51
|
Lea-Carnall CA, Williams SR, Sanaei-Nezhad F, Trujillo-Barreto NJ, Montemurro MA, El-Deredy W, Parkes LM. GABA Modulates Frequency-Dependent Plasticity in Humans. iScience 2020; 23:101657. [PMID: 33163932 PMCID: PMC7599432 DOI: 10.1016/j.isci.2020.101657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/27/2020] [Accepted: 10/05/2020] [Indexed: 11/18/2022] Open
Abstract
Frequency-dependent reorganization of the primary somatosensory cortex, together with perceptual changes, arises following repetitive sensory stimulation. Here, we investigate the role of GABA in this process. We co-stimulated two finger tips and measured GABA and Glx using magnetic resonance (MR) spectroscopy at the beginning and end of the stimulation. Participants performed a perceptual learning task before and after stimulation. There were 2 sessions with stimulation frequency either at or above the resonance frequency of the primary somatosensory cortex (23 and 39 Hz, respectively). Perceptual learning occurred following above resonance stimulation only, while GABA reduced during this condition. Lower levels of early GABA were associated with greater perceptual learning. One possible mechanism underlying this finding is that cortical disinhibition “unmasks” lateral connections within the cortex to permit adaptation to the sensory environment. These results provide evidence in humans for a frequency-dependent inhibitory mechanism underlying learning and suggest a mechanism-based approach for optimizing neurostimulation frequency. In the context of repetitive sensory stimulation, GABA release is frequency dependent Stimulating above the resonance frequency of the somatosensory cortex reduces GABA Perceptual learning is associated with a reduction in GABA Early GABA reduction opens a window for plasticity and learning
Collapse
Affiliation(s)
- Caroline A. Lea-Carnall
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Corresponding author
| | - Stephen R. Williams
- Division of Informatics, Imaging and Data Science, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Faezeh Sanaei-Nezhad
- Division of Informatics, Imaging and Data Science, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nelson J. Trujillo-Barreto
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Marcelo A. Montemurro
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Wael El-Deredy
- Centro de Investigación y Desarrollo en Ingeniería en Salud, Universidad de Valparaíso, Valparaíso, Chile
- Corresponding author
| | - Laura M. Parkes
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
52
|
Sethumadhavan N, Hoang TH, Strauch C, Manahan-Vaughan D. Involvement of the Postrhinal and Perirhinal Cortices in Microscale and Macroscale Visuospatial Information Encoding. Front Behav Neurosci 2020; 14:556645. [PMID: 33192363 PMCID: PMC7584114 DOI: 10.3389/fnbeh.2020.556645] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/28/2020] [Indexed: 11/29/2022] Open
Abstract
Whereas the postrhinal cortex (POR) is a critical center for the integration of egocentric and allocentric spatial information, the perirhinal cortex (PRC) plays an important role in the encoding of objects that supports spatial learning. The POR and PRC send afferents to the hippocampus, a structure that builds complex associative memories from the spatial experience. Hippocampal encoding of item-place experience is accompanied by the nuclear expression of immediate early gene (IEGs). Subfields of the Cornus ammonius and subregions of the hippocampus exhibit differentiated and distinct encoding responses, depending on whether the spatial location and relationships of large highly visible items (macroscale encoding) or small partially concealed items (microscale encoding), is learned. But to what extent the PRC and POR support hippocampal processing of different kinds of item-place representations is unclear. Using fluorescence in situ hybridization (FISH), we examined the effect of macroscale (overt, landmark) and microscale (subtle, discrete) item-place learning on the nuclear expression of the IEG, Arc. We observed an increase in Arc mRNA in the caudal part of PRC area 35 and the caudal part of the POR after macroscale, but not microscale item-place learning. The caudal part of PRC area 36, the rostral and middle parts of PRC areas 35 and 36, as well as the middle part of the POR responded to neither type of item. These results suggest that macroscale items may contain a strong identity component that is processed by specific compartments of the PRC and POR. In contrast small, microscale items are not encoded by the POR or PRC, indicating that item dimensions may play a role in the involvement of these structures in item processing.
Collapse
Affiliation(s)
- Nithya Sethumadhavan
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Thu-Huong Hoang
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Christina Strauch
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
53
|
Perez DM. α 1-Adrenergic Receptors in Neurotransmission, Synaptic Plasticity, and Cognition. Front Pharmacol 2020; 11:581098. [PMID: 33117176 PMCID: PMC7553051 DOI: 10.3389/fphar.2020.581098] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
α1-adrenergic receptors are G-Protein Coupled Receptors that are involved in neurotransmission and regulate the sympathetic nervous system through binding and activating the neurotransmitter, norepinephrine, and the neurohormone, epinephrine. There are three α1-adrenergic receptor subtypes (α1A, α1B, α1D) that are known to play various roles in neurotransmission and cognition. They are related to two other adrenergic receptor families that also bind norepinephrine and epinephrine, the β- and α2-, each with three subtypes (β1, β2, β3, α2A, α2B, α2C). Previous studies assessing the roles of α1-adrenergic receptors in neurotransmission and cognition have been inconsistent. This was due to the use of poorly-selective ligands and many of these studies were published before the characterization of the cloned receptor subtypes and the subsequent development of animal models. With the availability of more-selective ligands and the development of animal models, a clearer picture of their role in cognition and neurotransmission can be assessed. In this review, we highlight the significant role that the α1-adrenergic receptor plays in regulating synaptic efficacy, both short and long-term synaptic plasticity, and its regulation of different types of memory. We will also present evidence that the α1-adrenergic receptors, and particularly the α1A-adrenergic receptor subtype, are a potentially good target to treat a wide variety of neurological conditions with diminished cognition.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
54
|
Sumner RL, Spriggs MJ, Muthukumaraswamy SD, Kirk IJ. The role of Hebbian learning in human perception: a methodological and theoretical review of the human Visual Long-Term Potentiation paradigm. Neurosci Biobehav Rev 2020; 115:220-237. [PMID: 32562886 DOI: 10.1016/j.neubiorev.2020.03.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 11/17/2022]
Abstract
Long-term potentiation (LTP) is one of the most widely studied forms of neural plasticity, and is thought to be the principle mechanism underlying long-term memory and learning in the brain. Sensory paradigms utilising electroencephalography (EEG) and sensory stimulation to induce LTP have allowed translation from rodent and primate invasive research to non-invasive human investigations. This review focusses on visual sensory LTP induced using repetitive visual stimulation, resulting in changes in the visually evoked response recorded at the scalp with EEG. Across 15 years of use and replication in humans several major paradigm variants for eliciting visual LTP have emerged. The application of different paradigms, and the broad implementation of visual LTP across different populations combines to provide a rich and sensitive account of Hebbian LTP, and potentially non-Hebbian plasticity mechanisms. This review will conclude with a discussion of how these findings have advanced existing theories of perceptual learning by positioning Hebbian learning both alongside and within other major theories such as Predictive Coding and The Free Energy Principle.
Collapse
Affiliation(s)
| | - Meg J Spriggs
- Centre for Psychedelic Research, Division of Brain Sciences, Centre for Psychiatry, Imperial College London, UK
| | | | - Ian J Kirk
- Brain Research, New Zealand; School of Psychology, University of Auckland, New Zealand
| |
Collapse
|
55
|
Feldmann M, Beckmann D, Eysel UT, Manahan-Vaughan D. Early Loss of Vision Results in Extensive Reorganization of Plasticity-Related Receptors and Alterations in Hippocampal Function That Extend Through Adulthood. Cereb Cortex 2020; 29:892-905. [PMID: 30535137 PMCID: PMC6319173 DOI: 10.1093/cercor/bhy297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/07/2018] [Indexed: 11/15/2022] Open
Abstract
Although by adulthood cortical structures and their capacity for processing sensory information have become established and stabilized, under conditions of cortical injury, or sensory deprivation, rapid reorganization occurs. Little is known as to the impact of this kind of adaptation on cellular processes related to memory encoding. However, imaging studies in humans suggest that following loss or impairment of a sensory modality, not only cortical but also subcortical structures begin to reorganize. It is likely that these processes are supported by neurotransmitter receptors that enable synaptic and cortical plasticity. Here, we explored to what extent the expression of plasticity-related proteins (GABA-A, GABA-B, GluN1, GluN2A, GluN2B) is altered following early vision loss, and whether this impacts on hippocampal function. We observed that in the period of 2-4 months postnatally in CBA/J-mice that experience hereditary postnatal retinal degeneration, systematic changes of GABA-receptor and NMDA-receptor subunit expression occurred that emerged first in the hippocampus and developed later in the cortex, compared to control mice that had normal vision. Changes were accompanied by significant impairments in hippocampal long-term potentiation and hippocampus-dependent learning. These data indicate that during cortical adaptation to early loss of vision, hippocampal information processing is compromised, and this status impacts on the acquisition of spatial representations.
Collapse
Affiliation(s)
- Mirko Feldmann
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Daniela Beckmann
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Ulf T Eysel
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
56
|
Hippocampal Arc Induces Decay of Object Recognition Memory in Male Mice. Neuroscience 2020; 431:193-204. [DOI: 10.1016/j.neuroscience.2020.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 11/19/2022]
|
57
|
Beckmann D, Feldmann M, Shchyglo O, Manahan-Vaughan D. Hippocampal Synaptic Plasticity, Spatial Memory, and Neurotransmitter Receptor Expression Are Profoundly Altered by Gradual Loss of Hearing Ability. Cereb Cortex 2020; 30:4581-4596. [PMID: 32202614 PMCID: PMC7325716 DOI: 10.1093/cercor/bhaa061] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/20/2020] [Accepted: 02/12/2020] [Indexed: 12/27/2022] Open
Abstract
Sensory information comprises the substrate from which memories are created. Memories of spatial sensory experience are encoded by means of synaptic plasticity in the hippocampus. Hippocampal dependency on sensory information is highlighted by the fact that sudden and complete loss of a sensory modality results in an impairment of hippocampal function that persists for months. Effects are accompanied by extensive changes in the expression of neurotransmitter receptors in cortex and hippocampus, consistent with a substantial adaptive reorganization of cortical function. Whether gradual sensory loss affects hippocampal function is unclear. Progressive age-dependent hearing loss (presbycusis) is a risk factor for cognitive decline. Here, we scrutinized C57BL/6 mice that experience hereditary and cumulative deafness starting in young adulthood. We observed that 2–4 months postnatally, increases in the cortical and hippocampal expression of GluN2A and GluN2B subunits of the N-methyl-D-aspartate receptor occurred compared to control mice that lack sensory deficits. Furthermore, GABA and metabotropic glutamate receptor expression were significantly altered. Hippocampal synaptic plasticity was profoundly impaired and mice exhibited significant deficits in spatial memory. These data show that during cortical adaptation to cumulative loss of hearing, plasticity-related neurotransmitter expression is extensively altered in the cortex and hippocampus. Furthermore, cumulative sensory loss compromises hippocampal function.
Collapse
Affiliation(s)
- Daniela Beckmann
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum 44780, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum 44780, Germany
| | - Mirko Feldmann
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum 44780, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum 44780, Germany
| | - Olena Shchyglo
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum 44780, Germany
| | - Denise Manahan-Vaughan
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum 44780, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum 44780, Germany
| |
Collapse
|
58
|
Kim K, Shin W, Kang M, Lee S, Kim D, Kang R, Jung Y, Cho Y, Yang E, Kim H, Bae YC, Kim E. Presynaptic PTPσ regulates postsynaptic NMDA receptor function through direct adhesion-independent mechanisms. eLife 2020; 9:54224. [PMID: 32142410 PMCID: PMC7069723 DOI: 10.7554/elife.54224] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/04/2020] [Indexed: 12/14/2022] Open
Abstract
Synaptic adhesion molecules regulate synapse development and function. However, whether and how presynaptic adhesion molecules regulate postsynaptic NMDAR function remains largely unclear. Presynaptic LAR family receptor tyrosine phosphatases (LAR-RPTPs) regulate synapse development through mechanisms that include trans-synaptic adhesion; however, whether they regulate postsynaptic receptor functions remains unknown. Here we report that presynaptic PTPσ, a LAR-RPTP, enhances postsynaptic NMDA receptor (NMDAR) currents and NMDAR-dependent synaptic plasticity in the hippocampus. This regulation does not involve trans-synaptic adhesions of PTPσ, suggesting that the cytoplasmic domains of PTPσ, known to have tyrosine phosphatase activity and mediate protein-protein interactions, are important. In line with this, phosphotyrosine levels of presynaptic proteins, including neurexin-1, are strongly increased in PTPσ-mutant mice. Behaviorally, PTPσ-dependent NMDAR regulation is important for social and reward-related novelty recognition. These results suggest that presynaptic PTPσ regulates postsynaptic NMDAR function through trans-synaptic and direct adhesion-independent mechanisms and novelty recognition in social and reward contexts.
Collapse
Affiliation(s)
- Kyungdeok Kim
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Wangyong Shin
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Muwon Kang
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Suho Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Ryeonghwa Kang
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Yewon Jung
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Yisul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Esther Yang
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Eunjoon Kim
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| |
Collapse
|
59
|
Shandilya MCV, Gautam A. The temporal effect of hippocampal Arc in the working memory paradigm during novelty exploration. Brain Res Bull 2020; 158:51-58. [PMID: 32114002 DOI: 10.1016/j.brainresbull.2020.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/30/2020] [Accepted: 02/25/2020] [Indexed: 10/24/2022]
Abstract
Arc (activity-regulated cytoskeleton-associated protein) is one of the neuronal Immediate Early Genes (IEG), which is involved in the consolidation of memory and is an essential factor in the induction of Long-term Potentiation (LTP), Long-term Depression (LTD) and homeostatic synaptic plasticity. It has also been implicated in the increased familiarization of novel environments during reference memory paradigms. However, the Arc associated temporal effects in a working memory paradigm during novelty exploration are not well studied. Therefore, in the present study, we used spontaneous alternation behavior (SAB) test along with the expression analysis of Arc to study its temporal effects on the working memory paradigms. Using a modified SAB test, we found that the increase in the duration of exposure to a novel environment in the short time-scale (<min) increases the alternations showing that short-term habituation increases the alternation rate. Additionally, during repeated exposure to a novel environment, the alternation rates decrease after shorter inter-session interval. Parallelly, we observed the upregulation of Arc mRNA and protein level 30 min after the SAB test in the cortex and hippocampus of mice, which returns to near-basal level after two hours. The novel experience, associated with the enhanced expression of Arc, helps in the decrease of alternations in subsequent sessions. This change in alternations was absent if the environment was familiar. Further, the role of Arc during these SAB test was confirmed by the inhibition of hippocampal Arc protein through the stereotaxic infusion of Arc antisense oligodeoxynucleotides. We observed that the Arc is involved in the temporal decrease of spontaneous alternations during a series of exposures to a novel environment. Finally, the significance of these results has been discussed in the light of Wagner's Sometimes Opponent Processes model, where we suggest that Arc reduces the ability for short-term habituation during repeated exposures in the working memory paradigm, and the loss of this ability is more prominent when subjected to a novel environment.
Collapse
Affiliation(s)
- M C Vishnu Shandilya
- Molecular Neurobiology Lab, Centre for Neural and Cognitive Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Akash Gautam
- Molecular Neurobiology Lab, Centre for Neural and Cognitive Sciences, University of Hyderabad, Hyderabad, 500046, India.
| |
Collapse
|
60
|
Schill Y, Bijata M, Kopach O, Cherkas V, Abdel-Galil D, Böhm K, Schwab MH, Matsuda M, Compan V, Basu S, Bijata K, Wlodarczyk J, Bard L, Cole N, Dityatev A, Zeug A, Rusakov DA, Ponimaskin E. Serotonin 5-HT 4 receptor boosts functional maturation of dendritic spines via RhoA-dependent control of F-actin. Commun Biol 2020; 3:76. [PMID: 32060357 PMCID: PMC7021812 DOI: 10.1038/s42003-020-0791-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/23/2020] [Indexed: 01/24/2023] Open
Abstract
Activity-dependent remodeling of excitatory connections underpins memory formation in the brain. Serotonin receptors are known to contribute to such remodeling, yet the underlying molecular machinery remains poorly understood. Here, we employ high-resolution time-lapse FRET imaging in neuroblastoma cells and neuronal dendrites to establish that activation of serotonin receptor 5-HT4 (5-HT4R) rapidly triggers spatially-restricted RhoA activity and G13-mediated phosphorylation of cofilin, thus locally boosting the filamentous actin fraction. In neuroblastoma cells, this leads to cell rounding and neurite retraction. In hippocampal neurons in situ, 5-HT4R-mediated RhoA activation triggers maturation of dendritic spines. This is paralleled by RhoA-dependent, transient alterations in cell excitability, as reflected by increased spontaneous synaptic activity, apparent shunting of evoked synaptic responses, and enhanced long-term potentiation of excitatory transmission. The 5-HT4R/G13/RhoA signaling thus emerges as a previously unrecognized molecular pathway underpinning use-dependent functional remodeling of excitatory synaptic connections.
Collapse
Affiliation(s)
- Yvonne Schill
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Monika Bijata
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Olga Kopach
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Volodymyr Cherkas
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Dalia Abdel-Galil
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Katrin Böhm
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Markus H Schwab
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Michiyuki Matsuda
- Bioimaging and Cell Signaling, Kyoto University, Kyoto, 606-8501, Japan
| | | | - Subhadip Basu
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
- Computer Science and Engineering, Jadavpur University, Kolkata, 700032, India
| | - Krystian Bijata
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Jakub Wlodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Lucie Bard
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Nicholas Cole
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Dmitri A Rusakov
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
61
|
Cicvaric A, Sachernegg HM, Stojanovic T, Symmank D, Smani T, Moeslinger T, Uhrin P, Monje FJ. Podoplanin Gene Disruption in Mice Promotes in vivo Neural Progenitor Cells Proliferation, Selectively Impairs Dentate Gyrus Synaptic Depression and Induces Anxiety-Like Behaviors. Front Cell Neurosci 2020; 13:561. [PMID: 32009902 PMCID: PMC6974453 DOI: 10.3389/fncel.2019.00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Podoplanin (Pdpn), a brain-tumor-related glycoprotein identified in humans and animals, is endogenously expressed in several organs critical for life support such as kidney, lung, heart and brain. In the brain, Pdpn has been identified in proliferative nestin-positive adult neural progenitor cells and in neurons of the neurogenic hippocampal dentate gyrus (DG), a structure associated to anxiety, critical for learning and memory functions and severely damaged in people with Alzheimer's Disease (AD). The in vivo role of Pdpn in adult neurogenesis and anxiety-like behavior remained however unexplored. Using mice with disrupted Pdpn gene as a model organism and applying combined behavioral, molecular biological and electrophysiological assays, we here show that the absence of Pdpn selectively impairs long-term synaptic depression in the neurogenic DG without affecting the CA3-Schaffer's collateral-CA1 synapses. Pdpn deletion also enhanced the proliferative capacity of DG neural progenitor cells and diminished survival of differentiated neuronal cells in vitro. In addition, mice with podoplanin gene disruption showed increased anxiety-like behaviors in experimentally validated behavioral tests as compared to wild type littermate controls. Together, these findings broaden our knowledge on the molecular mechanisms influencing hippocampal synaptic plasticity and neurogenesis in vivo and reveal Pdpn as a novel molecular target for future studies addressing general anxiety disorder and synaptic depression-related memory dysfunctions.
Collapse
Affiliation(s)
- Ana Cicvaric
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannah M. Sachernegg
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Dörte Symmank
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville (IBiS)/University of Seville/CIBERCV, Seville, Spain
| | - Thomas Moeslinger
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
62
|
Strauch C, Manahan-Vaughan D. Orchestration of Hippocampal Information Encoding by the Piriform Cortex. Cereb Cortex 2020; 30:135-147. [PMID: 31220213 PMCID: PMC7029697 DOI: 10.1093/cercor/bhz077] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/08/2019] [Accepted: 03/18/2019] [Indexed: 01/03/2023] Open
Abstract
The hippocampus utilizes olfactospatial information to encode sensory experience by means of synaptic plasticity. Odor exposure is also a potent impetus for hippocampus-dependent memory retrieval. Here, we explored to what extent the piriform cortex directly impacts upon hippocampal information processing and storage. In behaving rats, test-pulse stimulation of the anterior piriform cortex (aPC) evoked field potentials in the dentate gyrus (DG). Patterned stimulation of the aPC triggered both long-term potentiation (LTP > 24 h) and short-term depression (STD), in a frequency-dependent manner. Dual stimulation of the aPC and perforant path demonstrated subordination of the aPC response, which was nonetheless completely distinct in profile to perforant path-induced DG plasticity. Correspondingly, patterned aPC stimulation resulted in somatic immediate early gene expression in the DG that did not overlap with responses elicited by perforant path stimulation. Our results support that the piriform cortex engages in specific control of hippocampal information processing and encoding. This process may underlie the unique role of olfactory cues in information encoding and retrieval of hippocampus-dependent associative memories.
Collapse
Affiliation(s)
- Christina Strauch
- Department of Neurophysiology, Medical Faculty
- International Graduate School for Neuroscience, Ruhr University Bochum, Universitaetsstr. Bochum, Germany
| | - Denise Manahan-Vaughan
- Department of Neurophysiology, Medical Faculty
- International Graduate School for Neuroscience, Ruhr University Bochum, Universitaetsstr. Bochum, Germany
| |
Collapse
|
63
|
Spatial-Memory Formation After Spaced Learning Involves ERKs1/2 Activation Through a Behavioral-Tagging Process. Sci Rep 2020; 10:98. [PMID: 31919427 PMCID: PMC6952433 DOI: 10.1038/s41598-019-57007-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/20/2019] [Indexed: 12/25/2022] Open
Abstract
The superiority of spaced over massed learning is an established fact in the formation of long-term memories (LTM). Here we addressed the cellular processes and the temporal demands of this phenomenon using a weak spatial object recognition (wSOR) training, which induces short-term memories (STM) but not LTM. We observed SOR-LTM promotion when two identical wSOR training sessions were spaced by an inter-trial interval (ITI) ranging from 15 min to 7 h, consistently with spaced training. The promoting effect was dependent on neural activity, protein synthesis and ERKs1/2 activity in the hippocampus. Based on the “behavioral tagging” hypothesis, which postulates that learning induces a neural tag that requires proteins to induce LTM formation, we propose that retraining will mainly retag the sites initially labeled by the prior training. Thus, when weak, consecutive training sessions are experienced within an appropriate spacing, the intracellular mechanisms triggered by each session would add, thereby reaching the threshold for protein synthesis required for memory consolidation. Our results suggest in addition that ERKs1/2 kinases play a dual role in SOR-LTM formation after spaced learning, both inducing protein synthesis and setting the SOR learning-tag. Overall, our findings bring new light to the mechanisms underlying the promoting effect of spaced trials on LTM formation.
Collapse
|
64
|
Uniyal A, Singh R, Akhtar A, Dhaliwal J, Kuhad A, Sah SP. Pharmacological rewriting of fear memories: A beacon for post-traumatic stress disorder. Eur J Pharmacol 2019; 870:172824. [PMID: 31778672 DOI: 10.1016/j.ejphar.2019.172824] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/13/2019] [Accepted: 11/22/2019] [Indexed: 01/08/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a psychopathological response that develops after exposure to an extreme life-threatening traumatic event. Its prevalence ranges from 0.5% to 14.5% worldwide. Due to the complex pathophysiology of PTSD, currently available treatment approaches are associated with high chances of failure, thus further research to identify better pharmacotherapeutic approaches is needed. The traumatic event associated with fear memories plays an important role in the development of PTSD and could be considered as the main culprit. PTSD patient feels frightened in a safe environment as the memories of the traumatic event are revisited. Neurocircuit involving normal processing of fear memories get disturbed in PTSD hence making a fear memory to remain to dominate even after years of trauma. Persistence of fear memories could be explained by acquisition, re-(consolidation) and extinction triad as all of these processes have been widely explored in preclinical as well as clinical studies and set a therapeutic platform for fear memory associated disorders. This review focuses on neurocircuit and pathophysiology of PTSD in context to fear memories and pharmacological targeting of fear memory for the management of PTSD.
Collapse
Affiliation(s)
- Ankit Uniyal
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, 160014, India; Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.) Varanasi, 221005, Uttar Pradesh, India
| | - Raghunath Singh
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, 160014, India
| | - Ansab Akhtar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, 160014, India
| | - Jatinder Dhaliwal
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, 160014, India
| | - Anurag Kuhad
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, 160014, India
| | - Sangeeta Pilkhwal Sah
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC-CAS, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
65
|
Prenatal melamine exposure impairs cognitive flexibility and hippocampal synaptic plasticity in adolescent and adult female rats. Pharmacol Biochem Behav 2019; 186:172791. [DOI: 10.1016/j.pbb.2019.172791] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 08/20/2019] [Accepted: 09/09/2019] [Indexed: 11/24/2022]
|
66
|
Loss of MsrB1 perturbs spatial learning and long-term potentiation/long-term depression in mice. Neurobiol Learn Mem 2019; 166:107104. [PMID: 31672630 DOI: 10.1016/j.nlm.2019.107104] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/21/2019] [Accepted: 10/27/2019] [Indexed: 12/22/2022]
Abstract
MsrB1 belongs to the methionine sulfoxide reductase family, it is also known as selenoprotein R for the sake of possessing a selenocysteine residue. It has been reported that MsrB1 could interact with actin, TRPM6, clusterin, and amyloid-beta in vitro. Thus, we presumed that MsrB1 may play an important role in central nervous system. To examine whether MsrB1 knockout has any effects on brain development or learning behavior, we carried out histological study on brains of MsrB1 deficient mice, and further tested spatial learning ability and long-term synaptic plasticity of these mice by using Morris water maze and electrophysiological methods. It was observed that loss of MsrB1 did not perturb the overall development of central nervous system except for the astrogliosis in hippocampus, however, it led mice to be incapable in spatial learning and severe impairments in LTP/LTD expression in CA1 of brain slices, along with the down-regulation of the synaptic proteins including PSD95, SYP, GluN2A and GluN2B, as well as the dramatic decrease of CaMKIIs phosphorylation at 286(287) compared with wild type mice. Taken together, these results suggest that MsrB1 is essential for mice spatial learning and LTP/LTD induction, and the MsrB1 related redox homeostasis may be involved in regulating the phosphorylation of CaMKIIs.
Collapse
|
67
|
Sadeghian A, Fathollahi Y, Javan M, Shojaei A, Kosarmadar N, Rezaei M, Mirnajafi-Zadeh J. Spatial Learning and Memory in Barnes Maze Test and Synaptic Potentiation in Schaffer Collateral-CA1 Synapses of Dorsal Hippocampus in Freely Moving Rats. Basic Clin Neurosci 2019; 10:461-468. [PMID: 32284835 PMCID: PMC7149949 DOI: 10.32598/bcn.9.10.330] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 09/02/2018] [Accepted: 12/25/2018] [Indexed: 12/26/2022] Open
Abstract
Introduction Synaptic plasticity has been suggested as the primary physiological mechanism underlying memory formation. Many experimental approaches have been used to investigate whether the mechanisms underlying Long-Term Potentiation (LTP) are activated during learning. Nevertheless, little evidence states that hippocampal-dependent learning triggers synaptic plasticity. In this study, we investigated if learning and memory in the Barnes maze test are accompanied by the occurrence of LTP in Schaffer collateral to CA1 synapses in freely moving rats. Methods The rats were implanted with a recording electrode in stratum radiatum and stimulating electrodes in Schaffer collaterals of the CA1 region in the dorsal hippocampus of the right hemisphere. Following the recovery period of at least 10 days, field potentials were recorded in freely moving animals before and after training them in Barnes maze as a hippocampal-dependent spatial learning and memory test. The slope of extracellular field Excitatory Postsynaptic Potentials (fEPSPs) was measured before and after the Barnes maze test. Results The results showed that the fEPSP slope did not change after learning and memory in the Barnes maze test, and this spatial learning did not result in a change in synaptic potentiation in the CA1 region of the hippocampus. Conclusion Spatial learning and memory in the Barnes maze test are not accompanied by LTP induction in Schaffer collateral-CA1 synapses.
Collapse
Affiliation(s)
- Azam Sadeghian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Shojaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nastaran Kosarmadar
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahmoud Rezaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
68
|
Zhao F, Siu JJ, Huang W, Askwith C, Cao L. Insulin Modulates Excitatory Synaptic Transmission and Synaptic Plasticity in the Mouse Hippocampus. Neuroscience 2019; 411:237-254. [PMID: 31146008 PMCID: PMC6612444 DOI: 10.1016/j.neuroscience.2019.05.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022]
Abstract
The administration of exogenous insulin into the hippocampus has the potential to enhance cognitive function and exert other beneficial effects. Elucidating the neurobiological substrates of insulin action and its underlying physiological mechanisms may further improve treatment efficacy. Previous work has shown that insulin affects synaptic plasticity, however there are discrepancies and contradictory conclusions between studies. Here, we used extracellular field recordings in mouse hippocampal slices to investigate how insulin acutely modulates synaptic transmission and synaptic plasticity, both of which are correlated with learning and memory processes. Our data demonstrate that insulin application inhibited basal excitatory synaptic transmission and promoted long-term potentiation (LTP) induction at hippocampal Schaffer collateral-CA1 synapses. Under similar conditions, insulin strongly activated the PI3K/AKT pathway, but had only a weak effect on the MAPK/ERK pathway. Although insulin-induced inhibition of field excitatory post-synaptic potentials (fEPSPs) was previously termed insulin-long-term depression (insulin-LTD), insulin application potentiated recovery from classically induced LTD. Further analysis suggests suppression of presynaptic neurotransmitter release contributed to the insulin-LTD. At low concentrations, insulin primarily inhibited fEPSPs; however, at high concentration, its effects were of mixed inhibition and enhancement in different recordings. Moreover, a broad spectrum protein kinase C blocker, cannabinoid receptor type 1 activator, or a high glucose concentration inhibited fEPSPs per se, and disturbed insulin's effect on fEPSP. Insulin also caused depotentiation during LTP expression and triggered depression during LTD recovery. Given the essential roles of dynamic synaptic transmission and plasticity in learning and memory, our data provide more evidence that insulin application may actively modulate hippocampal-dependent cognitive events.
Collapse
Affiliation(s)
- Fangli Zhao
- College of Medicine, The Ohio State University
| | - Jason J Siu
- College of Medicine, The Ohio State University
| | - Wei Huang
- College of Medicine, The Ohio State University
| | | | - Lei Cao
- College of Medicine, The Ohio State University.
| |
Collapse
|
69
|
Restless REM Sleep Impedes Overnight Amygdala Adaptation. Curr Biol 2019; 29:2351-2358.e4. [DOI: 10.1016/j.cub.2019.06.034] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/14/2019] [Accepted: 06/11/2019] [Indexed: 11/23/2022]
|
70
|
O'Riordan KJ, Hu NW, Rowan MJ. Aß Facilitates LTD at Schaffer Collateral Synapses Preferentially in the Left Hippocampus. Cell Rep 2019; 22:2053-2065. [PMID: 29466733 DOI: 10.1016/j.celrep.2018.01.085] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/16/2017] [Accepted: 01/26/2018] [Indexed: 01/03/2023] Open
Abstract
Promotion of long-term depression (LTD) mechanisms by synaptotoxic soluble oligomers of amyloid-β (Aß) has been proposed to underlie synaptic dysfunction in Alzheimer's disease (AD). Previously, LTD was induced by relatively non-specific electrical stimulation. Exploiting optogenetics, we studied LTD using a more physiologically diffuse spatial pattern of selective pathway activation in the rat hippocampus in vivo. This relatively sparse synaptic LTD requires both the ion channel function and GluN2B subunit of the NMDA receptor but, in contrast to electrically induced LTD, is not facilitated by boosting endogenous muscarinic acetylcholine or metabotropic glutamate 5 receptor activation. Although in the absence of Aß, there is no evidence of hippocampal LTD asymmetry, in the presence of Aß, the induction of LTD is preferentially enhanced in the left hippocampus in an mGluR5-dependent manner. This circuit-selective disruption of synaptic plasticity by Aß provides a route to understanding the development of aberrant brain lateralization in AD.
Collapse
Affiliation(s)
- Kenneth J O'Riordan
- Department of Pharmacology and Therapeutics and Institute of Neuroscience, Watts Building, Trinity College, Dublin 2, Ireland
| | - Neng-Wei Hu
- Department of Pharmacology and Therapeutics and Institute of Neuroscience, Watts Building, Trinity College, Dublin 2, Ireland; Department of Gerontology, Yijishan Hospital, Wannan Medical College, Wuhu, China.
| | - Michael J Rowan
- Department of Pharmacology and Therapeutics and Institute of Neuroscience, Watts Building, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
71
|
Neyman S, Braunewell KH, O'Connell KE, Dev KK, Manahan-Vaughan D. Inhibition of the Interaction Between Group I Metabotropic Glutamate Receptors and PDZ-Domain Proteins Prevents Hippocampal Long-Term Depression, but Not Long-Term Potentiation. Front Synaptic Neurosci 2019; 11:13. [PMID: 31057390 PMCID: PMC6482240 DOI: 10.3389/fnsyn.2019.00013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/04/2019] [Indexed: 01/07/2023] Open
Abstract
The group I metabotropic glutamate (mGlu) receptor subtypes, mGlu1 and mGlu5, strongly regulate hippocampal synaptic plasticity. Both harbor PSD-95/discs-large/ZO-1 (PDZ) motifs at their extreme carboxyl terminals, which allow interaction with the PDZ domain of Tamalin, regulate the cell surface expression of group I mGlu receptors, and may modulate their coupling to signaling proteins. We investigated the functional role of this interaction in hippocampal long-term depression (LTD). Acute intracerebral treatment of adult rats with a cell-permeable PDZ-blocking peptide (pep-mGluR-STL), designed to competitively inhibit the interaction between Tamalin and group 1 mGlu receptors, prevented expression of LTD in the hippocampal CA1 region without affecting long-term potentiation (LTP) or basal synaptic transmission. Pep-mGluR-STL prevented facilitation by the group I mGlu receptor agonist, (S)-3,5-Dihydroxyphenylglycine (DHPG), and the mGlu5 agonist, (R,S)-2-chloro-5-Hydroxyphenylglycine (CHPG), of short-term depression (STD) into LTD, suggesting that Tamalin preferentially acts by mediating signaling through mGlu5. These data support that Tamalin is essential for the persistent expression of LTD and that it subserves the effective signaling of group 1 mGlu receptors.
Collapse
Affiliation(s)
- Sergey Neyman
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Karl-Heinz Braunewell
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Kara E O'Connell
- Drug Development, School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
72
|
Hernández-Frausto M, López-Rubalcava C, Galván EJ. Progressive Alterations in Synaptic Transmission and Plasticity of Area CA1 Precede the Cognitive Impairment Associated with Neonatal Administration of MK-801. Neuroscience 2019; 404:205-217. [DOI: 10.1016/j.neuroscience.2019.01.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/26/2018] [Accepted: 01/21/2019] [Indexed: 11/26/2022]
|
73
|
Latzer P, Shchyglo O, Hartl T, Matschke V, Schlegel U, Manahan-Vaughan D, Theiss C. Blocking VEGF by Bevacizumab Compromises Electrophysiological and Morphological Properties of Hippocampal Neurons. Front Cell Neurosci 2019; 13:113. [PMID: 30971896 PMCID: PMC6445260 DOI: 10.3389/fncel.2019.00113] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/07/2019] [Indexed: 12/20/2022] Open
Abstract
A hallmark of glioblastoma multiforme (GBM) is neoangiogenesis, mediated by the overexpression of vascular endothelial growth factor (VEGF). Anti-VEGF antibodies, like bevacizumab, prolong progression-free survival in GBM, however, this treatment has been reported to be associated with a decline in neurocognitive function. Therefore, this study focused on the effects of bevacizumab on neuronal function and plasticity. We analyzed neuronal membrane properties and synaptic plasticity in rat hippocampal slices, as well as spine dynamics in dissociated hippocampal neurons, to examine the impact of bevacizumab on hippocampal function and viability. VEGF inhibition resulted in profound impairments in hippocampal synaptic plasticity as well as reductions in dendritic spine number and length. Physiological properties of hippocampal neurons were also affected. These effects of VEGF blockade on hippocampal function may play a role in compromising memory and information processing and thus, may contribute to neurocognitive dysfunction in GBM patients treated with bevacizumab.
Collapse
Affiliation(s)
- Pauline Latzer
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Olena Shchyglo
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Tim Hartl
- International Graduate School of Neuroscience, Ruhr University Bochum, Bochum, Germany.,Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Uwe Schlegel
- Department of Neurology, Knappschaftskrankenhaus, Ruhr University Bochum, Bochum, Germany
| | | | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
74
|
Ondrejcak T, Hu NW, Qi Y, Klyubin I, Corbett GT, Fraser G, Perkinton MS, Walsh DM, Billinton A, Rowan MJ. Soluble tau aggregates inhibit synaptic long-term depression and amyloid β-facilitated LTD in vivo. Neurobiol Dis 2019; 127:582-590. [PMID: 30910746 DOI: 10.1016/j.nbd.2019.03.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/01/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023] Open
Abstract
Soluble synaptotoxic aggregates of the main pathological proteins of Alzheimer's disease, amyloid β-protein (Aß) and tau, have rapid and potent inhibitory effects on long-term potentiation (LTP). Although the promotion of synaptic weakening mechanisms, including long-term depression (LTD), is posited to mediate LTP inhibition by Aß, little is known regarding the action of exogenous tau on LTD. The present study examined the ability of different assemblies of full-length human tau to affect LTD in the dorsal hippocampus of the anaesthetized rat. Unlike Aß, intracerebroventricular injection of soluble aggregates of tau (SτAs), but not monomers or fibrils, potently increased the threshold for LTD induction in a manner that required cellular prion protein. However, MTEP, an antagonist of the putative prion protein coreceptor metabotropic glutamate receptor 5, did not prevent the disruption of synaptic plasticity by SτAs. In contrast, systemic treatment with Ro 25-6981, a selective antagonist at GluN2B subunit-containing NMDA receptors, reduced SτA-mediated inhibition of LTD, but not LTP. Intriguingly, SτAs completely blocked Aß-facilitated LTD, whereas a subthreshold dose of SτAs facilitated Aß-mediated inhibition of LTP. Overall, these findings support the importance of cellular prion protein in mediating a range of, sometimes opposing, actions of soluble Aß and tau aggregates with different effector mechanisms on synaptic plasticity.
Collapse
Affiliation(s)
- Tomas Ondrejcak
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| | - Neng-Wei Hu
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland; Department of Physiology and Neurobiology, Zhengzhou University School of Medicine, Zhengzhou 450001, China
| | - Yingjie Qi
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Igor Klyubin
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Grant T Corbett
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Graham Fraser
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, UK
| | | | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Billinton
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge CB21 6GH, UK
| | - Michael J Rowan
- Department of Pharmacology & Therapeutics, Institute of Neuroscience, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
75
|
Martucci LL, Amar M, Chaussenot R, Benet G, Bauer O, de Zélicourt A, Nosjean A, Launay JM, Callebert J, Sebrié C, Galione A, Edeline JM, de la Porte S, Fossier P, Granon S, Vaillend C, Cancela JM. A multiscale analysis in CD38 -/- mice unveils major prefrontal cortex dysfunctions. FASEB J 2019; 33:5823-5835. [PMID: 30844310 DOI: 10.1096/fj.201800489r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Autism spectrum disorder (ASD) is characterized by early onset of behavioral and cognitive alterations. Low plasma levels of oxytocin (OT) have also been found in ASD patients; recently, a critical role for the enzyme CD38 in the regulation of OT release was demonstrated. CD38 is important in regulating several Ca2+-dependent pathways, but beyond its role in regulating OT secretion, it is not known whether a deficit in CD38 expression leads to functional modifications of the prefrontal cortex (PFC), a structure involved in social behavior. Here, we report that CD38-/- male mice show an abnormal cortex development, an excitation-inhibition balance shifted toward a higher excitation, and impaired synaptic plasticity in the PFC such as those observed in various mouse models of ASD. We also show that a lack of CD38 alters social behavior and emotional responses. Finally, examining neuromodulators known to control behavioral flexibility, we found elevated monoamine levels in the PFC of CD38-/- adult mice. Overall, our study unveiled major changes in PFC physiologic mechanisms and provides new evidence that the CD38-/- mouse could be a relevant model to study pathophysiological brain mechanisms of mental disorders such as ASD.-Martucci, L. L., Amar, M., Chaussenot, R., Benet, G., Bauer, O., de Zélicourt, A., Nosjean, A., Launay, J.-M., Callebert, J., Sebrié, C., Galione, A., Edeline, J.-M., de la Porte, S., Fossier, P., Granon, S., Vaillend, C., Cancela, J.-M., A multiscale analysis in CD38-/- mice unveils major prefrontal cortex dysfunctions.
Collapse
Affiliation(s)
- Lora L Martucci
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France.,INSERM Unité 1179, Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, Unité de Formation et de Recherche (UFR) des Sciences de la Santé Simone Veil, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Montigny-le-Bretonneux, France
| | - Muriel Amar
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Remi Chaussenot
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Gabriel Benet
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Oscar Bauer
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France.,Génétique Humaine et Fonctions Cognitives, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 3571, Gènes, Synapses et Cognition, CNRS, Institut Pasteur, Paris, France
| | - Antoine de Zélicourt
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France.,INSERM Unité 1179, Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, Unité de Formation et de Recherche (UFR) des Sciences de la Santé Simone Veil, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Montigny-le-Bretonneux, France
| | - Anne Nosjean
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | | | | | - Catherine Sebrié
- Imagerie par Résonance Magnétique Médicale et Multimodalité (IR4M) Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8081, Paris-Sud University, Paris-Saclay University, CNRS, Orsay, France
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Jean-Marc Edeline
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Sabine de la Porte
- INSERM Unité 1179, Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, Unité de Formation et de Recherche (UFR) des Sciences de la Santé Simone Veil, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Montigny-le-Bretonneux, France
| | - Philippe Fossier
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Sylvie Granon
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Cyrille Vaillend
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - José-Manuel Cancela
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| |
Collapse
|
76
|
Gimse K, Gorzek RC, Olin A, Osting S, Burger C. Hippocampal Homer1b/c is necessary for contextual fear conditioning and group I metabotropic glutamate receptor mediated long-term depression. Neurobiol Learn Mem 2018; 156:17-23. [PMID: 30336208 PMCID: PMC6226007 DOI: 10.1016/j.nlm.2018.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 01/24/2023]
Abstract
Coiled-coil forms of Homer1, including Homer1b and c (Homer1b/c) have been shown to play a role in hippocampal learning and memory and synaptic plasticity. We have previously found that overexpression of hippocampal Homer1c is sufficient to rescue learning and memory ability in aged learning impaired rats and in Homer1 knockout (KO) mice, and to rescue group I metabotropic glutamate receptor (mGluR1/5) mediated long-term potentiation in KO mice. Here, to determine if Homer1b/c is necessary for successful learning and memory we have utilized a rAAV5 vector expressing a Homer1b/c-targeting short hairpin RNA to knock down the expression of hippocampal Homer1b/c in adult 4-6-month old male Sprague Dawley rats. We have found that reduced hippocampal Homer1b/c expression elicits significant learning deficits in contextual fear conditioning, but not in the Morris water maze or novel object recognition tasks. Furthermore, we demonstrate that reduced hippocampal Homer1b/c is sufficient to completely block mGluR1/5 mediated long-term depression in the Schaffer collateral pathway. These results support a significant role for Homer1b/c in learning and synaptic plasticity; however, the exact role of each of these two protein isoforms in learning and memory remains elusive.
Collapse
Affiliation(s)
- Kirstan Gimse
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Medical Sciences Center, 1300 University Ave, Room 73 Bardeen, Madison, WI 53706, USA
| | - Ryan C Gorzek
- College of Letters and Science, University of Wisconsin, Madison, WI, USA
| | - Andrew Olin
- Department of Neurology, University of Wisconsin, Madison, WI, USA
| | - Sue Osting
- Department of Neurology, University of Wisconsin, Madison, WI, USA
| | - Corinna Burger
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Medical Sciences Center, 1300 University Ave, Room 73 Bardeen, Madison, WI 53706, USA; Department of Neurology, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
77
|
Lovinger DM, Abrahao KP. Synaptic plasticity mechanisms common to learning and alcohol use disorder. ACTA ACUST UNITED AC 2018; 25:425-434. [PMID: 30115764 PMCID: PMC6097767 DOI: 10.1101/lm.046722.117] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/21/2018] [Indexed: 11/24/2022]
Abstract
Alcohol use disorders include drinking problems that span a range from binge drinking to alcohol abuse and dependence. Plastic changes in synaptic efficacy, such as long-term depression and long-term potentiation are widely recognized as mechanisms involved in learning and memory, responses to drugs of abuse, and addiction. In this review, we focus on the effects of chronic ethanol (EtOH) exposure on the induction of synaptic plasticity in different brain regions. We also review findings indicating that synaptic plasticity occurs in vivo during EtOH exposure, with a focus on ex vivo electrophysiological indices of plasticity. Evidence for effects of EtOH-induced or altered synaptic plasticity on learning and memory and EtOH-related behaviors is also reviewed. As this review indicates, there is much work needed to provide more information about the molecular, cellular, circuit, and behavioral consequences of EtOH interactions with synaptic plasticity mechanisms.
Collapse
Affiliation(s)
- David M Lovinger
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland 20892, USA
| | - Karina P Abrahao
- Laboratory for Integrative Neuroscience, Division of Intramural Clinical and Biological Research, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland 20892, USA
| |
Collapse
|
78
|
Berthoux C, Barre A, Bockaert J, Marin P, Bécamel C. Sustained Activation of Postsynaptic 5-HT2A Receptors Gates Plasticity at Prefrontal Cortex Synapses. Cereb Cortex 2018; 29:1659-1669. [DOI: 10.1093/cercor/bhy064] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/26/2018] [Indexed: 01/01/2023] Open
Affiliation(s)
- Coralie Berthoux
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Montpellier, Montpellier, France
| | - Alexander Barre
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Montpellier, Montpellier, France
| | - Joël Bockaert
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Montpellier, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Montpellier, Montpellier, France
| | - Carine Bécamel
- Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Montpellier, Montpellier, France
| |
Collapse
|
79
|
Gajardo I, Salazar CS, Lopez-Espíndola D, Estay C, Flores-Muñoz C, Elgueta C, Gonzalez-Jamett AM, Martínez AD, Muñoz P, Ardiles ÁO. Lack of Pannexin 1 Alters Synaptic GluN2 Subunit Composition and Spatial Reversal Learning in Mice. Front Mol Neurosci 2018; 11:114. [PMID: 29692709 PMCID: PMC5902501 DOI: 10.3389/fnmol.2018.00114] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/22/2018] [Indexed: 01/24/2023] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are two forms of synaptic plasticity that have been considered as the cellular substrate of memory formation. Although LTP has received considerable more attention, recent evidences indicate that LTD plays also important roles in the acquisition and storage of novel information in the brain. Pannexin 1 (Panx1) is a membrane protein that forms non-selective channels which have been shown to modulate the induction of hippocampal synaptic plasticity. Animals lacking Panx1 or blockade of Pannexin 1 channels precludes the induction of LTD and facilitates LTP. To evaluate if the absence of Panx1 also affects the acquisition of rapidly changing information we trained Panx1 knockout (KO) mice and wild type (WT) littermates in a visual and hidden version of the Morris water maze (MWM). We found that KO mice find the hidden platform similarly although slightly quicker than WT animals, nonetheless, when the hidden platform was located in the opposite quadrant (OQ) to the previous learned location, KO mice spent significantly more time in the previous quadrant than in the new location indicating that the absence of Panx1 affects the reversion of a previously acquired spatial memory. Consistently, we observed changes in the content of synaptic proteins critical to LTD, such as GluN2 subunits of N-methyl-D-aspartate receptors (NMDARs), which changed their contribution to synaptic plasticity in conditions of Panx1 ablation. Our findings give further support to the role of Panx1 channels on the modulation of synaptic plasticity induction, learning and memory processes.
Collapse
Affiliation(s)
- Ivana Gajardo
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudia S Salazar
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniela Lopez-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigaciones Biomédicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Estay
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudio Elgueta
- Institute for Physiology I, University of Freiburg, Freiburg, Germany
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Muñoz
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigaciones Biomédicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Center for Applied Neurological Sciences, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile
| | - Álvaro O Ardiles
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| |
Collapse
|
80
|
Wang W, Duclot F, Groveman BR, Carrier N, Qiao H, Fang XQ, Wang H, Xin W, Jiang XH, Salter MW, Ding XS, Kabbaj M, Yu XM. Hippocampal protein kinase D1 is necessary for DHPG-induced learning and memory impairments in rats. PLoS One 2018; 13:e0195095. [PMID: 29614089 PMCID: PMC5882104 DOI: 10.1371/journal.pone.0195095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 03/18/2018] [Indexed: 11/18/2022] Open
Abstract
Background Understanding molecular mechanisms underlying the induction of learning and memory impairments remains a challenge. Recent investigations have shown that the activation of group I mGluRs (mGluR1 and mGluR5) in cultured hippocampal neurons by application of (S)-3,5-Dihydroxyphenylglycine (DHPG) causes the regulated internalization of N-methyl-D-aspartate receptors (NMDARs), which subsequently activates protein kinase D1 (PKD1). Through phosphorylating the C-terminals of the NMDAR GluN2 subunits, PKD1 down-regulates the activity of remaining (non-internalized) surface NMDARs. The knockdown of PKD1 does not affect the DHPG-induced inhibition of AMPA receptor-mediated miniature excitatory post-synaptic currents (mEPSCs) but prevents the DHPG-induced inhibition of NMDAR-mediated mEPSCs in vitro. Thus, we investigated the in vivo effects of bilateral infusions of DHPG into the hippocampal CA1 area of rats in the Morris water maze (MWM) and the novel object discrimination (NOD) tests. Methods A total of 300 adult male Sprague Dawley rats (250–280 g) were used for behavioral tests. One hundred ninety four were used in MWM test and the other 106 rats in the NOD test. Following one week of habituation to the vivarium, rats were bilaterally implanted under deep anesthesia with cannulas aimed at the CA1 area of the hippocampus (CA1 coordinates in mm from Bregma: AP -3.14; lateral +/-2; DV -3.0). Through implanted cannulas artificial cerebrospinal fluid (ACSF), the group1 mGluR antagonist 6-Methyl-2-(phenylethynyl)pyridine (MPEP), the dynamin-dependent internalization inhibitor Dynasore, or the PKD1 inhibitor CID755673 were infused into the bilateral hippocampal CA1 areas (2 μL per side, over 5 min). The effects of these infusions and the effects of PKD1 knockdown were examined in MWM or NOD test. Results DHPG infusion increased the latency to reach the platform in the MWM test and reduced the preference for the novel object in the NOD task. We found that the DHPG effects were dose-dependent and could be maintained for up to 2 days. Notably, these effects could be prevented by pre-infusion of the group1 mGluR antagonist MPEP, the dynamin-dependent internalization inhibitor Dynasore, the PKD1 inhibitor CID755673, or by PKD1 knockdown in the hippocampal CA1 area. Conclusion Altogether, these findings provide direct evidence that PKD1-mediated signaling may play a critical role in the induction of learning and memory impairments by DHPG infusion into the hippocampal CA1 area.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
- BenQ Affiliated Hospital and Neurological Institute, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Florian Duclot
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
| | - Bradley R. Groveman
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
| | - Nicole Carrier
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
| | - Haifa Qiao
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
| | - Xiao-Qian Fang
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
- Department of Biomedical Sciences, University of Texas Rio Grande Valley School of Medicine, Edinburg, Texas, United States of America
| | - Hui Wang
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
| | - Wenkuan Xin
- College of Pharmaceutical Sciences, Southwest University, Chongqing, People’s Republic of China
| | - Xing-Hong Jiang
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, People’s Republic of China
| | - Michael W. Salter
- Program in Neuroscience and Mental Health, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Xin-Sheng Ding
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
- BenQ Affiliated Hospital and Neurological Institute, Nanjing Medical University, Nanjing, People’s Republic of China
- * E-mail: (XD); (MK); (XMY)
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
- * E-mail: (XD); (MK); (XMY)
| | - Xian-Min Yu
- BenQ Affiliated Hospital and Neurological Institute, Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, United States of America
- Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, People’s Republic of China
- * E-mail: (XD); (MK); (XMY)
| |
Collapse
|
81
|
Sun DG, Kang H, Tetteh H, Su J, Lee J, Park SW, He J, Jo J, Yang S, Yang S. Long term potentiation, but not depression, in interlamellar hippocampus CA1. Sci Rep 2018; 8:5187. [PMID: 29581468 PMCID: PMC5979950 DOI: 10.1038/s41598-018-23369-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/12/2018] [Indexed: 01/23/2023] Open
Abstract
Synaptic plasticity in the lamellar CA3 to CA1 circuitry has been extensively studied while interlamellar CA1 to CA1 connections have not yet received much attention. One of our earlier studies demonstrated that axons of CA1 pyramidal neurons project to neighboring CA1 neurons, implicating information transfer along a longitudinal interlamellar network. Still, it remains unclear whether long-term synaptic plasticity is present within this longitudinal CA1 network. Here, we investigate long-term synaptic plasticity between CA1 pyramidal cells, using in vitro and in vivo extracellular recordings and 3D holography glutamate uncaging. We found that the CA1-CA1 network exhibits NMDA receptor-dependent long-term potentiation (LTP) without direction or layer selectivity. By contrast, we find no significant long-term depression (LTD) under various LTD induction protocols. These results implicate unique synaptic properties in the longitudinal projection suggesting that the interlamellar CA1 network could be a promising structure for hippocampus-related information processing and brain diseases.
Collapse
Affiliation(s)
- Duk-Gyu Sun
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, Korea
| | - Hyeri Kang
- Department of Nano-bioengineering, Incheon National University, Incheon, Korea
| | - Hannah Tetteh
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Junfeng Su
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Jihwan Lee
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Sung-Won Park
- Department of Nano-bioengineering, Incheon National University, Incheon, Korea
| | - Jufang He
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Jihoon Jo
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, Korea. .,Department of Neurology, Chonnam National University Medical School, Gwangju, Korea. .,NeuroMedical Convergence Laboratory, Biomedical Research Institute, Chonnam National University Hospital, Gwangju, Korea.
| | - Sungchil Yang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong.
| | - Sunggu Yang
- Department of Nano-bioengineering, Incheon National University, Incheon, Korea.
| |
Collapse
|
82
|
Edelmann E, Lessmann V. Dopaminergic innervation and modulation of hippocampal networks. Cell Tissue Res 2018; 373:711-727. [PMID: 29470647 DOI: 10.1007/s00441-018-2800-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
The catecholamine dopamine plays an important role in hippocampus-dependent plasticity and related learning and memory processes. Dopamine secretion in the hippocampus is activated by, e.g., salient or novel stimuli, thereby helping to establish and to stabilize hippocampus-dependent memories. Disturbed dopaminergic function in the hippocampus leads to severe pathophysiological conditions. While the role and importance of dopaminergic modulation of hippocampal networks have been unequivocally proven, there is still a lack of detailed molecular and cellular mechanistic understanding of how dopamine orchestrates these hippocampal processes. In this chapter of the special issue "Hippocampal structure and function," we will discuss the current understanding of dopaminergic modulation of basal synaptic transmission and long-lasting, activity-dependent potentiation or depression.
Collapse
Affiliation(s)
- Elke Edelmann
- Institut für Physiologie, Otto-von-Guericke-Universität, Medizinische Fakultät, Leipziger Str. 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto-von-Guericke University, Universitätsplatz 2, 39106, Magdeburg, Germany.
| | - Volkmar Lessmann
- Institut für Physiologie, Otto-von-Guericke-Universität, Medizinische Fakultät, Leipziger Str. 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto-von-Guericke University, Universitätsplatz 2, 39106, Magdeburg, Germany.
| |
Collapse
|
83
|
Hoang TH, Aliane V, Manahan-Vaughan D. Novel encoding and updating of positional, or directional, spatial cues are processed by distinct hippocampal subfields: Evidence for parallel information processing and the "what" stream. Hippocampus 2018; 28:315-326. [PMID: 29394518 PMCID: PMC5947642 DOI: 10.1002/hipo.22833] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 11/12/2017] [Accepted: 01/23/2018] [Indexed: 11/08/2022]
Abstract
The specific roles of hippocampal subfields in spatial information processing and encoding are, as yet, unclear. The parallel map theory postulates that whereas the CA1 processes discrete environmental features (positional cues used to generate a “sketch map”), the dentate gyrus (DG) processes large navigation‐relevant landmarks (directional cues used to generate a “bearing map”). Additionally, the two‐streams hypothesis suggests that hippocampal subfields engage in differentiated processing of information from the “where” and the “what” streams. We investigated these hypotheses by analyzing the effect of exploration of discrete “positional” features and large “directional” spatial landmarks on hippocampal neuronal activity in rats. As an indicator of neuronal activity we measured the mRNA induction of the immediate early genes (IEGs), Arc and Homer1a. We observed an increase of this IEG mRNA in CA1 neurons of the distal neuronal compartment and in proximal CA3, after novel spatial exploration of discrete positional cues, whereas novel exploration of directional cues led to increases in IEG mRNA in the lower blade of the DG and in proximal CA3. Strikingly, the CA1 did not respond to directional cues and the DG did not respond to positional cues. Our data provide evidence for both the parallel map theory and the two‐streams hypothesis and suggest a precise compartmentalization of the encoding and processing of “what” and “where” information occurs within the hippocampal subfields.
Collapse
Affiliation(s)
- Thu-Huong Hoang
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum 44780, Germany.,International Graduate School of Neuroscience, Ruhr University Bochum, Bochum 44780, Germany
| | - Verena Aliane
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum 44780, Germany
| | - Denise Manahan-Vaughan
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum 44780, Germany
| |
Collapse
|
84
|
Recording Field Potentials and Synaptic Plasticity From Freely Behaving Rodents. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2018. [DOI: 10.1016/b978-0-12-812028-6.00001-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
85
|
Item-Place Encoding Through Hippocampal Long-Term Depression. HANDBOOK OF OBJECT NOVELTY RECOGNITION 2018. [DOI: 10.1016/b978-0-12-812012-5.00019-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
86
|
Manahan-Vaughan D. Special Considerations When Using Mice for In Vivo Electrophysiology and Long-Term Studies of Hippocampal Synaptic Plasticity During Behavior. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2018. [DOI: 10.1016/b978-0-12-812028-6.00003-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
87
|
Long Term Depression in Rat Hippocampus and the Effect of Ethanol during Fetal Life. Brain Sci 2017; 7:brainsci7120157. [PMID: 29182556 PMCID: PMC5742760 DOI: 10.3390/brainsci7120157] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/20/2017] [Accepted: 11/27/2017] [Indexed: 12/11/2022] Open
Abstract
Alcohol (ethanol) disturbs cognitive functions including learning and memory in humans, non-human primates, and laboratory animals such as rodents. As studied in animals, cellular mechanisms for learning and memory include bidirectional synaptic plasticity, long-term potentiation (LTP), and long-term depression (LTD), primarily in the hippocampus. Most of the research in the field of alcohol has analyzed the effects of ethanol on LTP; however, with recent advances in the understanding of the physiological role of LTD in learning and memory, some authors have examined the effects of ethanol exposure on this particular signal. In the present review, I will focus on hippocampal LTD recorded in rodents and the effects of fetal alcohol exposure on this signal. A synthesis of the findings indicates that prenatal ethanol exposure disturbs LTD concurrently with LTP in offspring and that both glutamatergic and γ-aminobutyric acid (GABA) neurotransmissions are altered and contribute to LTD disturbances. Although the ultimate mode of action of ethanol on these two transmitter systems is not yet clear, novel suggestions have recently appeared in the literature.
Collapse
|
88
|
Berger SM, Fernández-Lamo I, Schönig K, Fernández Moya SM, Ehses J, Schieweck R, Clementi S, Enkel T, Grothe S, von Bohlen Und Halbach O, Segura I, Delgado-García JM, Gruart A, Kiebler MA, Bartsch D. Forebrain-specific, conditional silencing of Staufen2 alters synaptic plasticity, learning, and memory in rats. Genome Biol 2017; 18:222. [PMID: 29149906 PMCID: PMC5693596 DOI: 10.1186/s13059-017-1350-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/26/2017] [Indexed: 12/16/2022] Open
Abstract
Background Dendritic messenger RNA (mRNA) localization and subsequent local translation in dendrites critically contributes to synaptic plasticity and learning and memory. Little is known, however, about the contribution of RNA-binding proteins (RBPs) to these processes in vivo. Results To delineate the role of the double-stranded RBP Staufen2 (Stau2), we generate a transgenic rat model, in which Stau2 expression is conditionally silenced by Cre-inducible expression of a microRNA (miRNA) targeting Stau2 mRNA in adult forebrain neurons. Known physiological mRNA targets for Stau2, such as RhoA, Complexin 1, and Rgs4 mRNAs, are found to be dysregulated in brains of Stau2-deficient rats. In vivo electrophysiological recordings reveal synaptic strengthening upon stimulation, showing a shift in the frequency-response function of hippocampal synaptic plasticity to favor long-term potentiation and impair long-term depression in Stau2-deficient rats. These observations are accompanied by deficits in hippocampal spatial working memory, spatial novelty detection, and in tasks investigating associative learning and memory. Conclusions Together, these experiments reveal a critical contribution of Stau2 to various forms of synaptic plasticity including spatial working memory and cognitive management of new environmental information. These findings might contribute to the development of treatments for conditions associated with learning and memory deficits. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1350-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefan M Berger
- Department of Molecular Biology, CIMH and Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Iván Fernández-Lamo
- Division of Neurosciences, Pablo de Olavide University, 41013, Seville, Spain.,Present Address: Institute Cajal (CSIC), 28002, Madrid, Spain
| | - Kai Schönig
- Department of Molecular Biology, CIMH and Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Sandra M Fernández Moya
- BioMedical Center, Medical Faculty, Ludwig Maximilians University, 82152, Planegg-Martinsried, Germany
| | - Janina Ehses
- BioMedical Center, Medical Faculty, Ludwig Maximilians University, 82152, Planegg-Martinsried, Germany
| | - Rico Schieweck
- BioMedical Center, Medical Faculty, Ludwig Maximilians University, 82152, Planegg-Martinsried, Germany
| | - Stefano Clementi
- Department of Molecular Biology, CIMH and Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Thomas Enkel
- Department of Molecular Biology, CIMH and Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Sascha Grothe
- Institute for Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany
| | | | - Inmaculada Segura
- BioMedical Center, Medical Faculty, Ludwig Maximilians University, 82152, Planegg-Martinsried, Germany.
| | | | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, 41013, Seville, Spain
| | - Michael A Kiebler
- BioMedical Center, Medical Faculty, Ludwig Maximilians University, 82152, Planegg-Martinsried, Germany.
| | - Dusan Bartsch
- Department of Molecular Biology, CIMH and Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany.
| |
Collapse
|
89
|
Aidil-Carvalho M, Carmo A, Ribeiro J, Cunha-Reis D. Mismatch novelty exploration training enhances hippocampal synaptic plasticity: A tool for cognitive stimulation? Neurobiol Learn Mem 2017; 145:240-250. [DOI: 10.1016/j.nlm.2017.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 07/31/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023]
|
90
|
Wilkerson JR, Albanesi JP, Huber KM. Roles for Arc in metabotropic glutamate receptor-dependent LTD and synapse elimination: Implications in health and disease. Semin Cell Dev Biol 2017; 77:51-62. [PMID: 28969983 DOI: 10.1016/j.semcdb.2017.09.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 10/18/2022]
Abstract
The Arc gene is robustly transcribed in specific neural ensembles in response to experience-driven activity. Upon induction, Arc mRNA is transported to dendrites, where it can be rapidly and locally translated by activation of metabotropic glutamate receptors (mGluR1/5). mGluR-induced dendritic synthesis of Arc is implicated in weakening or elimination of excitatory synapses by triggering endocytosis of postsynaptic AMPARs in both hippocampal CA1 and cerebellar Purkinje neurons. Importantly, CA1 neurons with experience-induced Arc mRNA are susceptible, or primed for mGluR-induced long-term synaptic depression (mGluR-LTD). Here we review mechanisms and function of Arc in mGluR-LTD and synapse elimination and propose roles for these forms of plasticity in Arc-dependent formation of sparse neural representations of learned experience. We also discuss accumulating evidence linking dysregulation of Arc and mGluR-LTD in human cognitive disorders such as intellectual disability, autism and Alzheimer's disease.
Collapse
Affiliation(s)
- Julia R Wilkerson
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Joseph P Albanesi
- Departments of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Kimberly M Huber
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
91
|
Abstract
Progress in clinical and affective neuroscience is redefining psychiatric illness as symptomatic expression of cellular/molecular dysfunctions in specific brain circuits. Post-traumatic stress disorder (PTSD) has been an exemplar of this progress, with improved understanding of neurobiological systems subserving fear learning, salience detection, and emotion regulation explaining much of its phenomenology and neurobiology. However, many features remain unexplained and a parsimonious model that more fully accounts for symptoms and the core neurobiology remains elusive. Contextual processing is a key modulatory function of hippocampal-prefrontal-thalamic circuitry, allowing organisms to disambiguate cues and derive situation-specific meaning from the world. We propose that dysregulation within this context-processing circuit is at the core of PTSD pathophysiology, accounting for much of its phenomenology and most of its biological findings. Understanding core mechanisms like this, and their underlying neural circuits, will sharpen diagnostic precision and understanding of risk factors, enhancing our ability to develop preventive and "personalized" interventions.
Collapse
Affiliation(s)
- Israel Liberzon
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-2700, USA; Mental Health Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, MI 48105, USA.
| | - James L Abelson
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-2700, USA
| |
Collapse
|
92
|
Kim DH, Kang M, Kim CH, Huh YH, Cho IH, Ryu HH, Chung KH, Park CS, Rhee S, Lee YS, Song WK. SPIN90 Modulates Long-Term Depression and Behavioral Flexibility in the Hippocampus. Front Mol Neurosci 2017; 10:295. [PMID: 28979184 PMCID: PMC5611360 DOI: 10.3389/fnmol.2017.00295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 09/01/2017] [Indexed: 12/23/2022] Open
Abstract
The importance of actin-binding proteins (ABPs) in the regulation of synapse morphology and plasticity has been well established. SH3 protein interacting with Nck, 90 kDa (SPIN90), an Nck-interacting protein highly expressed in synapses, is essential for actin remodeling and dendritic spine morphology. Synaptic targeting of SPIN90 to spine heads or dendritic shafts depends on its phosphorylation state, leading to blockage of cofilin-mediated actin depolymerization and spine shrinkage. However, the physiological role of SPIN90 in long-term plasticity, learning and memory are largely unknown. In this study, we demonstrate that Spin90-knockout (KO) mice exhibit substantial deficits in synaptic plasticity and behavioral flexibility. We found that loss of SPIN90 disrupted dendritic spine density in CA1 neurons of the hippocampus and significantly impaired long-term depression (LTD), leaving basal synaptic transmission and long-term potentiation (LTP) intact. These impairments were due in part to deficits in AMPA receptor endocytosis and its pre-requisites, GluA1 dephosphorylation and postsynaptic density (PSD) 95 phosphorylation, but also by an intrinsic activation of Akt-GSK3β signaling as a result of Spin90-KO. In accordance with these defects, mice lacking SPIN90 were found to carry significant deficits in object-recognition and behavioral flexibility, while learning ability was largely unaffected. Collectively, these findings demonstrate a novel modulatory role for SPIN90 in hippocampal LTD and behavioral flexibility.
Collapse
Affiliation(s)
- Dae Hwan Kim
- Bio Imaging and Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - Minkyung Kang
- Department of Physiology, Department of Biomedical Sciences, Seoul National University College of MedicineSeoul, South Korea
| | - Chong-Hyun Kim
- Center for Neuroscience, Korea Institute of Science and Technology, Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and TechnologySeoul, South Korea
| | - Yun Hyun Huh
- Bio Imaging and Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - In Ha Cho
- Department of Biological Sciences, Dartmouth CollegeHanover, NH, United States
| | - Hyun-Hee Ryu
- Department of Physiology, Department of Biomedical Sciences, Seoul National University College of MedicineSeoul, South Korea.,Department of Life Science, Chung-Ang UniversitySeoul, South Korea
| | - Kyung Hwun Chung
- Electron Microscope Facility, Dental Research Institute, Seoul National UniversitySeoul, South Korea
| | - Chul-Seung Park
- School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang UniversitySeoul, South Korea
| | - Yong-Seok Lee
- Department of Physiology, Department of Biomedical Sciences, Seoul National University College of MedicineSeoul, South Korea
| | - Woo Keun Song
- Bio Imaging and Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and TechnologyGwangju, South Korea
| |
Collapse
|
93
|
Esmaeilpour K, Sheibani V, Shabani M, Mirnajafi-Zadeh J. Low frequency electrical stimulation has time dependent improving effect on kindling-induced impairment in long-term potentiation in rats. Brain Res 2017; 1668:20-27. [DOI: 10.1016/j.brainres.2017.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 04/28/2017] [Accepted: 05/08/2017] [Indexed: 10/19/2022]
|
94
|
Abstract
It is possible that one of the essential functions of sleep is to take out the garbage, as it were, erasing and "forgetting" information built up throughout the day that would clutter the synaptic network that defines us. It may also be that this cleanup function of sleep is a general principle of neuroscience, applicable to every creature with a nervous system.
Collapse
|
95
|
The Longevity of Hippocampus-Dependent Memory Is Orchestrated by the Locus Coeruleus-Noradrenergic System. Neural Plast 2017; 2017:2727602. [PMID: 28695015 PMCID: PMC5485371 DOI: 10.1155/2017/2727602] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/17/2017] [Accepted: 05/23/2017] [Indexed: 12/24/2022] Open
Abstract
The locus coeruleus is connected to the dorsal hippocampus via strong fiber projections. It becomes activated after arousal and novelty, whereupon noradrenaline is released in the hippocampus. Noradrenaline from the locus coeruleus is involved in modulating the encoding, consolidation, retrieval, and reversal of hippocampus-based memory. Memory storage can be modified by the activation of the locus coeruleus and subsequent facilitation of hippocampal long-term plasticity in the forms of long-term depression and long-term potentiation. Recent evidence indicates that noradrenaline and dopamine are coreleased in the hippocampus from locus coeruleus terminals, thus fostering neuromodulation of long-term synaptic plasticity and memory. Noradrenaline is an inductor of epigenetic modifications regulating transcriptional control of synaptic long-term plasticity to gate the endurance of memory storage. In conclusion, locus coeruleus activation primes the persistence of hippocampus-based long-term memory.
Collapse
|
96
|
Jansen S, Gottschling C, Faissner A, Manahan-Vaughan D. Intrinsic cellular and molecular properties of in vivo hippocampal synaptic plasticity are altered in the absence of key synaptic matrix molecules. Hippocampus 2017; 27:920-933. [DOI: 10.1002/hipo.22742] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 04/19/2017] [Accepted: 05/12/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Stephan Jansen
- Department of Neurophysiology, Medical Faculty; Ruhr University Bochum; Bochum Germany
| | - Christine Gottschling
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology; Ruhr University Bochum; Bochum Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology; Ruhr University Bochum; Bochum Germany
| | | |
Collapse
|
97
|
Raven F, Van der Zee EA, Meerlo P, Havekes R. The role of sleep in regulating structural plasticity and synaptic strength: Implications for memory and cognitive function. Sleep Med Rev 2017. [PMID: 28641933 DOI: 10.1016/j.smrv.2017.05.002] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dendritic spines are the major sites of synaptic transmission in the central nervous system. Alterations in the strength of synaptic connections directly affect the neuronal communication, which is crucial for brain function as well as the processing and storage of information. Sleep and sleep loss bidirectionally alter structural plasticity, by affecting spine numbers and morphology, which ultimately can affect the functional output of the brain in terms of alertness, cognition, and mood. Experimental data from studies in rodents suggest that sleep deprivation may impact structural plasticity in different ways. One of the current views, referred to as the synaptic homeostasis hypothesis, suggests that wake promotes synaptic potentiation whereas sleep facilitates synaptic downscaling. On the other hand, several studies have now shown that sleep deprivation can reduce spine density and attenuate synaptic efficacy in the hippocampus. These data are the basis for the view that sleep promotes hippocampal structural plasticity critical for memory formation. Altogether, the impact of sleep and sleep loss may vary between regions of the brain. A better understanding of the role that sleep plays in regulating structural plasticity may ultimately lead to novel therapeutic approaches for brain disorders that are accompanied by sleep disturbances and sleep loss.
Collapse
Affiliation(s)
- Frank Raven
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Eddy A Van der Zee
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Peter Meerlo
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Robbert Havekes
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
98
|
Kalweit AN, Amanpour-Gharaei B, Colitti-Klausnitzer J, Manahan-Vaughan D. Changes in Neuronal Oscillations Accompany the Loss of Hippocampal LTP that Occurs in an Animal Model of Psychosis. Front Behav Neurosci 2017; 11:36. [PMID: 28337131 PMCID: PMC5340772 DOI: 10.3389/fnbeh.2017.00036] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/21/2017] [Indexed: 12/17/2022] Open
Abstract
The first-episode of psychosis is followed by a transient time-window of ca. 60 days during which therapeutic interventions have a higher likelihood of being effective than interventions that are started with a greater latency. This suggests that, in the immediate time-period after first-episode psychosis, functional changes occur in the brain that render it increasingly resistant to intervention. The precise mechanistic nature of these changes is unclear, but at the cognitive level, sensory and hippocampus-based dysfunctions become increasingly manifest. In an animal model of first-episode psychosis that comprises acute treatment of rats with the irreversible N-methyl-D-aspartate receptor (NMDAR)-antagonist, MK801, acute but also chronic deficits in long-term potentiation (LTP) and spatial memory occur. Neuronal oscillations, especially in the form of information transfer through θ and γ frequency oscillations are an intrinsic component of normal information processing in the hippocampus. Changes in θ-γ coupling and power are known to accompany deficits in hippocampal plasticity. Here, we examined whether changes in δ, θ, α, β and γ oscillations, or θ-γ coupling accompany the chronic loss of LTP that is observed in the MK801-animal model of psychosis. One and 4 weeks after acute systemic treatment of adult rats with MK801, a potent loss of hippocampal in vivo LTP was evident compared to vehicle-treated controls. Overall, the typical pattern of θ-γ oscillations that are characteristic for the successful induction of LTP was altered. In particular, θ-power was lower and an uncoupling of θ-γ oscillations was evident in MK801-treated rats. The alterations in network oscillations that accompany LTP deficits in this animal model may comprise a mechanism through which disturbances in sensory information processing and hippocampal function occur in psychosis. These data suggest that the hippocampus is likely to comprise a very early locus of functional change after instigation of a first-episode psychosis-like state in rodents.
Collapse
Affiliation(s)
- Alexander N Kalweit
- Department of Neurophysiology, Medical Faculty, Ruhr University BochumBochum, Germany; International Graduate School of Neuroscience, Ruhr University BochumBochum, Germany
| | - Bezhad Amanpour-Gharaei
- Department of Neurophysiology, Medical Faculty, Ruhr University BochumBochum, Germany; International Graduate School of Neuroscience, Ruhr University BochumBochum, Germany
| | | | | |
Collapse
|
99
|
Meunier CNJ, Chameau P, Fossier PM. Modulation of Synaptic Plasticity in the Cortex Needs to Understand All the Players. Front Synaptic Neurosci 2017; 9:2. [PMID: 28203201 PMCID: PMC5285384 DOI: 10.3389/fnsyn.2017.00002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/13/2017] [Indexed: 12/19/2022] Open
Abstract
The prefrontal cortex (PFC) is involved in cognitive tasks such as working memory, decision making, risk assessment and regulation of attention. These functions performed by the PFC are supposed to rely on rhythmic electrical activity generated by neuronal network oscillations determined by a precise balance between excitation and inhibition balance (E/I balance) resulting from the coordinated activities of recurrent excitation and feedback and feedforward inhibition. Functional alterations in PFC functions have been associated with cognitive deficits in several pathologies such as major depression, anxiety and schizophrenia. These pathological situations are correlated with alterations of different neurotransmitter systems (i.e., serotonin (5-HT), dopamine (DA), acetylcholine…) that result in alterations of the E/I balance. The aim of this review article is to cover the basic aspects of the regulation of the E/I balance as well as to highlight the importance of the complementarity role of several neurotransmitters in the modulation of the plasticity of excitatory and inhibitory synapses. We illustrate our purpose by recent findings that demonstrate that 5-HT and DA cooperate to regulate the plasticity of excitatory and inhibitory synapses targeting layer 5 pyramidal neurons (L5PyNs) of the PFC and to fine tune the E/I balance. Using a method based on the decomposition of the synaptic conductance into its excitatory and inhibitory components, we show that concomitant activation of D1-like receptors (D1Rs) and 5-HT1ARs, through a modulation of NMDA receptors, favors long term potentiation (LTP) of both excitation and inhibition and consequently does not modify the E/I balance. We also demonstrate that activation of D2-receptors requires functional 5-HT1ARs to shift the E-I balance towards more inhibition and to favor long term depression (LTD) of excitatory synapses through the activation of glycogen synthase kinase 3β (GSK3β). This cooperation between different neurotransmitters is particularly relevant in view of pathological situations in which alterations of one neurotransmitter system will also have consequences on the regulation of synaptic efficacy by other neurotransmitters. This opens up new perspectives in the development of therapeutic strategies for the pharmacological treatment of neuronal disorders.
Collapse
Affiliation(s)
- Claire N J Meunier
- Institut de Neurosciences Paris-Saclay (NeuroPSI), UMR 91197 CNRS-Université Paris-Saclay Paris, France
| | - Pascal Chameau
- Swammerdam Institute for Life Sciences, Center for NeuroScience, University of Amsterdam Amsterdam, Netherlands
| | - Philippe M Fossier
- Institut de Neurosciences Paris-Saclay (NeuroPSI), UMR 91197 CNRS-Université Paris-Saclay Paris, France
| |
Collapse
|
100
|
Liu C, Yin H, Gao J, Xu X, Zhang T, Yang Z. Leonurine ameliorates cognitive dysfunction via antagonizing excitotoxic glutamate insults and inhibiting autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1638-1646. [PMID: 27823628 DOI: 10.1016/j.phymed.2016.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/28/2016] [Accepted: 10/02/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Chronic cerebral hypoperfusion is related with cognitive deficits in different types of dementia. PURPOSE In this study, we aimed to investigate the effect and potential mechanisms of leonurine on chronic cerebral hypoperfusion both in vitro and in vivo. STUDY DESIGN Chronic cerebral hypoperfusion was duplicated by oxygen-glucose deprivation (OGD) in vitro and by ligation of bilateral common carotid arteries (2-VO) in vivo. METHODS In in vitro study, there were control group, OGD group, OGD+ 100µM leonurin group, and OGD+ 10µM donepezil group. The spontaneous excitatory postsynaptic current amplitude and frequency were recorded. In in vivo study, the chronic cerebral hypoperfusion model was induced by ligated bilateral common carotid arteries. Rats were randomly divided into Sham group, 2-VO group, 2-VO+ 60mg/kg/day leonurine group, and 2-VO+ 4mg/kg/day donepezil group. After three weeks, the Morris water maze and Long-term depression recording were observed. Then N-methyl-D-aspartate receptor-associated proteins and autophagy-associated proteins were detected by Western blot assay. RESULTS In in vitro experiment, results showed that leonurine could obviously attenuate the spontaneous excitatory postsynaptic current amplitude and frequency on pyramidal neurons. In in vivo experiment, leonurine significantly decreased levels of glutamate and hydrogen peroxide, improved both the cognitive flexibility and the spatial learning and memory abilities. Moreover, leonurine obviously enhanced long-term depression, elevated the ratio of N-methyl-D-aspartate receptor 2A/2B, and decreased the expression of postsynaptic density protein-95. Interestingly, the ratio of LC3II/LC3I and beclin-1 expression were markedly down-regulated by leonurine. CONCLUSION These findings suggest that leonurine ameliorates cognitive dysfunction at least partly via antagonizing excitotoxic glutamate insults and inhibiting autophagy. Furthermore, it might become a potential drug candidate of chronic cerebral hyperfusion in future.
Collapse
Affiliation(s)
- Chunhua Liu
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, China
| | - Hongqiang Yin
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, China
| | - Jing Gao
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, China
| | - Xiaxia Xu
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Tao Zhang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhuo Yang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|