http://dx.doi.org/10.7554/eLife.08969.002
Collapse
Affiliation(s)
- Graham L Jones
- Department of Psychiatry and Behavioral Sciences and the Department of Pharmacology, University of Washington, Seattle, United States
| | - Marta E Soden
- Department of Psychiatry and Behavioral Sciences and the Department of Pharmacology, University of Washington, Seattle, United States
| | - Cerise R Knakal
- Department of Psychiatry and Behavioral Sciences and the Department of Pharmacology, University of Washington, Seattle, United States
| | - Heather Lee
- Department of Psychiatry and Behavioral Sciences and the Department of Pharmacology, University of Washington, Seattle, United States
| | - Amanda S Chung
- Department of Psychiatry and Behavioral Sciences and the Department of Pharmacology, University of Washington, Seattle, United States
| | - Elliott B Merriam
- Department of Psychiatry and Behavioral Sciences and the Department of Pharmacology, University of Washington, Seattle, United States
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences and the Department of Pharmacology, University of Washington, Seattle, United States
| |
Collapse
|
33
|
Enhanced dopamine-dependent hippocampal plasticity after single MK-801 application. Neuropsychopharmacology 2015; 40:987-95. [PMID: 25315194 PMCID: PMC4330513 DOI: 10.1038/npp.2014.276] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 09/17/2014] [Accepted: 09/25/2014] [Indexed: 11/09/2022]
Abstract
Dopaminergic hyperfunction and N-methyl-D-aspartate receptor (NMDAR) hypofunction have both been implicated in psychosis. Dopamine-releasing drugs and NMDAR antagonists replicate symptoms associated with psychosis in healthy humans and exacerbate symptoms in patients with schizophrenia. Though hippocampal dysfunction contributes to psychosis, the impact of NMDAR hypofunction on hippocampal plasticity remains poorly understood. Here, we used an NMDAR antagonist rodent model of psychosis to investigate hippocampal long-term potentiation (LTP). We found that single systemic NMDAR antagonism results in a region-specific, presynaptic LTP at hippocampal CA1-subiculum synapses that is induced by activation of D1/D5 dopamine receptors and modulated by L-type voltage-gated Ca(2+) channels. Thereby, our findings may provide a cellular mechanism how NMDAR antagonism can lead to an enhanced hippocampal output causing activation of the hippocampus-ventral tegmental area-loop and overdrive of the dopamine system.
Collapse
|
34
|
Hippocampal cAMP/PKA/CREB is required for neuroprotective effect of acupuncture. Physiol Behav 2014; 139:482-90. [PMID: 25481359 DOI: 10.1016/j.physbeh.2014.12.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 11/21/2014] [Accepted: 12/02/2014] [Indexed: 01/18/2023]
Abstract
Acupuncture has beneficial effects in vascular dementia (VaD) patients. The underlying mechanism, however, remains unknown. The present study was designed to investigate whether the cAMP/PKA/CREB cascade is involved in the mechanism of acupuncture in cerebral multi-infarction rats. In this study, cerebral multi-infarction was modeled in adult Wistar rats by homologous blood clot emboli. After a two-week acupuncture treatment at Zusanli (ST36), hippocampal-dependent memory was tested by employing a radial arm maze test. The hippocampus was isolated for analyses of cAMP concentration, phosphodiesterase (PDE) activity and CREB/pCREB and ERK/pERK expressions. The Morris water maze (MWM) task and CREB phosphorylation were evaluated in the presence of PKA-selective peptide inhibitor (H89). The radial arm maze test results demonstrated that acupuncture treatment at ST36 reversed hippocampal-dependent memory in impaired animals. Compared to those of the impaired group, cAMP concentration, PKA activity and pCREB and pERK expressions were increased following acupuncture therapy. Finally, the blockade of PKA reversed the increase in CREB phosphorylation and the improvement in recognitive function induced by acupuncture treatment. These results suggest that acupuncture could improve hippocampus function by modulating the cAMP/PKA/CREB signaling pathway, which represents a molecular mechanism of acupuncture for recognitive function in cerebral multi-infarction rats.
Collapse
|
35
|
Ghosh S, Chattarji S. Neuronal encoding of the switch from specific to generalized fear. Nat Neurosci 2014; 18:112-20. [DOI: 10.1038/nn.3888] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/29/2014] [Indexed: 12/14/2022]
|
36
|
Kaeser PS. Pushing synaptic vesicles over the RIM. CELLULAR LOGISTICS 2014; 1:106-110. [PMID: 21922075 DOI: 10.4161/cl.1.3.16429] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/01/2011] [Accepted: 06/01/2011] [Indexed: 01/19/2023]
Abstract
In a presynaptic nerve terminal, neurotransmitter release is largely restricted to specialized sites called active zones. Active zones consist of a complex protein network, and they organize fusion of synaptic vesicles with the presynaptic plasma membrane in response to action potentials. Rab3-interacting molecules (RIMs) are central components of active zones. In a recent series of experiments, we have systematically dissected the molecular mechanisms by which RIMs operate in synaptic vesicle release. We found that RIMs execute two critical functions of active zones by virtue of independent protein domains. They tether presyanptic Ca(2+) channels to the active zone, and they activate priming of synaptic vesicles by monomerizing homodimeric, constitutively inactive Munc13. These data indicate that RIMs orchestrate synaptic vesicle release into a coherent process. In conjunction with previous studies, they suggest that RIMs form a molecular platform on which plasticity of synaptic vesicle release can operate.
Collapse
Affiliation(s)
- Pascal S Kaeser
- Stanford Institute for Neuro-Innovation & Translational Neurosciences; Department of Molecular and Cellular Physiology; Stanford University; Stanford, CA USA
| |
Collapse
|
37
|
Li C, Rainnie DG. Bidirectional regulation of synaptic plasticity in the basolateral amygdala induced by the D1-like family of dopamine receptors and group II metabotropic glutamate receptors. J Physiol 2014; 592:4329-51. [PMID: 25107924 PMCID: PMC4215780 DOI: 10.1113/jphysiol.2014.277715] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022] Open
Abstract
Competing mechanisms of long-term potentiation (LTP) and long-term depression (LTD) in principal neurons of the basolateral amygdala (BLA) are thought to underlie the acquisition and consolidation of fear memories, and their subsequent extinction. However, no study to date has examined the locus of action and/or the cellular mechanism(s) by which these processes interact. Here, we report that synaptic plasticity in the cortical pathway onto BLA principal neurons is frequency-dependent and shows a transition from LTD to LTP at stimulation frequencies of ∼10 Hz. At the crossover point from LTD to LTP induction we show that concurrent activation of D1 and group II metabotropic glutamate (mGluR2/3) receptors act to nullify any net change in synaptic strength. Significantly, blockade of either D1 or mGluR2/3 receptors unmasked 10 Hz stimulation-induced LTD and LTP, respectively. Significantly, prior activation of presynaptic D1 receptors caused a time-dependent attenuation of mGluR2/3-induced depotentiation of previously induced LTP. Furthermore, studies with cell type-specific postsynaptic transgene expression of designer receptors activated by designer drugs (DREADDs) suggest that the interaction results via bidirectional modulation of adenylate cyclase activity in presynaptic glutamatergic terminals. The results of our study raise the possibility that the temporal sequence of activation of either presynaptic D1 receptors or mGluR2/3 receptors may critically regulate the direction of synaptic plasticity in afferent pathways onto BLA principal neurons. Hence, the interaction of these two neurotransmitter systems may represent an important mechanism for bidirectional metaplasticity in BLA circuits and thus modulate the acquisition and extinction of fear memory.
Collapse
Affiliation(s)
- Chenchen Li
- Division of Behavioural Neuroscience & Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA, 30329, USA Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, 30329, USA
| | - Donald G Rainnie
- Division of Behavioural Neuroscience & Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA, 30329, USA Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, 30329, USA
| |
Collapse
|
38
|
Kintscher M, Wozny C, Johenning FW, Schmitz D, Breustedt J. Role of RIM1α in short- and long-term synaptic plasticity at cerebellar parallel fibres. Nat Commun 2014; 4:2392. [PMID: 23999086 DOI: 10.1038/ncomms3392] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/02/2013] [Indexed: 11/09/2022] Open
Abstract
The presynaptic terminals of synaptic connections are composed of a complex network of interacting proteins that collectively ensure proper synaptic transmission and plasticity characteristics. The key components of this network are the members of the RIM protein family. Here we show that RIM1α can influence short-term plasticity at cerebellar parallel-fibre synapses. We demonstrate that the loss of a single RIM isoform, RIM1α, leads to reduced calcium influx in cerebellar granule cell terminals, decreased release probability and consequently an enhanced short-term facilitation. In contrast, we find that presynaptic long-term plasticity is fully intact in the absence of RIM1α, arguing against its necessary role in the expression of this important process. Our data argue for a universal role of RIM1α in setting release probability via interaction with voltage-dependent calcium channels at different connections instead of synapse-specific functions.
Collapse
Affiliation(s)
- Michael Kintscher
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | | | | | | | | |
Collapse
|
39
|
M1-muscarinic receptors promote fear memory consolidation via phospholipase C and the M-current. J Neurosci 2014; 34:1570-8. [PMID: 24478341 DOI: 10.1523/jneurosci.1040-13.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuromodulators released during and after a fearful experience promote the consolidation of long-term memory for that experience. Because overconsolidation may contribute to the recurrent and intrusive memories of post-traumatic stress disorder, neuromodulatory receptors provide a potential pharmacological target for prevention. Stimulation of muscarinic receptors promotes memory consolidation in several conditioning paradigms, an effect primarily associated with the M1 receptor (M1R). However, neither inhibiting nor genetically disrupting M1R impairs the consolidation of cued fear memory. Using the M1R agonist cevimeline and antagonist telenzepine, as well as M1R knock-out mice, we show here that M1R, along with β2-adrenergic (β2AR) and D5-dopaminergic (D5R) receptors, regulates the consolidation of cued fear memory by redundantly activating phospholipase C (PLC) in the basolateral amygdala (BLA). We also demonstrate that fear memory consolidation in the BLA is mediated in part by neuromodulatory inhibition of the M-current, which is conducted by KCNQ channels and is known to be inhibited by muscarinic receptors. Manipulating the M-current by administering the KCNQ channel blocker XE991 or the KCNQ channel opener retigabine reverses the effects on consolidation caused by manipulating β2AR, D5R, M1R, and PLC. Finally, we show that cAMP and protein kinase A (cAMP/PKA) signaling relevant to this stage of consolidation is upstream of these neuromodulators and PLC, suggesting an important presynaptic role for cAMP/PKA in consolidation. These results support the idea that neuromodulatory regulation of ion channel activity and neuronal excitability is a critical mechanism for promoting consolidation well after acquisition has occurred.
Collapse
|
40
|
Jayachandran R, Liu X, BoseDasgupta S, Müller P, Zhang CL, Moshous D, Studer V, Schneider J, Genoud C, Fossoud C, Gambino F, Khelfaoui M, Müller C, Bartholdi D, Rossez H, Stiess M, Houbaert X, Jaussi R, Frey D, Kammerer RA, Deupi X, de Villartay JP, Lüthi A, Humeau Y, Pieters J. Coronin 1 regulates cognition and behavior through modulation of cAMP/protein kinase A signaling. PLoS Biol 2014; 12:e1001820. [PMID: 24667537 PMCID: PMC3965382 DOI: 10.1371/journal.pbio.1001820] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 02/12/2014] [Indexed: 01/14/2023] Open
Abstract
Cognitive and behavioral disorders are thought to be a result of neuronal dysfunction, but the underlying molecular defects remain largely unknown. An important signaling pathway involved in the regulation of neuronal function is the cyclic AMP/Protein kinase A pathway. We here show an essential role for coronin 1, which is encoded in a genomic region associated with neurobehavioral dysfunction, in the modulation of cyclic AMP/PKA signaling. We found that coronin 1 is specifically expressed in excitatory but not inhibitory neurons and that coronin 1 deficiency results in loss of excitatory synapses and severe neurobehavioral disabilities, including reduced anxiety, social deficits, increased aggression, and learning defects. Electrophysiological analysis of excitatory synaptic transmission in amygdala revealed that coronin 1 was essential for cyclic-AMP-protein kinase A-dependent presynaptic plasticity. We further show that upon cell surface stimulation, coronin 1 interacted with the G protein subtype Gαs to stimulate the cAMP/PKA pathway. The absence of coronin 1 or expression of coronin 1 mutants unable to interact with Gαs resulted in a marked reduction in cAMP signaling. Strikingly, synaptic plasticity and behavioral defects of coronin 1-deficient mice were restored by in vivo infusion of a membrane-permeable cAMP analogue. Together these results identify coronin 1 as being important for cognition and behavior through its activity in promoting cAMP/PKA-dependent synaptic plasticity and may open novel avenues for the dissection of signal transduction pathways involved in neurobehavioral processes.
Collapse
Affiliation(s)
| | - Xiaolong Liu
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | - Chun-Lei Zhang
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | | | - Vera Studer
- Biozentrum, University of Basel, Basel, Switzerland
| | - Jacques Schneider
- Department of Radiology, University Children Hospital, UKBB, Basel, Switzerland
| | - Christel Genoud
- Center for Cellular Imaging and NanoAnalytics, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute, Basel, Switzerland
| | | | | | - Malik Khelfaoui
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | | | | | | | | | - Xander Houbaert
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | - Rolf Jaussi
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
| | - Daniel Frey
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
| | - Richard A. Kammerer
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
| | - Xavier Deupi
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
- Condensed Matter Theory, Paul Scherrer Institute, Villigen, Switzerland
| | | | | | - Yann Humeau
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | - Jean Pieters
- Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
41
|
Vitureira N, Goda Y. Cell biology in neuroscience: the interplay between Hebbian and homeostatic synaptic plasticity. ACTA ACUST UNITED AC 2013; 203:175-86. [PMID: 24165934 PMCID: PMC3812972 DOI: 10.1083/jcb.201306030] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Synaptic plasticity, a change in the efficacy of synaptic signaling, is a key property of synaptic communication that is vital to many brain functions. Hebbian forms of long-lasting synaptic plasticity-long-term potentiation (LTP) and long-term depression (LTD)-have been well studied and are considered to be the cellular basis for particular types of memory. Recently, homeostatic synaptic plasticity, a compensatory form of synaptic strength change, has attracted attention as a cellular mechanism that counteracts changes brought about by LTP and LTD to help stabilize neuronal network activity. New findings on the cellular mechanisms and molecular players of the two forms of plasticity are uncovering the interplay between them in individual neurons.
Collapse
Affiliation(s)
- Nathalia Vitureira
- Departmento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo 11100, Uruguay
| | | |
Collapse
|
42
|
Khelfaoui M, Gambino F, Houbaert X, Ragazzon B, Müller C, Carta M, Lanore F, Srikumar BN, Gastrein P, Lepleux M, Zhang CL, Kneib M, Poulain B, Reibel-Foisset S, Vitale N, Chelly J, Billuart P, Lüthi A, Humeau Y. Lack of the presynaptic RhoGAP protein oligophrenin1 leads to cognitive disabilities through dysregulation of the cAMP/PKA signalling pathway. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130160. [PMID: 24298161 DOI: 10.1098/rstb.2013.0160] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Loss-of-function mutations in the gene encoding for the RhoGAP protein of oligophrenin-1 (OPHN1) lead to cognitive disabilities (CDs) in humans, yet the underlying mechanisms are not known. Here, we show that in mice constitutive lack of Ophn1 is associated with dysregulation of the cyclic adenosine monophosphate/phosphate kinase A (cAMP/PKA) signalling pathway in a brain-area-specific manner. Consistent with a key role of cAMP/PKA signalling in regulating presynaptic function and plasticity, we found that PKA-dependent presynaptic plasticity was completely abolished in affected brain regions, including hippocampus and amygdala. At the behavioural level, lack of OPHN1 resulted in hippocampus- and amygdala-related learning disabilities which could be fully rescued by the ROCK/PKA kinase inhibitor fasudil. Together, our data identify OPHN1 as a key regulator of presynaptic function and suggest that, in addition to reported postsynaptic deficits, loss of presynaptic plasticity contributes to the pathophysiology of CDs.
Collapse
Affiliation(s)
- Malik Khelfaoui
- Centre National de la Recherche Scientifique UPR3212, CNRS, Université de Strasbourg, , Strasbourg 67084, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
The thalamus integrates and transmits sensory information to the neocortex. The activity of thalamocortical relay (TC) cells is modulated by specific inhibitory circuits. Although this inhibition plays a crucial role in regulating thalamic activity, little is known about long-term changes in synaptic strength at these inhibitory synapses. Therefore, we studied long-term plasticity of inhibitory inputs to TC cells in the posterior medial nucleus of the thalamus by combining patch-clamp recordings with two-photon fluorescence microscopy in rat brain slices. We found that specific activity patterns in the postsynaptic TC cell induced inhibitory long-term potentiation (iLTP). This iLTP was non-Hebbian because it did not depend on the timing between presynaptic and postsynaptic activity, but it could be induced by postsynaptic burst activity alone. iLTP required postsynaptic dendritic Ca(2+) influx evoked by low-threshold Ca(2+) spikes. In contrast, tonic postsynaptic spiking from a depolarized membrane potential (-50 mV), which suppressed these low-threshold Ca(2+) spikes, induced no plasticity. The postsynaptic dendritic Ca(2+) increase triggered the synthesis of nitric oxide that retrogradely activated presynaptic guanylyl cyclase, resulting in the presynaptic expression of iLTP. The dependence of iLTP on the membrane potential and therefore on the postsynaptic discharge mode suggests that this form of iLTP might occur during sleep, when TC cells discharge in bursts. Therefore, iLTP might be involved in sleep state-dependent modulation of thalamic information processing and thalamic oscillations.
Collapse
|
44
|
Yang Y, Calakos N. Presynaptic long-term plasticity. Front Synaptic Neurosci 2013; 5:8. [PMID: 24146648 PMCID: PMC3797957 DOI: 10.3389/fnsyn.2013.00008] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/09/2013] [Indexed: 01/01/2023] Open
Abstract
Long-term synaptic plasticity is a major cellular substrate for learning, memory, and behavioral adaptation. Although early examples of long-term synaptic plasticity described a mechanism by which postsynaptic signal transduction was potentiated, it is now apparent that there is a vast array of mechanisms for long-term synaptic plasticity that involve modifications to either or both the presynaptic terminal and postsynaptic site. In this article, we discuss current and evolving approaches to identify presynaptic mechanisms as well as discuss their limitations. We next provide examples of the diverse circuits in which presynaptic forms of long-term synaptic plasticity have been described and discuss the potential contribution this form of plasticity might add to circuit function. Finally, we examine the present evidence for the molecular pathways and cellular events underlying presynaptic long-term synaptic plasticity.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pediatrics, Stanford University School of Medicine Stanford, CA, USA
| | | |
Collapse
|
45
|
NPY Y1 receptors differentially modulate GABAA and NMDA receptors via divergent signal-transduction pathways to reduce excitability of amygdala neurons. Neuropsychopharmacology 2013; 38:1352-64. [PMID: 23358240 PMCID: PMC3656378 DOI: 10.1038/npp.2013.33] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuropeptide Y (NPY) administration into the basolateral amygdala (BLA) decreases anxiety-like behavior, mediated in part through the Y1 receptor (Y1R) isoform. Activation of Y1Rs results in G-protein-mediated reduction of cAMP levels, which results in reduced excitability of amygdala projection neurons. Understanding the mechanisms linking decreased cAMP levels to reduced excitability in amygdala neurons is important for identifying novel anxiolytic targets. We studied the intracellular mechanisms of activation of Y1Rs on synaptic transmission in the BLA. Activating Y1Rs by [Leu(31),Pro(34)]-NPY (L-P NPY) reduced the amplitude of evoked NMDA-mediated excitatory postsynaptic currents (eEPSCs), without affecting AMPA-mediated eEPSCs, but conversely increased the amplitude of GABAA-mediated evoked inhibitory postsynaptic currents (eIPSCs). Both effects were abolished by the Y1R antagonist, PD160170. Intracellular GDP-β-S, or pre-treatment with either forskolin or 8Br-cAMP, eliminated the effects of L-P NPY on both NMDA- and GABAA-mediated currents. Thus, both the NMDA and GABAA effects of Y1R activation in the BLA are G-protein-mediated and cAMP-dependent. Pipette inclusion of protein kinase A (PKA) catalytic subunit blocked the effect of L-P NPY on GABAA-mediated eIPSCs, but not on NMDA-mediated eEPSCs. Conversely, activating the exchange protein activated by cAMP (Epac) with 8CPT-2Me-cAMP blocked the effect of L-P NPY on NMDA-mediated eEPSCs, but not on GABAA-mediated eIPSCs. Thus, NPY regulates amygdala excitability via two signal-transduction events, with reduced PKA activity enhancing GABAA-mediated eIPSCs and Epac deactivation reducing NMDA-mediated eEPSCs. This multipathway regulation of NMDA- and GABAA-mediated currents may be important for NPY plasticity and stress resilience in the amygdala.
Collapse
|
46
|
Pre- and postsynaptic localization of NMDA receptor subunits at hippocampal mossy fibre synapses. Neuroscience 2013; 230:139-50. [DOI: 10.1016/j.neuroscience.2012.10.061] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Revised: 10/25/2012] [Accepted: 10/26/2012] [Indexed: 11/20/2022]
|
47
|
The presynaptic active zone protein RIM1α controls epileptogenesis following status epilepticus. J Neurosci 2012; 32:12384-95. [PMID: 22956829 DOI: 10.1523/jneurosci.0223-12.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
To ensure operation of synaptic transmission within an appropriate dynamic range, neurons have evolved mechanisms of activity-dependent plasticity, including changes in presynaptic efficacy. The multidomain protein RIM1α is an integral component of the cytomatrix at the presynaptic active zone and has emerged as key mediator of presynaptically expressed forms of synaptic plasticity. We have therefore addressed the role of RIM1α in aberrant cellular plasticity and structural reorganization after an episode of synchronous neuronal activity pharmacologically induced in vivo [status epilepticus (SE)]. Post-SE, all animals developed spontaneous seizure events, but their frequency was dramatically increased in RIM1α-deficient mice (RIM1α(-/-)). We found that in wild-type mice (RIM1α(+/+)) SE caused an increase in paired-pulse facilitation in the CA1 region of the hippocampus to the level observed in RIM1α(-/-) mice before SE. In contrast, this form of short-term plasticity was not further enhanced in RIM1α-deficient mice after SE. Intriguingly, RIM1α(-/-) mice showed a unique pattern of selective hilar cell loss (i.e., endfolium sclerosis), which so far has not been observed in a genetic epilepsy animal model, as well as less severe astrogliosis and attenuated mossy fiber sprouting. These findings indicate that the decrease in release probability and altered short- and long-term plasticity as present in RIM1α(-/-) mice result in the formation of a hyperexcitable network but act in part neuroprotectively with regard to neuropathological alterations associated with epileptogenesis. In summary, our results suggest that presynaptic plasticity and proper function of RIM1α play an important part in a neuron's adaptive response to aberrant electrical activity.
Collapse
|
48
|
Rizo J, Südhof TC. The Membrane Fusion Enigma: SNAREs, Sec1/Munc18 Proteins, and Their Accomplices—Guilty as Charged? Annu Rev Cell Dev Biol 2012; 28:279-308. [DOI: 10.1146/annurev-cellbio-101011-155818] [Citation(s) in RCA: 318] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Josep Rizo
- Departments of Biophysics, Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390;
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305;
| |
Collapse
|
49
|
Miura Y, Naka M, Matsuki N, Nomura H. Differential calcium dependence in basal and forskolin-potentiated spontaneous transmitter release in basolateral amygdala neurons. Neurosci Lett 2012; 529:1-6. [PMID: 22989859 DOI: 10.1016/j.neulet.2012.09.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 08/16/2012] [Accepted: 09/04/2012] [Indexed: 11/30/2022]
Abstract
Action potential-independent transmitter release, or spontaneous release, is postulated to produce multiple postsynaptic effects (e.g., maintenance of dendritic spines and suppression of local dendritic protein synthesis). Potentiation of spontaneous release may contribute to the precise modulation of synaptic function. However, the expression mechanism underlying potentiated spontaneous release remains unclear. In this study, we investigated the involvement of extracellular and intracellular calcium in basal and potentiated spontaneous release. Miniature excitatory postsynaptic currents (mEPSCs) of the basolateral amygdala neurons in acute brain slices were recorded. Forskolin, an adenylate cyclase activator, increased mEPSC frequency, and the increase lasted at least 25 min after washout. Removal of the extracellular calcium decreased mEPSC frequency in both naïve and forskolin-treated slices. On the other hand, chelation of intracellular calcium by BAPTA-AM decreased mEPSC frequency in naïve, but not in forskolin-treated slices. A blockade of the calcium-sensing receptor (CaSR) resulted in an increase in mEPSC frequency in forskolin-treated, but not in naïve slices. These findings indicate that forskolin-induced potentiation is accompanied by changes in the mechanisms underlying Ca(2+)-dependent spontaneous release.
Collapse
Affiliation(s)
- Yuki Miura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | |
Collapse
|
50
|
Sarabdjitsingh RA, Kofink D, Karst H, de Kloet ER, Joëls M. Stress-induced enhancement of mouse amygdalar synaptic plasticity depends on glucocorticoid and ß-adrenergic activity. PLoS One 2012; 7:e42143. [PMID: 22900007 PMCID: PMC3416843 DOI: 10.1371/journal.pone.0042143] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 07/02/2012] [Indexed: 12/16/2022] Open
Abstract
Background Glucocorticoid hormones, in interaction with noradrenaline, enable the consolidation of emotionally arousing and stressful experiences in rodents and humans. Such interaction is thought to occur at least partly in the basolateral nucleus of the amygdala (BLA) which is crucially involved in emotional memory formation. Extensive evidence points to long-term synaptic potentiation (LTP) as a mechanism contributing to memory formation. Here we determined in adolescent C57/Bl6 mice the effects of stress on LTP in the LA-BLA pathway and the specific roles of corticosteroid and β-adrenergic receptor activation in this process. Principal Findings Exposure to 20 min of restraint stress (compared to control treatment) prior to slice preparation enhanced subsequent LTP induction in vitro, without affecting baseline fEPSP responses. The role of glucocorticoid receptors, mineralocorticoid receptors and β2-adrenoceptors in the effects of stress was studied by treating mice with the antagonists mifepristone, spironolactone or propranolol respectively (or the corresponding vehicles) prior to stress or control treatment. In undisturbed controls, mifepristone and propranolol administration in vivo did not influence LTP induced in vitro. By contrast, spironolactone caused a gradually attenuating form of LTP, both in unstressed and stressed mice. Mifepristone treatment prior to stress strongly reduced the ability to induce LTP in vitro. Propranolol normalized the stress-induced enhancement of LTP to control levels during the first 10 min after high frequency stimulation, after which synaptic responses further declined. Conclusions Acute stress changes BLA electrical properties such that subsequent LTP induction is facilitated. Both β-adrenergic and glucocorticoid receptors are involved in the development of these changes. Mineralocorticoid receptors are important for the maintenance of LTP in the BLA, irrespective of stress-induced changes in the circuit. The prolonged changes in BLA network function after stress may contribute to effective memory formation of emotional and stressful events.
Collapse
Affiliation(s)
- Ratna Angela Sarabdjitsingh
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|