51
|
Mellado S, Moreno-Ruiz B, Expósito S, Fernández M, Martín ED. Prolactin Reduces Hippocampal Parvalbumin and GABAA Receptor Expression in Female Mice. Neuroendocrinology 2022; 112:796-806. [PMID: 34666336 PMCID: PMC9533442 DOI: 10.1159/000520279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Parvalbumin (PV)-positive cells are strategic elements of neuronal networks capable of influencing memory and learning processes. However, it is not known whether pituitary hormones may be related to PV expression in the hippocampus - a part of the limbic system with important functions in learning and memory. OBJECTIVE Since previous studies indicate that prolactin (PRL) plays a significant role in hippocampal-dependent learning and synaptic plasticity, we hypothesized that a rise in PRL levels can modify PV expression in the hippocampus. METHODS We employed biochemical, immunohistochemistry, and densitometry techniques - as well as a behavioural assay - in a hyperprolactinemia model using subcutaneous osmotic pumps in female mice. RESULTS PRL treatment via osmotic pump induced an increase in PRL receptor (PRLR) expression in most regions of the hippocampus analysed by Western blotting and immunohistochemistry methods. Fluorescent densitometry analysis revealed that PV expression decreases in the same layers in the hippocampus following PRL treatment, while double labelling immunostaining indicated close localization of PV and PRLR in PV-positive interneurons. In addition, we found that PRL induced a reduction in the β2/3 subunit of GABAA receptor (GABAAR) expression that was linearly correlated with the reduction in PV expression. This reduction in the β2/3 subunit of GABAAR expression was maintained in trained animals in which PRL treatment improved the learning of a spatial memory task. CONCLUSIONS These data show, for the first time, that an increase in PRL level is associated with changes in key constituent elements of inhibitory circuits in the hippocampus and may be of relevance for the alterations in cognitive function reported in hyperprolactinemia.
Collapse
Affiliation(s)
- Susana Mellado
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Beatriz Moreno-Ruiz
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Sara Expósito
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Miriam Fernández
- Research Institute for Neurological Disabilities (IDINE), Medical School, University of Castilla-La Mancha, Albacete, Spain
| | - Eduardo D. Martín
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- *Eduardo D. Martín,
| |
Collapse
|
52
|
Park J, Khan S, Yun DH, Ku T, Villa KL, Lee JE, Zhang Q, Park J, Feng G, Nedivi E, Chung K. Epitope-preserving magnified analysis of proteome (eMAP). SCIENCE ADVANCES 2021; 7:eabf6589. [PMID: 34767453 PMCID: PMC8589305 DOI: 10.1126/sciadv.abf6589] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 09/24/2021] [Indexed: 05/28/2023]
Abstract
Synthetic tissue-hydrogel methods have enabled superresolution investigation of biological systems using diffraction-limited microscopy. However, chemical modification by fixatives can cause loss of antigenicity, limiting molecular interrogation of the tissue gel. Here, we present epitope-preserving magnified analysis of proteome (eMAP) that uses purely physical tissue-gel hybridization to minimize the loss of antigenicity while allowing permanent anchoring of biomolecules. We achieved success rates of 96% and 94% with synaptic antibodies for mouse and marmoset brains, respectively. Maximal preservation of antigenicity allows imaging of nanoscopic architectures in 1000-fold expanded tissues without additional signal amplification. eMAP-processed tissue gel can endure repeated staining and destaining without epitope loss or structural damage, enabling highly multiplexed proteomic analysis. We demonstrated the utility of eMAP as a nanoscopic proteomic interrogation tool by investigating molecular heterogeneity in inhibitory synapses in the mouse brain neocortex and characterizing the spatial distributions of synaptic proteins within synapses in mouse and marmoset brains.
Collapse
Affiliation(s)
- Joha Park
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Sarim Khan
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Chemical Engineering, Indian Institute of Technology (IIT), Roorkee, Uttarakhand 247667, India
| | - Dae Hee Yun
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Taeyun Ku
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Katherine L. Villa
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Jiachen E. Lee
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Qiangge Zhang
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Juhyuk Park
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Chemical Engineering, MIT, Cambridge, MA 02142, USA
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Guoping Feng
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Elly Nedivi
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Kwanghun Chung
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
- Department of Chemical Engineering, MIT, Cambridge, MA 02142, USA
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
53
|
Wu K, Castellano D, Tian Q, Lu W. Distinct regulation of tonic GABAergic inhibition by NMDA receptor subtypes. Cell Rep 2021; 37:109960. [PMID: 34758303 PMCID: PMC8630577 DOI: 10.1016/j.celrep.2021.109960] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/03/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022] Open
Abstract
Tonic inhibition mediated by extrasynaptic GABAARs regulates various brain functions. However, the mechanisms that regulate tonic inhibition remain largely unclear. Here, we report distinct actions of GluN2A- and GluN2B-NMDA receptors (NMDARs) on tonic inhibition in hippocampal neurons under basal and high activity conditions. Specifically, overexpression of GluN2B, but not GluN2A, reduces α5-GABAAR surface expression and tonic currents. Additionally, knockout of GluN2A and GluN2B decreases and increases tonic currents, respectively. Mechanistically, GluN2A-NMDARs inhibit and GluN2B-NMDARs promote α5-GABAAR internalization, resulting in increased and decreased surface α5-GABAAR expression, respectively. Furthermore, GluN2A-NMDARs, but not GluN2B-NMDARs, are required for homeostatic potentiation of tonic inhibition induced by prolonged increase of neuronal activity. Last, tonic inhibition decreases during acute seizures, whereas it increases 24 h later, involving GluN2-NMDAR-dependent signaling. Collectively, these data reveal an NMDAR subunit-specific regulation of tonic inhibition in physiological and pathological conditions and provide mechanistic insight into activity-dependent modulation of tonic inhibition.
Collapse
Affiliation(s)
- Kunwei Wu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Castellano
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qingjun Tian
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
54
|
Ahmadian Y, Miller KD. What is the dynamical regime of cerebral cortex? Neuron 2021; 109:3373-3391. [PMID: 34464597 PMCID: PMC9129095 DOI: 10.1016/j.neuron.2021.07.031] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 07/05/2021] [Accepted: 07/30/2021] [Indexed: 01/13/2023]
Abstract
Many studies have shown that the excitation and inhibition received by cortical neurons remain roughly balanced across many conditions. A key question for understanding the dynamical regime of cortex is the nature of this balancing. Theorists have shown that network dynamics can yield systematic cancellation of most of a neuron's excitatory input by inhibition. We review a wide range of evidence pointing to this cancellation occurring in a regime in which the balance is loose, meaning that the net input remaining after cancellation of excitation and inhibition is comparable in size with the factors that cancel, rather than tight, meaning that the net input is very small relative to the canceling factors. This choice of regime has important implications for cortical functional responses, as we describe: loose balance, but not tight balance, can yield many nonlinear population behaviors seen in sensory cortical neurons, allow the presence of correlated variability, and yield decrease of that variability with increasing external stimulus drive as observed across multiple cortical areas.
Collapse
Affiliation(s)
- Yashar Ahmadian
- Computational and Biological Learning Lab, Department of Engineering, University of Cambridge, Cambridge, UK.
| | - Kenneth D Miller
- Center for Theoretical Neuroscience, Swartz Program in Theoretical Neuroscience, Kavli Institute for Brain Science, and Department of Neuroscience, College of Physicians and Surgeons and Morton B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
55
|
Maksymetz J, Byun NE, Luessen DJ, Li B, Barry RL, Gore JC, Niswender CM, Lindsley CW, Joffe ME, Conn PJ. mGlu 1 potentiation enhances prelimbic somatostatin interneuron activity to rescue schizophrenia-like physiological and cognitive deficits. Cell Rep 2021; 37:109950. [PMID: 34731619 PMCID: PMC8628371 DOI: 10.1016/j.celrep.2021.109950] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 08/09/2021] [Accepted: 10/14/2021] [Indexed: 01/03/2023] Open
Abstract
Evidence for prefrontal cortical (PFC) GABAergic dysfunction is one of the most consistent findings in schizophrenia and may contribute to cognitive deficits. Recent studies suggest that the mGlu1 subtype of metabotropic glutamate receptor regulates cortical inhibition; however, understanding the mechanisms through which mGlu1 positive allosteric modulators (PAMs) regulate PFC microcircuit function and cognition is essential for advancing these potential therapeutics toward the clinic. We report a series of electrophysiology, optogenetic, pharmacological magnetic resonance imaging, and animal behavior studies demonstrating that activation of mGlu1 receptors increases inhibitory transmission in the prelimbic PFC by selective excitation of somatostatin-expressing interneurons (SST-INs). An mGlu1 PAM reverses cortical hyperactivity and concomitant cognitive deficits induced by N-methyl-d-aspartate (NMDA) receptor antagonists. Using in vivo optogenetics, we show that prelimbic SST-INs are necessary for mGlu1 PAM efficacy. Collectively, these findings suggest that mGlu1 PAMs could reverse cortical GABAergic deficits and exhibit efficacy in treating cognitive dysfunction in schizophrenia.
Collapse
Affiliation(s)
- James Maksymetz
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Nellie E Byun
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Deborah J Luessen
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Brianna Li
- Vanderbilt University, Nashville, TN 37232, USA
| | - Robert L Barry
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Max E Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
56
|
Melzer S, Newmark ER, Mizuno GO, Hyun M, Philson AC, Quiroli E, Righetti B, Gregory MR, Huang KW, Levasseur J, Tian L, Sabatini BL. Bombesin-like peptide recruits disinhibitory cortical circuits and enhances fear memories. Cell 2021; 184:5622-5634.e25. [PMID: 34610277 PMCID: PMC8556345 DOI: 10.1016/j.cell.2021.09.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 07/12/2021] [Accepted: 09/08/2021] [Indexed: 01/09/2023]
Abstract
Disinhibitory neurons throughout the mammalian cortex are powerful enhancers of circuit excitability and plasticity. The differential expression of neuropeptide receptors in disinhibitory, inhibitory, and excitatory neurons suggests that each circuit motif may be controlled by distinct neuropeptidergic systems. Here, we reveal that a bombesin-like neuropeptide, gastrin-releasing peptide (GRP), recruits disinhibitory cortical microcircuits through selective targeting and activation of vasoactive intestinal peptide (VIP)-expressing cells. Using a genetically encoded GRP sensor, optogenetic anterograde stimulation, and trans-synaptic tracing, we reveal that GRP regulates VIP cells most likely via extrasynaptic diffusion from several local and long-range sources. In vivo photometry and CRISPR-Cas9-mediated knockout of the GRP receptor (GRPR) in auditory cortex indicate that VIP cells are strongly recruited by novel sounds and aversive shocks, and GRP-GRPR signaling enhances auditory fear memories. Our data establish peptidergic recruitment of selective disinhibitory cortical microcircuits as a mechanism to regulate fear memories.
Collapse
Affiliation(s)
- Sarah Melzer
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Elena R Newmark
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Grace Or Mizuno
- Departments of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, USA
| | - Minsuk Hyun
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Adrienne C Philson
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Eleonora Quiroli
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Beatrice Righetti
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Malika R Gregory
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Kee Wui Huang
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - James Levasseur
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Lin Tian
- Departments of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, USA
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
57
|
Liu D, Liang M, Zhu L, Zhou TT, Wang Y, Wang R, Wu FF, Goh ELK, Chen T. Potential Ago2/miR-3068-5p Cascades in the Nucleus Accumbens Contribute to Methamphetamine-Induced Locomotor Sensitization of Mice. Front Pharmacol 2021; 12:708034. [PMID: 34483916 PMCID: PMC8414410 DOI: 10.3389/fphar.2021.708034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/12/2021] [Indexed: 01/24/2023] Open
Abstract
Dysregulation of microRNA (miRNA) biogenesis is involved in drug addiction. Argonaute2 (Ago2), a specific splicing protein involved in the generation of miRNA, was found to be dysregulated in the nucleus accumbens (NAc) of methamphetamine (METH)-sensitized mice in our previous study. Here, we determined whether Ago2 in the NAc regulates METH sensitization in mice and identified Ago2-dependent miRNAs involved in this process. We found a gradual reduction in Ago2 expression in the NAc following repeated METH use. METH-induced hyperlocomotor activity in mice was strengthened by knocking down NAc neuronal levels of Ago2 but reduced by overexpressing Ago2 in NAc neurons. Surprisingly, miR-3068-5p was upregulated following overexpression of Ago2 and downregulated by silencing Ago2 in the NAc. Knocking down miR-3068-5p, serving as an Ago2-dependent miRNA, strengthened the METH sensitization responses in mice. These findings demonstrated that dysregulated Ago2 in neurons in the NAc is capable of regulating METH sensitization and suggested a potential role of Ago2-dependent miR-3068-5p in METH sensitization.
Collapse
Affiliation(s)
- Dan Liu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China.,Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Min Liang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Li Zhu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Ting-Ting Zhou
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Yu Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Rui Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Fei-Fei Wu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Eyleen L K Goh
- Department of Research, National Neuroscience Institute, Singapore, Singapore.,Neuroscience and Mental Health Faculty, Lee Kong China School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Teng Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
58
|
Extracellular Metalloproteinases in the Plasticity of Excitatory and Inhibitory Synapses. Cells 2021; 10:cells10082055. [PMID: 34440823 PMCID: PMC8391609 DOI: 10.3390/cells10082055] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Long-term synaptic plasticity is shaped by the controlled reorganization of the synaptic proteome. A key component of this process is local proteolysis performed by the family of extracellular matrix metalloproteinases (MMPs). In recent years, considerable progress was achieved in identifying extracellular proteases involved in neuroplasticity phenomena and their protein substrates. Perisynaptic metalloproteinases regulate plastic changes at synapses through the processing of extracellular and membrane proteins. MMP9 was found to play a crucial role in excitatory synapses by controlling the NMDA-dependent LTP component. In addition, MMP3 regulates the L-type calcium channel-dependent form of LTP as well as the plasticity of neuronal excitability. Both MMP9 and MMP3 were implicated in memory and learning. Moreover, altered expression or mutations of different MMPs are associated with learning deficits and psychiatric disorders, including schizophrenia, addiction, or stress response. Contrary to excitatory drive, the investigation into the role of extracellular proteolysis in inhibitory synapses is only just beginning. Herein, we review the principal mechanisms of MMP involvement in the plasticity of excitatory transmission and the recently discovered role of proteolysis in inhibitory synapses. We discuss how different matrix metalloproteinases shape dynamics and turnover of synaptic adhesome and signal transduction pathways in neurons. Finally, we discuss future challenges in exploring synapse- and plasticity-specific functions of different metalloproteinases.
Collapse
|
59
|
Giorgi C, Marinelli S. Roles and Transcriptional Responses of Inhibitory Neurons in Learning and Memory. Front Mol Neurosci 2021; 14:689952. [PMID: 34211369 PMCID: PMC8239217 DOI: 10.3389/fnmol.2021.689952] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence supports a model whereby memories are encoded by sparse ensembles of neurons called engrams, activated during memory encoding and reactivated upon recall. An engram consists of a network of cells that undergo long-lasting modifications of their transcriptional programs and connectivity. Ground-breaking advancements in this field have been made possible by the creative exploitation of the characteristic transcriptional responses of neurons to activity, allowing both engram labeling and manipulation. Nevertheless, numerous aspects of engram cell-type composition and function remain to be addressed. As recent transcriptomic studies have revealed, memory encoding induces persistent transcriptional and functional changes in a plethora of neuronal subtypes and non-neuronal cells, including glutamatergic excitatory neurons, GABAergic inhibitory neurons, and glia cells. Dissecting the contribution of these different cellular classes to memory engram formation and activity is quite a challenging yet essential endeavor. In this review, we focus on the role played by the GABAergic inhibitory component of the engram through two complementary lenses. On one hand, we report on available physiological evidence addressing the involvement of inhibitory neurons to different stages of memory formation, consolidation, storage and recall. On the other, we capitalize on a growing number of transcriptomic studies that profile the transcriptional response of inhibitory neurons to activity, revealing important clues on their potential involvement in learning and memory processes. The picture that emerges suggests that inhibitory neurons are an essential component of the engram, likely involved in engram allocation, in tuning engram excitation and in storing the memory trace.
Collapse
Affiliation(s)
- Corinna Giorgi
- CNR, Institute of Molecular Biology and Pathology, Rome, Italy.,European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Silvia Marinelli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| |
Collapse
|
60
|
Wyroślak M, Lebida K, Mozrzymas JW. Induction of Inhibitory Synaptic Plasticity Enhances Tonic Current by Increasing the Content of α5-Subunit Containing GABA A Receptors in Hippocampal Pyramidal Neurons. Neuroscience 2021; 467:39-46. [PMID: 34033868 DOI: 10.1016/j.neuroscience.2021.05.020] [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: 03/24/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 11/16/2022]
Abstract
It is known that besides synaptic inhibition, there is a persistent component of inhibitory drive mediated by tonic currents which is believed to mediate majority of the total inhibitory charge in hippocampal neurons. Tonic currents, depending on cell types, can be mediated by a variety of GABAA receptor (GABAAR) subtypes but in pyramidal neurons, α5-subunit containing receptors were found to be predominant. Importantly, α5-GABAARs were implicated in both inhibitory and excitatory synaptic plasticity as well as in a variety of cognitive tasks. In the present study, we asked whether the protocol that evokes NMDAR-dependent GABAergic inhibitory long-term potentiation (iLTP) also induces the plasticity of tonic inhibition in hippocampal pyramidal neurons. Our whole-cell patch-clamp recordings revealed that the induction of this type of iLTP is associated with a marked increase in tonic current. By using the specific inverse agonist of α5-containing GABAARs (L-655,709) we provide evidence that this plastic change in tonic current is correlated with an increased proportion of this type of GABAARs. On the contrary, the iLTP induction did not affect the tonic current potentiated by THIP, indicating that the pool of δ subunit-containing GABAARs receptors remains unaffected. We conclude that the α5-GABAARs-dependent plasticity of tonic inhibition is a novel dimension of the neuroplasticity of the inhibitory drive in the hippocampal principal neurons. Overall, α5-containing GABAARs emerge as key players in a variety of plasticity mechanisms operating over a large span of time and spatial scales.
Collapse
Affiliation(s)
- Marcin Wyroślak
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland.
| | - Katarzyna Lebida
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367 Wroclaw, Poland
| |
Collapse
|
61
|
CCL2/CCR2 Contributes to the Altered Excitatory-inhibitory Synaptic Balance in the Nucleus Accumbens Shell Following Peripheral Nerve Injury-induced Neuropathic Pain. Neurosci Bull 2021; 37:921-933. [PMID: 34003466 DOI: 10.1007/s12264-021-00697-6] [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: 07/17/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
The medium spiny neurons (MSNs) in the nucleus accumbens (NAc) integrate excitatory and inhibitory synaptic inputs and gate motivational and emotional behavior output. Here we report that the relative intensity of excitatory and inhibitory synaptic inputs to MSNs of the NAc shell was decreased in mice with neuropathic pain induced by spinal nerve ligation (SNL). SNL increased the frequency, but not the amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs), and decreased both the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) in the MSNs. SNL also decreased the paired-pulse ratio (PPR) of evoked IPSCs but increased the PPR of evoked EPSCs. Moreover, acute bath application of C-C motif chemokine ligand 2 (CCL2) increased the frequency and amplitude of sIPSCs and sEPSCs in the MSNs, and especially strengthened the amplitude of N-methyl-D-aspartate receptor (NMDAR)-mediated miniature EPSCs. Further Ccl2 overexpression in the NAc in vivo decreased the peak amplitude of the sEPSC/sIPSC ratio. Finally, Ccr2 knock-down improved the impaired induction of NMDAR-dependent long-term depression (LTD) in the NAc after SNL. These results suggest that CCL2/CCR2 signaling plays a role in the integration of excitatory/inhibitory synaptic transmission and leads to an increase of the LTD induction threshold at the synapses of MSNs during neuropathic pain.
Collapse
|
62
|
Akil AE, Rosenbaum R, Josić K. Balanced networks under spike-time dependent plasticity. PLoS Comput Biol 2021; 17:e1008958. [PMID: 33979336 PMCID: PMC8143429 DOI: 10.1371/journal.pcbi.1008958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/24/2021] [Accepted: 04/12/2021] [Indexed: 11/28/2022] Open
Abstract
The dynamics of local cortical networks are irregular, but correlated. Dynamic excitatory–inhibitory balance is a plausible mechanism that generates such irregular activity, but it remains unclear how balance is achieved and maintained in plastic neural networks. In particular, it is not fully understood how plasticity induced changes in the network affect balance, and in turn, how correlated, balanced activity impacts learning. How do the dynamics of balanced networks change under different plasticity rules? How does correlated spiking activity in recurrent networks change the evolution of weights, their eventual magnitude, and structure across the network? To address these questions, we develop a theory of spike–timing dependent plasticity in balanced networks. We show that balance can be attained and maintained under plasticity–induced weight changes. We find that correlations in the input mildly affect the evolution of synaptic weights. Under certain plasticity rules, we find an emergence of correlations between firing rates and synaptic weights. Under these rules, synaptic weights converge to a stable manifold in weight space with their final configuration dependent on the initial state of the network. Lastly, we show that our framework can also describe the dynamics of plastic balanced networks when subsets of neurons receive targeted optogenetic input. Animals are able to learn complex tasks through changes in individual synapses between cells. Such changes lead to the coevolution of neural activity patterns and the structure of neural connectivity, but the consequences of these interactions are not fully understood. We consider plasticity in model neural networks which achieve an average balance between the excitatory and inhibitory synaptic inputs to different cells, and display cortical–like, irregular activity. We extend the theory of balanced networks to account for synaptic plasticity and show which rules can maintain balance, and which will drive the network into a different state. This theory of plasticity can provide insights into the relationship between stimuli, network dynamics, and synaptic circuitry.
Collapse
Affiliation(s)
- Alan Eric Akil
- Department of Mathematics, University of Houston, Houston, Texas, United States of America
| | - Robert Rosenbaum
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, Indiana, United States of America
- Interdisciplinary Center for Network Science and Applications, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Krešimir Josić
- Department of Mathematics, University of Houston, Houston, Texas, United States of America
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
63
|
Puścian A, Benisty H, Higley MJ. NMDAR-Dependent Emergence of Behavioral Representation in Primary Visual Cortex. Cell Rep 2021; 32:107970. [PMID: 32726633 DOI: 10.1016/j.celrep.2020.107970] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/28/2020] [Accepted: 07/08/2020] [Indexed: 10/23/2022] Open
Abstract
Although neocortical sensory areas are generally thought to faithfully represent external stimuli, cortical networks exhibit considerable functional plasticity, allowing them to modify their output to reflect ongoing behavioral demands. We apply longitudinal 2-photon imaging of activity in the primary visual cortex (V1) of mice learning a conditioned eyeblink task to investigate the dynamic representations of task-relevant information. We find that, although all V1 neurons robustly and stably encode visual input, pyramidal cells and parvalbumin-expressing interneurons exhibit experience-dependent emergence of accurate behavioral representations during learning. The functional plasticity driving performance-predictive activity requires cell-autonomous expression of NMDA-type glutamate receptors. Our findings demonstrate that accurate encoding of behavioral output is not inherent to V1 but develops during learning to support visual task performance.
Collapse
Affiliation(s)
- Alicja Puścian
- Department of Neuroscience, Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Nencki-EMBL Partnership for Neural Plasticity and Brain Disorders - BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3 Street, 02-093 Warsaw, Poland.
| | - Hadas Benisty
- Department of Neuroscience, Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Michael J Higley
- Department of Neuroscience, Kavli Institute of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
64
|
Local miRNA-Dependent Translational Control of GABA AR Synthesis during Inhibitory Long-Term Potentiation. Cell Rep 2021; 31:107785. [PMID: 32579917 PMCID: PMC7486624 DOI: 10.1016/j.celrep.2020.107785] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 04/14/2020] [Accepted: 05/28/2020] [Indexed: 12/29/2022] Open
Abstract
Molecular mechanisms underlying plasticity at brain inhibitory synapses remain poorly characterized. Increased postsynaptic clustering of GABAA receptors (GABAARs) rapidly strengthens inhibition during inhibitory long-term potentiation (iLTP). However, it is unclear how synaptic GABAAR clustering is maintained to sustain iLTP. Here, we identify a role for miR376c in regulating the translation of mRNAs encoding the synaptic α1 and γ2 GABAAR subunits, GABRA1 and GABRG2, respectively. Following iLTP induction, transcriptional repression of miR376c is induced through a calcineurin-NFAT-HDAC signaling pathway and promotes increased translation and clustering of synaptic GABAARs. This pathway is essential for the long-term expression of iLTP and is blocked by miR376c overexpression, specifically impairing inhibitory synaptic strength. Finally, we show that local de novo synthesis of synaptic GABAARs occurs exclusively in dendrites and in a miR376c-dependent manner following iLTP. Together, this work describes a local post-transcriptional mechanism that regulates inhibitory synaptic plasticity via miRNA control of dendritic protein synthesis. Clustering of GABAARs at inhibitory synapses is crucial for synaptic inhibition. Rajgor et al. discover that synaptic GABAAR expression is controlled by their local translation, regulated by miR376c. During inhibitory synaptic potentiation, miR376c is downregulated, relieving its translational repression of GABAAR mRNAs and leading to de novo synthesis of dendritic GABAARs.
Collapse
|
65
|
Cook SG, Buonarati OR, Coultrap SJ, Bayer KU. CaMKII holoenzyme mechanisms that govern the LTP versus LTD decision. SCIENCE ADVANCES 2021; 7:7/16/eabe2300. [PMID: 33853773 PMCID: PMC8046365 DOI: 10.1126/sciadv.abe2300] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/24/2021] [Indexed: 05/30/2023]
Abstract
Higher brain functions are thought to require synaptic frequency decoding that can lead to long-term potentiation (LTP) or depression (LTD). We show that the LTP versus LTD decision is determined by complex cross-regulation of T286 and T305/306 autophosphorylation within the 12meric CaMKII holoenzyme, which enabled molecular computation of stimulus frequency, amplitude, and duration. Both LTP and LTD require T286 phosphorylation, but T305/306 phosphorylation selectively promoted LTD. In response to excitatory LTP versus LTD stimuli, the differential T305/306 phosphorylation directed CaMKII movement to either excitatory or inhibitory synapses, thereby coordinating plasticity at both synapse types. Fast T305/306 phosphorylation required prior T286 phosphorylation and then curbed CaMKII activity by two mechanisms: (i) a cis-subunit reaction reduced both Ca2+ stimulation and autonomous activity and (ii) a trans-subunit reaction enabled complete activity shutdown and feed-forward inhibition of further T286 phosphorylation. These are fundamental additions to the long-studied CaMKII regulation and function in neuronal plasticity.
Collapse
Affiliation(s)
- Sarah G Cook
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Olivia R Buonarati
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Steven J Coultrap
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
66
|
Modulation of Coordinated Activity across Cortical Layers by Plasticity of Inhibitory Synapses. Cell Rep 2021; 30:630-641.e5. [PMID: 31968242 PMCID: PMC6988114 DOI: 10.1016/j.celrep.2019.12.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 11/21/2019] [Accepted: 12/13/2019] [Indexed: 11/25/2022] Open
Abstract
In the neocortex, synaptic inhibition shapes all forms of spontaneous and sensory evoked activity. Importantly, inhibitory transmission is highly plastic, but the functional role of inhibitory synaptic plasticity is unknown. In the mouse barrel cortex, activation of layer (L) 2/3 pyramidal neurons (PNs) elicits strong feedforward inhibition (FFI) onto L5 PNs. We find that FFI involving parvalbumin (PV)-expressing cells is strongly potentiated by postsynaptic PN burst firing. FFI plasticity modifies the PN excitation-to-inhibition (E/I) ratio, strongly modulates PN gain, and alters information transfer across cortical layers. Moreover, our LTPi-inducing protocol modifies firing of L5 PNs and alters the temporal association of PN spikes to γ-oscillations both in vitro and in vivo. All of these effects are captured by unbalancing the E/I ratio in a feedforward inhibition circuit model. Altogether, our results indicate that activity-dependent modulation of perisomatic inhibitory strength effectively influences the participation of single principal cortical neurons to cognition-relevant network activity. Feedforward inhibition (FFI) of layer 5 pyramidal neurons (PNs) is highly plastic Long-term potentiation of FFI modulates spiking activity of layer 5 PNs LTPi affects information transfer across cortical layers The strength of LTPi determines the phase locking of PN firing to γ-oscillations
Collapse
|
67
|
Wiera G, Lebida K, Lech AM, Brzdąk P, Van Hove I, De Groef L, Moons L, Petrini EM, Barberis A, Mozrzymas JW. Long-term plasticity of inhibitory synapses in the hippocampus and spatial learning depends on matrix metalloproteinase 3. Cell Mol Life Sci 2021; 78:2279-2298. [PMID: 32959071 PMCID: PMC7966195 DOI: 10.1007/s00018-020-03640-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/19/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022]
Abstract
Learning and memory are known to depend on synaptic plasticity. Whereas the involvement of plastic changes at excitatory synapses is well established, plasticity mechanisms at inhibitory synapses only start to be discovered. Extracellular proteolysis is known to be a key factor in glutamatergic plasticity but nothing is known about its role at GABAergic synapses. We reveal that pharmacological inhibition of MMP3 activity or genetic knockout of the Mmp3 gene abolishes induction of postsynaptic iLTP. Moreover, the application of exogenous active MMP3 mimics major iLTP manifestations: increased mIPSCs amplitude, enlargement of synaptic gephyrin clusters, and a decrease in the diffusion coefficient of synaptic GABAA receptors that favors their entrapment within the synapse. Finally, we found that MMP3 deficient mice show faster spatial learning in Morris water maze and enhanced contextual fear conditioning. We conclude that MMP3 plays a key role in iLTP mechanisms and in the behaviors that presumably in part depend on GABAergic plasticity.
Collapse
Affiliation(s)
- Grzegorz Wiera
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, 50-367, Wroclaw, Poland.
| | - Katarzyna Lebida
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, 50-367, Wroclaw, Poland.
| | - Anna Maria Lech
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, 50-367, Wroclaw, Poland
- Laboratory of Cellular Neurobiology, Department of Physiology and Molecular Neurobiology, Wroclaw University, 50-205, Wroclaw, Poland
| | - Patrycja Brzdąk
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, 50-367, Wroclaw, Poland
- Laboratory of Cellular Neurobiology, Department of Physiology and Molecular Neurobiology, Wroclaw University, 50-205, Wroclaw, Poland
| | - Inge Van Hove
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), 3000, Leuven, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), 3000, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), 3000, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Enrica Maria Petrini
- Laboratory of Synaptic Plasticity of Inhibitory Networks, Fondazione Istituto Italiano Di Tecnologia, 16163, Genoa, Italy
| | - Andrea Barberis
- Laboratory of Synaptic Plasticity of Inhibitory Networks, Fondazione Istituto Italiano Di Tecnologia, 16163, Genoa, Italy
| | - Jerzy W Mozrzymas
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, 50-367, Wroclaw, Poland
| |
Collapse
|
68
|
Uliana DL, Gomes FV, Grace AA. Stress impacts corticoamygdalar connectivity in an age-dependent manner. Neuropsychopharmacology 2021; 46:731-740. [PMID: 33096542 PMCID: PMC8027626 DOI: 10.1038/s41386-020-00886-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 01/13/2023]
Abstract
Stress is a socio-environmental risk factor for the development of psychiatric disorders, with the age of exposure potentially determining the outcome. Several brain regions mediate stress responsivity, with a prominent role of the medial prefrontal cortex (mPFC) and basolateral amygdala (BLA) and their reciprocal inhibitory connectivity. Here we investigated the impact of stress exposure during adolescence and adulthood on the activity of putative pyramidal neurons in the BLA and corticoamygdalar plasticity using in vivo electrophysiology. 155 male Sprague-Dawley rats were subjected to a combination of footshock/restraint stress in either adolescence (postnatal day 31-40) or adulthood (postnatal day 65-74). Both adolescent and adult stress increased the number of spontaneously active putative BLA pyramidal neurons 1-2 weeks, but not 5-6 weeks post stress. High-frequency stimulation (HFS) of BLA and mPFC depressed evoked spike probability in the mPFC and BLA, respectively, in adult but not adolescent rats. In contrast, an adult-like BLA HFS-induced decrease in spike probability of mPFC neurons was found 1-2 weeks post-adolescent stress. Changes in mPFC and BLA neuron discharge were found 1-2 weeks post-adult stress after BLA and mPFC HFS, respectively. All these changes were transient since they were not found 5-6 weeks post adolescent or adult stress. Our findings indicate that stress during adolescence may accelerate the development of BLA-PFC plasticity, probably due to BLA hyperactivity, which can also disrupt the reciprocal communication of BLA-mPFC after adult stress. Therefore, precocious BLA-mPFC connectivity alterations may represent an early adaptive stress response that ultimately may contribute to vulnerability to adult psychiatric disorders.
Collapse
Affiliation(s)
- Daniela L. Uliana
- grid.21925.3d0000 0004 1936 9000Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA USA
| | - Felipe V. Gomes
- grid.21925.3d0000 0004 1936 9000Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA USA ,grid.11899.380000 0004 1937 0722Present Address: Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP Brazil
| | - Anthony A. Grace
- grid.21925.3d0000 0004 1936 9000Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
69
|
Yang XY, Ma ZL, Storm DR, Cao H, Zhang YQ. Selective ablation of type 3 adenylyl cyclase in somatostatin-positive interneurons produces anxiety- and depression-like behaviors in mice. World J Psychiatry 2021; 11:35-49. [PMID: 33643860 PMCID: PMC7896247 DOI: 10.5498/wjp.v11.i2.35] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a highly disabling psychiatric syndrome associated with deficits of specific subpopulations of cortical GABAergic interneurons; however, the underlying molecular mechanism remains unknown. Type 3 adenylyl cyclase (ADCY3, AC3), which is important for neuronal excitability, has been implicated in MDD in a genome-wide association study in humans. Moreover, a study reported that ablation of AC3 in mice caused similar symptoms as MDD patients. AIM To determine if disruption of the AC3 gene in different subtypes of GABAergic interneurons of mice causes depression-like behaviors. METHODS Using immunohistochemistry, we investigated the expression of AC3 in two major subtypes GABAergic interneurons: Somatostatin-positive (SST+) and parvalbumin-positive (PV+) neurons. Genetic manipulations were used to selectively disrupt AC3 expression in SST+ or PV+ interneurons. A series of behavior tests including rotarod test, open field test (OFT), elevated plus maze test (EPM), forced swimming test (FST), and tail suspension test (TST) were used to evaluate the motor ability, anxiety- and depression- like behaviors, respectively. RESULTS Our results indicate that approximately 90.41% of SST+ and 91.22% of PV+ interneurons express AC3. After ablation of AC3 in SST+ interneurons, the mice spent comparable time in the center area in OFT, but significantly less time in the open arms and low frequency of entries to the open arms in EPM. Furthermore, these mice showed prolonged immobility in FST and more freezing in TST. However, there were no significant changes in these behaviors after specific disruption of AC3 in PV+ interneurons. CONCLUSION This study indicates that ablation of AC3 in SST+ interneurons of mice increases anxiety- and depression-like behaviors in mice, supporting the general hypothesis that decreased AC3 activity may play a role in human depression.
Collapse
Affiliation(s)
- Xiao-Yu Yang
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Zhao-Liang Ma
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Daniel R Storm
- Department of Pharmacology, University of Washington, Seattle, WA 98105, United States
| | - Hong Cao
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Yu-Qiu Zhang
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| |
Collapse
|
70
|
Davenport CM, Rajappa R, Katchan L, Taylor CR, Tsai MC, Smith CM, de Jong JW, Arnold DB, Lammel S, Kramer RH. Relocation of an Extrasynaptic GABA A Receptor to Inhibitory Synapses Freezes Excitatory Synaptic Strength and Preserves Memory. Neuron 2021; 109:123-134.e4. [PMID: 33096025 PMCID: PMC7790995 DOI: 10.1016/j.neuron.2020.09.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/21/2020] [Accepted: 09/25/2020] [Indexed: 11/27/2022]
Abstract
The excitatory synapse between hippocampal CA3 and CA1 pyramidal neurons exhibits long-term potentiation (LTP), a positive feedback process implicated in learning and memory in which postsynaptic depolarization strengthens synapses, promoting further depolarization. Without mechanisms for interrupting positive feedback, excitatory synapses could strengthen inexorably, corrupting memory storage. Here, we reveal a hidden form of inhibitory synaptic plasticity that prevents accumulation of excitatory LTP. We developed a knockin mouse that allows optical control of endogenous α5-subunit-containing γ-aminobutyric acid (GABA)A receptors (α5-GABARs). Induction of excitatory LTP relocates α5-GABARs, which are ordinarily extrasynaptic, to inhibitory synapses, quashing further NMDA receptor activation necessary for inducing more excitatory LTP. Blockade of α5-GABARs accelerates reversal learning, a behavioral test for cognitive flexibility dependent on repeated LTP. Hence, inhibitory synaptic plasticity occurs in parallel with excitatory synaptic plasticity, with the ensuing interruption of the positive feedback cycle of LTP serving as a possible critical early step in preserving memory.
Collapse
Affiliation(s)
- Christopher M Davenport
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rajit Rajappa
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ljudmila Katchan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Charlotte R Taylor
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ming-Chi Tsai
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Caleb M Smith
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Johannes W de Jong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Don B Arnold
- Department of Biology, Section of Molecular and Computational Biology, University of Southern California, Los Angeles, Los Angeles, CA 90089, USA
| | - Stephan Lammel
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
71
|
Savalia NK, Shao LX, Kwan AC. A Dendrite-Focused Framework for Understanding the Actions of Ketamine and Psychedelics. Trends Neurosci 2020; 44:260-275. [PMID: 33358035 DOI: 10.1016/j.tins.2020.11.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 02/09/2023]
Abstract
Pilot studies have hinted that serotonergic psychedelics such as psilocybin may relieve depression, and could possibly do so by promoting neural plasticity. Intriguingly, another psychotomimetic compound, ketamine, is a fast-acting antidepressant and induces synapse formation. The similarities in behavioral and neural effects have been puzzling because the compounds target distinct molecular receptors in the brain. In this opinion article, we develop a conceptual framework that suggests the actions of ketamine and serotonergic psychedelics may converge at the dendrites, to both enhance and suppress membrane excitability. We speculate that mismatches in the opposing actions on dendritic excitability may relate to these compounds' cell-type and region selectivity, their moderate range of effects and toxicity, and their plasticity-promoting capacities.
Collapse
Affiliation(s)
- Neil K Savalia
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Ling-Xiao Shao
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Alex C Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
72
|
Zamora-Moratalla A, Martín ED. Prolactin enhances hippocampal synaptic plasticity in female mice of reproductive age. Hippocampus 2020; 31:281-293. [PMID: 33285014 PMCID: PMC7983975 DOI: 10.1002/hipo.23288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 01/27/2023]
Abstract
Dynamic signaling between the endocrine system (ES) and the nervous system (NS) is essential for brain and body homeostasis. In particular, reciprocal interaction occurs during pregnancy and motherhood that may involve changes in some brain plasticity processes. Prolactin (PRL), a hormone with pleiotropic effects on the NS, promotes maternal behavior and has been linked to modifications in brain circuits during motherhood; however, it is unclear whether PRL may regulate synaptic plasticity. Therefore, the main aim of the present work was to determine the cellular and molecular mechanisms triggered by PRL that regulate synaptic plasticity in the hippocampus. By analyzing extracellular recordings in CA3‐CA1 synapses of hippocampal slices, we report that PRL modifies short and long‐term synaptic plasticity in female mice of reproductive age, but not in sexually immature females or adult males. This effect is carried out through mechanisms that include participation of GABAA receptors and activation of the JAK2‐mediated signaling pathway. These findings show for the first time how PRL enhances the synaptic strength in hippocampal circuits and that this effect is sexually dimorphic, which would influence complex brain processes in physiological conditions like pregnancy and lactation.
Collapse
Affiliation(s)
- Alfonsa Zamora-Moratalla
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eduardo D Martín
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
73
|
Interneuron-specific plasticity at parvalbumin and somatostatin inhibitory synapses onto CA1 pyramidal neurons shapes hippocampal output. Nat Commun 2020; 11:4395. [PMID: 32879322 PMCID: PMC7467931 DOI: 10.1038/s41467-020-18074-8] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 07/31/2020] [Indexed: 12/22/2022] Open
Abstract
The formation and maintenance of spatial representations within hippocampal cell assemblies is strongly dictated by patterns of inhibition from diverse interneuron populations. Although it is known that inhibitory synaptic strength is malleable, induction of long-term plasticity at distinct inhibitory synapses and its regulation of hippocampal network activity is not well understood. Here, we show that inhibitory synapses from parvalbumin and somatostatin expressing interneurons undergo long-term depression and potentiation respectively (PV-iLTD and SST-iLTP) during physiological activity patterns. Both forms of plasticity rely on T-type calcium channel activation to confer synapse specificity but otherwise employ distinct mechanisms. Since parvalbumin and somatostatin interneurons preferentially target perisomatic and distal dendritic regions respectively of CA1 pyramidal cells, PV-iLTD and SST-iLTP coordinate a reprioritisation of excitatory inputs from entorhinal cortex and CA3. Furthermore, circuit-level modelling reveals that PV-iLTD and SST-iLTP cooperate to stabilise place cells while facilitating representation of multiple unique environments within the hippocampal network. Inhibitory interneuron subtypes differentially control place cell representations in CA1. Here, the authors show that parvalbumin and somatostatin interneuron synapses onto CA1 pyramidal neurons exhibit distinct plasticity mechanisms and incorporating this insight into circuit-level modeling leads to stable place cell representations.
Collapse
|
74
|
Herstel LJ, Wierenga CJ. Network control through coordinated inhibition. Curr Opin Neurobiol 2020; 67:34-41. [PMID: 32853970 DOI: 10.1016/j.conb.2020.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 12/29/2022]
Abstract
Coordinated excitatory and inhibitory activity is required for proper brain functioning. Recent computational and experimental studies have demonstrated that activity patterns in recurrent cortical networks are dominated by inhibition. Whereas previous studies have suggested that inhibitory plasticity is important for homeostatic control, this new framework puts inhibition in the driver's seat. Complex neuronal networks in the brain comprise many configurations in parallel, controlled by external and internal 'switches'. Context-dependent modulation and plasticity of inhibitory connections play a key role in memory and learning. It is therefore important to realize that synaptic plasticity is often multisynaptic and that a proper balance between excitation and inhibition is not fixed, but depends on context and activity level.
Collapse
Affiliation(s)
- Lotte J Herstel
- Cell Biology, Neurobiology and Biophysics, Biology Department, Faculty of Science, Utrecht University, The Netherlands
| | - Corette J Wierenga
- Cell Biology, Neurobiology and Biophysics, Biology Department, Faculty of Science, Utrecht University, The Netherlands.
| |
Collapse
|
75
|
Capogna M, Castillo PE, Maffei A. The ins and outs of inhibitory synaptic plasticity: Neuron types, molecular mechanisms and functional roles. Eur J Neurosci 2020; 54:6882-6901. [PMID: 32663353 DOI: 10.1111/ejn.14907] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/30/2020] [Accepted: 07/08/2020] [Indexed: 01/05/2023]
Abstract
GABAergic interneurons are highly diverse, and their synaptic outputs express various forms of plasticity. Compelling evidence indicates that activity-dependent changes of inhibitory synaptic transmission play a significant role in regulating neural circuits critically involved in learning and memory and circuit refinement. Here, we provide an updated overview of inhibitory synaptic plasticity with a focus on the hippocampus and neocortex. To illustrate the diversity of inhibitory interneurons, we discuss the case of two highly divergent interneuron types, parvalbumin-expressing basket cells and neurogliaform cells, which support unique roles on circuit dynamics. We also present recent progress on the molecular mechanisms underlying long-term, activity-dependent plasticity of fast inhibitory transmission. Lastly, we discuss the role of inhibitory synaptic plasticity in neuronal circuits' function. The emerging picture is that inhibitory synaptic transmission in the CNS is extremely diverse, undergoes various mechanistically distinct forms of plasticity and contributes to a much more refined computational role than initially thought. Both the remarkable diversity of inhibitory interneurons and the various forms of plasticity expressed by GABAergic synapses provide an amazingly rich inhibitory repertoire that is central to a variety of complex neural circuit functions, including memory.
Collapse
Affiliation(s)
- Marco Capogna
- Department of Biomedicine, Danish National Research Foundation Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Pablo E Castillo
- Dominck P Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Arianna Maffei
- Center for Neural Circuit Dynamics and Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
76
|
Hu HY, Kruijssen DLH, Frias CP, Rózsa B, Hoogenraad CC, Wierenga CJ. Endocannabinoid Signaling Mediates Local Dendritic Coordination between Excitatory and Inhibitory Synapses. Cell Rep 2020; 27:666-675.e5. [PMID: 30995465 DOI: 10.1016/j.celrep.2019.03.078] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/21/2018] [Accepted: 03/21/2019] [Indexed: 01/01/2023] Open
Abstract
Dendritic inhibitory synapses are most efficient in modulating excitatory inputs localized on the same dendrite, but it is unknown whether their location is random or regulated. Here, we show that the formation of inhibitory synapses can be directed by excitatory synaptic activity on the same dendrite. We stimulated dendritic spines close to a GABAergic axon crossing by pairing two-photon glutamate uncaging with postsynaptic depolarization in CA1 pyramidal cells. We found that repeated spine stimulation promoted growth of a GABAergic bouton onto the same dendrite. The dendritic feedback signal required postsynaptic activation of DAGL, which produces the endocannabinoid 2-AG, and was mediated by CB1 receptors. We could also induce inhibitory bouton growth by local, brief applications of 2-AG. Our findings reveal a dendritic signaling mechanism to trigger growth of an inhibitory bouton at dendritic locations with strong excitatory synaptic activity, and this mechanism may serve to ensure inhibitory control over clustered excitatory inputs.
Collapse
Affiliation(s)
- Hai Yin Hu
- Department of Biology, Science for Life, Utrecht University, 3584CH Utrecht, the Netherlands
| | - Dennis L H Kruijssen
- Department of Biology, Science for Life, Utrecht University, 3584CH Utrecht, the Netherlands
| | - Cátia P Frias
- Department of Biology, Science for Life, Utrecht University, 3584CH Utrecht, the Netherlands
| | - Balázs Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest 1083, Hungary; Faculty of Information Technology, Pázmány Péter Catholic University, Budapest 1083, Hungary
| | - Casper C Hoogenraad
- Department of Biology, Science for Life, Utrecht University, 3584CH Utrecht, the Netherlands
| | - Corette J Wierenga
- Department of Biology, Science for Life, Utrecht University, 3584CH Utrecht, the Netherlands.
| |
Collapse
|
77
|
Bannai H, Niwa F, Sakuragi S, Mikoshiba K. Inhibitory synaptic transmission tuned by Ca 2+ and glutamate through the control of GABA A R lateral diffusion dynamics. Dev Growth Differ 2020; 62:398-406. [PMID: 32329058 PMCID: PMC7496684 DOI: 10.1111/dgd.12667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/08/2020] [Accepted: 04/18/2020] [Indexed: 11/30/2022]
Abstract
The GABAergic synapses, a primary inhibitory synapse in the mammalian brain, is important for the normal development of brain circuits, and for the regulation of the excitation‐inhibition balance critical for brain function from the developmental stage throughout life. However, the molecular mechanism underlying the formation, maintenance, and modulation of GABAergic synapses is less understood compared to that of excitatory synapses. Quantum dot‐single particle tracking (QD‐SPT), a super‐resolution imaging technique that enables the analysis of membrane molecule dynamics at single‐molecule resolution, is a powerful tool to analyze the behavior of proteins and lipids on the plasma membrane. In this review, we summarize the recent application of QD‐SPT in understanding of GABAergic synaptic transmission. Here we introduce QD‐SPT experiments that provide further insights into the molecular mechanism supporting GABAergic synapses. QD‐SPT studies revealed that glutamate and Ca2+ signaling is involved in (a) the maintenance of GABAergic synapses, (b) GABAergic long‐term depression, and GABAergic long‐term potentiation, by specifically activating signaling pathways unique to each phenomenon. We also introduce a novel Ca2+ imaging technique to describe the diversity of Ca2+ signals that may activate the downstream signaling pathways that induce specific biological output.
Collapse
Affiliation(s)
- Hiroko Bannai
- School of Advanced Science and Engineering, Department of Electrical Engineering and Biosciences, Waseda University, Tokyo, Japan.,Department of Neurophysiology, Keio University School of Medicine, Tokyo, Japan.,Japan Science and Technology Agency, PRESTO, Kawaguchi, Saitama, Japan.,Laboratory for Developmental Neurobiology, RIKEN Center for Brain Science, Wako, Japan
| | - Fumihiro Niwa
- Laboratory for Developmental Neurobiology, RIKEN Center for Brain Science, Wako, Japan.,Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, PSL Research University, INSERM, CNRS, Paris, France
| | - Shigeo Sakuragi
- School of Advanced Science and Engineering, Department of Electrical Engineering and Biosciences, Waseda University, Tokyo, Japan.,Department of Pharmacology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Center for Brain Science, Wako, Japan.,Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China.,Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Japan
| |
Collapse
|
78
|
Mossner JM, Batista-Brito R, Pant R, Cardin JA. Developmental loss of MeCP2 from VIP interneurons impairs cortical function and behavior. eLife 2020; 9:55639. [PMID: 32343226 PMCID: PMC7213975 DOI: 10.7554/elife.55639] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/28/2020] [Indexed: 12/24/2022] Open
Abstract
Rett Syndrome is a devastating neurodevelopmental disorder resulting from mutations in the gene MECP2. Mutations of Mecp2 that are restricted to GABAergic cell types largely replicate the behavioral phenotypes associated with mouse models of Rett Syndrome, suggesting a pathophysiological role for inhibitory interneurons. Recent work has suggested that vasoactive intestinal peptide-expressing (VIP) interneurons may play a critical role in the proper development and function of cortical circuits, making them a potential key point of vulnerability in neurodevelopmental disorders. However, little is known about the role of VIP interneurons in Rett Syndrome. Here we find that loss of MeCP2 specifically from VIP interneurons replicates key neural and behavioral phenotypes observed following global Mecp2 loss of function.
Collapse
Affiliation(s)
- James M Mossner
- Department of Neuroscience, Yale University, New Haven, United States
| | - Renata Batista-Brito
- Department of Neuroscience, Yale University, New Haven, United States.,Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States
| | - Rima Pant
- Department of Neuroscience, Yale University, New Haven, United States
| | - Jessica A Cardin
- Department of Neuroscience, Yale University, New Haven, United States.,Kavli Institute for Neuroscience, Yale University, New Haven, United States
| |
Collapse
|
79
|
Nitric Oxide Signaling Strengthens Inhibitory Synapses of Cerebellar Molecular Layer Interneurons through a GABARAP-Dependent Mechanism. J Neurosci 2020; 40:3348-3359. [PMID: 32169968 DOI: 10.1523/jneurosci.2211-19.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/13/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) is an important signaling molecule that fulfills diverse functional roles as a neurotransmitter or diffusible second messenger in the developing and adult CNS. Although the impact of NO on different behaviors such as movement, sleep, learning, and memory has been well documented, the identity of its molecular and cellular targets is still an area of ongoing investigation. Here, we identify a novel role for NO in strengthening inhibitory GABAA receptor-mediated transmission in molecular layer interneurons of the mouse cerebellum. NO levels are elevated by the activity of neuronal NO synthase (nNOS) following Ca2+ entry through extrasynaptic NMDA-type ionotropic glutamate receptors (NMDARs). NO activates protein kinase G with the subsequent production of cGMP, which prompts the stimulation of NADPH oxidase and protein kinase C (PKC). The activation of PKC promotes the selective strengthening of α3-containing GABAARs synapses through a GΑΒΑ receptor-associated protein-dependent mechanism. Given the widespread but cell type-specific expression of the NMDAR/nNOS complex in the mammalian brain, our data suggest that NMDARs may uniquely strengthen inhibitory GABAergic transmission in these cells through a novel NO-mediated pathway.SIGNIFICANCE STATEMENT Long-term changes in the efficacy of GABAergic transmission is mediated by multiple presynaptic and postsynaptic mechanisms. A prominent pathway involves crosstalk between excitatory and inhibitory synapses whereby Ca2+-entering through postsynaptic NMDARs promotes the recruitment and strengthening of GABAA receptor synapses via Ca2+/calmodulin-dependent protein kinase II. Although Ca2+ transport by NMDARs is also tightly coupled to nNOS activity and NO production, it has yet to be determined whether this pathway affects inhibitory synapses. Here, we show that activation of NMDARs trigger a NO-dependent pathway that strengthens inhibitory GABAergic synapses of cerebellar molecular layer interneurons. Given the widespread expression of NMDARs and nNOS in the mammalian brain, we speculate that NO control of GABAergic synapse efficacy may be more widespread than has been appreciated.
Collapse
|
80
|
Antidepressant mechanisms of ketamine: Focus on GABAergic inhibition. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 89:43-78. [PMID: 32616214 DOI: 10.1016/bs.apha.2020.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There has been much recent progress in understanding of the mechanism of ketamine's rapid and enduring antidepressant effects. Here we review recent insights from clinical and preclinical studies, with special emphasis of ketamine-induced changes in GABAergic synaptic transmission that are considered essential for its antidepressant therapeutic effects. Subanesthetic ketamine is now understood to exert its initial action by selectively blocking a subset of NMDA receptors on GABAergic interneurons, which results in disinhibition of glutamatergic target neurons, a surge in extracellular glutamate and correspondingly elevated glutamatergic synaptic transmission. This surge in glutamate appears to be corroborated by the rapid metabolism of ketamine into hydroxynorketamine, which acts at presynaptic sites to disinhibit the release of glutamate. Preclinical studies indicate that glutamate-induced activity triggers the release of BDNF, followed by transient activation of the mTOR pathway and increased expression of synaptic proteins, along with functional strengthening of glutamatergic synapses. This drug-on phase lasts for approximately 2h and is followed by a period of days characterized by structural maturation of newly formed glutamatergic synapses and prominently enhanced GABAergic synaptic inhibition. Evidence from mouse models with constitutive antidepressant-like phenotypes suggests that this phase involves strengthened inhibition of dendrites by somatostatin-positive GABAergic interneurons and correspondingly reduced NMDA receptor-mediated Ca2+ entry into dendrites, which activates an intracellular signaling cascade that converges with the mTOR pathway onto increased activity of the eukaryotic elongation factor eEF2 and enhanced translation of dendritic mRNAs. Newly synthesized proteins such as BDNF may be important for the prolonged therapeutic effects of ketamine.
Collapse
|
81
|
Field RE, D'amour JA, Tremblay R, Miehl C, Rudy B, Gjorgjieva J, Froemke RC. Heterosynaptic Plasticity Determines the Set Point for Cortical Excitatory-Inhibitory Balance. Neuron 2020; 106:842-854.e4. [PMID: 32213321 DOI: 10.1016/j.neuron.2020.03.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/27/2019] [Accepted: 03/03/2020] [Indexed: 01/24/2023]
Abstract
Excitation in neural circuits must be carefully controlled by inhibition to regulate information processing and network excitability. During development, cortical inhibitory and excitatory inputs are initially mismatched but become co-tuned or balanced with experience. However, little is known about how excitatory-inhibitory balance is defined at most synapses or about the mechanisms for establishing or maintaining this balance at specific set points. Here we show how coordinated long-term plasticity calibrates populations of excitatory-inhibitory inputs onto mouse auditory cortical pyramidal neurons. Pairing pre- and postsynaptic activity induced plasticity at paired inputs and different forms of heterosynaptic plasticity at the strongest unpaired synapses, which required minutes of activity and dendritic Ca2+ signaling to be computed. Theoretical analyses demonstrated how the relative rate of heterosynaptic plasticity could normalize and stabilize synaptic strengths to achieve any possible excitatory-inhibitory correlation. Thus, excitatory-inhibitory balance is dynamic and cell specific, determined by distinct plasticity rules across multiple excitatory and inhibitory synapses.
Collapse
Affiliation(s)
- Rachel E Field
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA; Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
| | - James A D'amour
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA; Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
| | - Robin Tremblay
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA; Department of Anesthesiology, New York University School of Medicine, New York, NY 10016, USA
| | - Christoph Miehl
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany; School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Bernardo Rudy
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA; Department of Anesthesiology, New York University School of Medicine, New York, NY 10016, USA
| | - Julijana Gjorgjieva
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany; School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Robert C Froemke
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA; Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
82
|
Whole-Neuron Synaptic Mapping Reveals Spatially Precise Excitatory/Inhibitory Balance Limiting Dendritic and Somatic Spiking. Neuron 2020; 106:566-578.e8. [PMID: 32169170 DOI: 10.1016/j.neuron.2020.02.015] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/19/2019] [Accepted: 02/11/2020] [Indexed: 02/02/2023]
Abstract
The balance between excitatory and inhibitory (E and I) synapses is thought to be critical for information processing in neural circuits. However, little is known about the spatial principles of E and I synaptic organization across the entire dendritic tree of mammalian neurons. We developed a new open-source reconstruction platform for mapping the size and spatial distribution of E and I synapses received by individual genetically-labeled layer 2/3 (L2/3) cortical pyramidal neurons (PNs) in vivo. We mapped over 90,000 E and I synapses across twelve L2/3 PNs and uncovered structured organization of E and I synapses across dendritic domains as well as within individual dendritic segments. Despite significant domain-specific variation in the absolute density of E and I synapses, their ratio is strikingly balanced locally across dendritic segments. Computational modeling indicates that this spatially precise E/I balance dampens dendritic voltage fluctuations and strongly impacts neuronal firing output.
Collapse
|
83
|
Gandolfi D, Bigiani A, Porro CA, Mapelli J. Inhibitory Plasticity: From Molecules to Computation and Beyond. Int J Mol Sci 2020; 21:E1805. [PMID: 32155701 PMCID: PMC7084224 DOI: 10.3390/ijms21051805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 11/17/2022] Open
Abstract
Synaptic plasticity is the cellular and molecular counterpart of learning and memory and, since its first discovery, the analysis of the mechanisms underlying long-term changes of synaptic strength has been almost exclusively focused on excitatory connections. Conversely, inhibition was considered as a fixed controller of circuit excitability. Only recently, inhibitory networks were shown to be finely regulated by a wide number of mechanisms residing in their synaptic connections. Here, we review recent findings on the forms of inhibitory plasticity (IP) that have been discovered and characterized in different brain areas. In particular, we focus our attention on the molecular pathways involved in the induction and expression mechanisms leading to changes in synaptic efficacy, and we discuss, from the computational perspective, how IP can contribute to the emergence of functional properties of brain circuits.
Collapse
Affiliation(s)
- Daniela Gandolfi
- Department of Biomedical, Metabolic and Neural Sciences and Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy; (D.G.); (A.B.); (C.A.P.)
- Department of Brain and behavioral sciences, University of Pavia, 27100 Pavia, Italy
| | - Albertino Bigiani
- Department of Biomedical, Metabolic and Neural Sciences and Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy; (D.G.); (A.B.); (C.A.P.)
| | - Carlo Adolfo Porro
- Department of Biomedical, Metabolic and Neural Sciences and Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy; (D.G.); (A.B.); (C.A.P.)
| | - Jonathan Mapelli
- Department of Biomedical, Metabolic and Neural Sciences and Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy; (D.G.); (A.B.); (C.A.P.)
| |
Collapse
|
84
|
Chiu CQ, Barberis A, Higley MJ. Preserving the balance: diverse forms of long-term GABAergic synaptic plasticity. Nat Rev Neurosci 2019; 20:272-281. [PMID: 30837689 DOI: 10.1038/s41583-019-0141-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cellular mechanisms that regulate the interplay of synaptic excitation and inhibition are thought to be central to the functional stability of healthy neuronal circuits. A growing body of literature demonstrates the capacity for inhibitory GABAergic synapses to exhibit long-term plasticity in response to changes in neuronal activity. Here, we review this expanding field of research, focusing on the diversity of mechanisms that link glutamatergic signalling, postsynaptic action potentials and inhibitory synaptic strength. Several lines of evidence indicate that multiple, parallel forms of plasticity serve to regulate activity at both the input and output domains of individual neurons. Overall, these varied phenomena serve to promote both stability and flexibility over the life of the organism.
Collapse
Affiliation(s)
- Chiayu Q Chiu
- Centro Interdisciplinario de Neurociencia de Valparaiso, Universidad de Valparaiso, Valparaiso, Chile
| | | | - Michael J Higley
- Department of Neuroscience, Yale University, New Haven, CT, USA.
| |
Collapse
|
85
|
Kruijssen DLH, Wierenga CJ. Single Synapse LTP: A Matter of Context? Front Cell Neurosci 2019; 13:496. [PMID: 31780899 PMCID: PMC6861208 DOI: 10.3389/fncel.2019.00496] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/21/2019] [Indexed: 12/18/2022] Open
Abstract
The most commonly studied form of synaptic plasticity is long-term potentiation (LTP). Over the last 15 years, it has been possible to induce structural and functional LTP in dendritic spines using two-photon glutamate uncaging, allowing for studying the signaling mechanisms of LTP with single synapse resolution. In this review, we compare different stimulation methods to induce single synapse LTP and discuss how LTP is expressed. We summarize the underlying signaling mechanisms that have been studied with high spatiotemporal resolution. Finally, we discuss how LTP in a single synapse can be affected by excitatory and inhibitory synapses nearby. We argue that single synapse LTP is highly dependent on context: the choice of induction method, the history of the dendritic spine and the dendritic vicinity crucially affect signaling pathways and expression of single synapse LTP.
Collapse
Affiliation(s)
- Dennis L H Kruijssen
- Department of Biology, Science for Life, Utrecht University, Utrecht, Netherlands
| | - Corette J Wierenga
- Department of Biology, Science for Life, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
86
|
Bayer KU, Schulman H. CaM Kinase: Still Inspiring at 40. Neuron 2019; 103:380-394. [PMID: 31394063 DOI: 10.1016/j.neuron.2019.05.033] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/12/2019] [Accepted: 05/21/2019] [Indexed: 01/07/2023]
Abstract
The Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) was touted as a memory molecule, even before its involvement in long-term potentiation (LTP) was shown. The enzyme has not disappointed, with subsequent demonstrations of remarkable structural and regulatory properties. Its neuronal functions now extend to long-term depression (LTD), and last year saw the first direct evidence for memory storage by CaMKII. Although CaMKII may have taken the spotlight, it is a member of a large family of diverse and interesting CaM kinases. Our aim is to place CaMKII in context of the other CaM kinases and then review certain aspects of this kinase that are of current interest.
Collapse
Affiliation(s)
- K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | |
Collapse
|
87
|
Functional flexibility in cortical circuits. Curr Opin Neurobiol 2019; 58:175-180. [PMID: 31585330 DOI: 10.1016/j.conb.2019.09.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/18/2019] [Accepted: 09/09/2019] [Indexed: 01/15/2023]
Abstract
Cortical networks receive a highly variable stream of inputs from internal and external influences, and must flexibly adapt their operations on a short timescale. Recent work has highlighted this state-dependent functional flexibility of cortical circuits and provided initial insights into underlying circuit-level mechanisms. Transitions from quiescent to aroused or task-engaged behavioral states are associated with common motifs of network activity, including changes in correlations and enhanced sensory encoding. Evidence points to a key role for selective activation of specific GABAergic interneuron populations in mediating mode-switching in cortical networks. Finally, inhibitory interneurons may function as a critical target for convergent state-dependent neuromodulatory sculpting of cortical circuits.
Collapse
|
88
|
Tiihonen J, Koskuvi M, Storvik M, Hyötyläinen I, Gao Y, Puttonen KA, Giniatullina R, Poguzhelskaya E, Ojansuu I, Vaurio O, Cannon TD, Lönnqvist J, Therman S, Suvisaari J, Kaprio J, Cheng L, Hill AF, Lähteenvuo M, Tohka J, Giniatullin R, Lehtonen Š, Koistinaho J. Sex-specific transcriptional and proteomic signatures in schizophrenia. Nat Commun 2019; 10:3933. [PMID: 31477693 PMCID: PMC6718673 DOI: 10.1038/s41467-019-11797-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022] Open
Abstract
It has remained unclear why schizophrenia typically manifests after adolescence and which neurobiological mechanisms are underlying the cascade leading to the actual onset of the illness. Here we show that the use of induced pluripotent stem cell-derived neurons of monozygotic twins from pairs discordant for schizophrenia enhances disease-specific signal by minimizing genetic heterogeneity. In proteomic and pathway analyses, clinical illness is associated especially with altered glycosaminoglycan, GABAergic synapse, sialylation, and purine metabolism pathways. Although only 12% of all 19,462 genes are expressed differentially between healthy males and females, up to 61% of the illness-related genes are sex specific. These results on sex-specific genes are replicated in another dataset. This implies that the pathophysiology differs between males and females, and may explain why symptoms appear after adolescence when the expression of many sex-specific genes change, and suggests the need for sex-specific treatments.
Collapse
Affiliation(s)
- Jari Tiihonen
- Department of Clinical Neuroscience, Karolinska Institutet, Byggnad R5, SE-171 76, Stockholm, Sweden. .,Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240, Kuopio, Finland.
| | - Marja Koskuvi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland.,Neuroscience Center, University of Helsinki, PO Box 63, FI-00271, Helsinki, Finland
| | - Markus Storvik
- Department of Pharmacology, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Ida Hyötyläinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Yanyan Gao
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Katja A Puttonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Raisa Giniatullina
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Ekaterina Poguzhelskaya
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Ilkka Ojansuu
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240, Kuopio, Finland
| | - Olli Vaurio
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240, Kuopio, Finland
| | - Tyrone D Cannon
- Department of Psychology and Psychiatry, Yale University, 1 Prospect Street, New Haven, Connecticut, 06511, USA
| | - Jouko Lönnqvist
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, PO Box 30, FI-00271, Helsinki, Finland.,Department of Psychiatry, University of Helsinki, PO Box 22, FI-00014, Helsinki, Finland
| | - Sebastian Therman
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, PO Box 30, FI-00271, Helsinki, Finland
| | - Jaana Suvisaari
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, PO Box 30, FI-00271, Helsinki, Finland
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, PO Box 20, FI-00014, Helsinki, Finland.,Institute for Molecular Medicine FIMM, University of Helsinki, PO Box 20, FI-00014, Helsinki, Finland
| | - Lesley Cheng
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Science Drive, Bundoora, VIC, 3083, Australia
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Science Drive, Bundoora, VIC, 3083, Australia
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240, Kuopio, Finland.,Institute for Molecular Medicine FIMM, University of Helsinki, PO Box 20, FI-00014, Helsinki, Finland
| | - Jussi Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland. .,Neuroscience Center, University of Helsinki, PO Box 63, FI-00271, Helsinki, Finland.
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland. .,Neuroscience Center, University of Helsinki, PO Box 63, FI-00271, Helsinki, Finland.
| |
Collapse
|
89
|
Campbell BFN, Tyagarajan SK. Cellular Mechanisms Contributing to the Functional Heterogeneity of GABAergic Synapses. Front Mol Neurosci 2019; 12:187. [PMID: 31456660 PMCID: PMC6700328 DOI: 10.3389/fnmol.2019.00187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/19/2019] [Indexed: 11/24/2022] Open
Abstract
GABAergic inhibitory neurotransmission contributes to diverse aspects of brain development and adult plasticity, including the expression of complex cognitive processes. This is afforded for in part by the dynamic adaptations occurring at inhibitory synapses, which show great heterogeneity both in terms of upstream signaling and downstream effector mechanisms. Single-particle tracking and live imaging have revealed that complex receptor-scaffold interactions critically determine adaptations at GABAergic synapses. Super-resolution imaging studies have shown that protein interactions at synaptic sites contribute to nano-scale scaffold re-arrangements through post-translational modifications (PTMs), facilitating receptor and scaffold recruitment to synaptic sites. Additionally, plasticity mechanisms may be affected by the protein composition at individual synapses and the type of pre-synaptic input. This mini-review article examines recent discoveries of plasticity mechanisms that are operational within GABAergic synapses and discusses their contribution towards functional heterogeneity in inhibitory neurotransmission.
Collapse
Affiliation(s)
| | - Shiva K Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
90
|
Barberis A. Postsynaptic plasticity of GABAergic synapses. Neuropharmacology 2019; 169:107643. [PMID: 31108109 DOI: 10.1016/j.neuropharm.2019.05.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 12/18/2022]
Abstract
The flexibility of neuronal networks is believed to rely mainly on the plasticity of excitatory synapses. However, like their excitatory counterparts, inhibitory synapses also undergo several forms of synaptic plasticity. This review examines recent advances in the understanding of the molecular mechanisms leading to postsynaptic GABAergic plasticity. Specifically, modulation of GABAA receptor (GABAAR) number at postsynaptic sites plays a key role, with the interaction of GABAARs with the scaffold protein gephyrin and other postsynaptic scaffold/regulatory proteins having particular importance. Our understanding of these molecular interactions are progressing, based on recent insights into the processes of GABAAR lateral diffusion, gephyrin dynamics, and gephyrin nanoscale organization. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Andrea Barberis
- Plasticity of Inhibitory Networks, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genova, Italy.
| |
Collapse
|
91
|
Duman RS, Sanacora G, Krystal JH. Altered Connectivity in Depression: GABA and Glutamate Neurotransmitter Deficits and Reversal by Novel Treatments. Neuron 2019; 102:75-90. [PMID: 30946828 PMCID: PMC6450409 DOI: 10.1016/j.neuron.2019.03.013] [Citation(s) in RCA: 627] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
The mechanisms underlying the pathophysiology and treatment of depression and stress-related disorders remain unclear, but studies in depressed patients and rodent models are beginning to yield promising insights. These studies demonstrate that depression and chronic stress exposure cause atrophy of neurons in cortical and limbic brain regions implicated in depression, and brain imaging studies demonstrate altered connectivity and network function in the brains of depressed patients. Studies of the neurobiological basis of the these alterations have focused on both the principle, excitatory glutamate neurons, as well as inhibitory GABA interneurons. They demonstrate structural, functional, and neurochemical deficits in both major neuronal types that could lead to degradation of signal integrity in cortical and hippocampal regions. The molecular mechanisms underlying these changes have not been identified but are thought to be related to stress induced excitotoxic effects in combination with elevated adrenal glucocorticoids and inflammatory cytokines as well as other environmental factors. Transcriptomic studies are beginning to demonstrate important sex differences and, together with genomic studies, are starting to reveal mechanistic domains of overlap and uniqueness with regards to risk and pathophysiological mechanisms with schizophrenia and bipolar disorder. These studies also implicate GABA and glutamate dysfunction as well as immunologic mechanisms. While current antidepressants have significant time lag and efficacy limitations, new rapid-acting agents that target the glutamate and GABA systems address these issues and offer superior therapeutic interventions for this widespread and debilitating disorder.
Collapse
Affiliation(s)
- Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA.
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA
| |
Collapse
|
92
|
Using computational models to predict in vivo synaptic inputs to interneuron specific 3 (IS3) cells of CA1 hippocampus that also allow their recruitment during rhythmic states. PLoS One 2019; 14:e0209429. [PMID: 30620732 PMCID: PMC6324795 DOI: 10.1371/journal.pone.0209429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/05/2018] [Indexed: 12/05/2022] Open
Abstract
Brain coding strategies are enabled by the balance of synaptic inputs that individual neurons receive as determined by the networks in which they reside. Inhibitory cell types contribute to brain function in distinct ways but recording from specific, inhibitory cell types during behaviour to determine their contributions is highly challenging. In particular, the in vivo activities of vasoactive intestinal peptide-expressing interneuron specific 3 (IS3) cells in the hippocampus that only target other inhibitory cells are unknown at present. We perform a massive, computational exploration of possible synaptic inputs to IS3 cells using multi-compartment models and optimized synaptic parameters. We find that asynchronous, in vivo-like states that are sensitive to additional theta-timed inputs (8 Hz) exist when excitatory and inhibitory synaptic conductances are approximately equally balanced and with low numbers of activated synapses receiving correlated inputs. Specifically, under these balanced conditions, the input resistance is larger with higher mean spike firing rates relative to other activated synaptic conditions investigated. Incoming theta-timed inputs result in strongly increased spectral power relative to baseline. Thus, using a generally applicable computational approach we predict the existence and features of background, balanced states in hippocampal circuits.
Collapse
|
93
|
Dorman DB, Jędrzejewska-Szmek J, Blackwell KT. Inhibition enhances spatially-specific calcium encoding of synaptic input patterns in a biologically constrained model. eLife 2018; 7:e38588. [PMID: 30355449 PMCID: PMC6235562 DOI: 10.7554/elife.38588] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/24/2018] [Indexed: 11/13/2022] Open
Abstract
Synaptic plasticity, which underlies learning and memory, depends on calcium elevation in neurons, but the precise relationship between calcium and spatiotemporal patterns of synaptic inputs is unclear. Here, we develop a biologically realistic computational model of striatal spiny projection neurons with sophisticated calcium dynamics, based on data from rodents of both sexes, to investigate how spatiotemporally clustered and distributed excitatory and inhibitory inputs affect spine calcium. We demonstrate that coordinated excitatory synaptic inputs evoke enhanced calcium elevation specific to stimulated spines, with lower but physiologically relevant calcium elevation in nearby non-stimulated spines. Results further show a novel and important function of inhibition-to enhance the difference in calcium between stimulated and non-stimulated spines. These findings suggest that spine calcium dynamics encode synaptic input patterns and may serve as a signal for both stimulus-specific potentiation and heterosynaptic depression, maintaining balanced activity in a dendritic branch while inducing pattern-specific plasticity.
Collapse
Affiliation(s)
- Daniel B Dorman
- Interdisciplinary Program in NeuroscienceGeorge Mason UniversityFairfaxUnited States
| | | | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience, Bioengineering DepartmentKrasnow Institute for Advanced Study, George Mason UniversityFairfaxUnited States
| |
Collapse
|
94
|
Rupprecht P, Friedrich RW. Precise Synaptic Balance in the Zebrafish Homolog of Olfactory Cortex. Neuron 2018; 100:669-683.e5. [PMID: 30318416 DOI: 10.1016/j.neuron.2018.09.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/04/2018] [Accepted: 09/06/2018] [Indexed: 01/04/2023]
Abstract
Neuronal computations critically depend on the connectivity rules that govern the convergence of excitatory and inhibitory synaptic signals onto individual neurons. To examine the functional synaptic organization of a distributed memory network, we performed voltage clamp recordings in telencephalic area Dp of adult zebrafish, the homolog of olfactory cortex. In neurons of posterior Dp, odor stimulation evoked large, recurrent excitatory and inhibitory inputs that established a transient state of high conductance and synaptic balance. Excitation and inhibition in individual neurons were co-tuned to different odors and correlated on slow and fast timescales. This precise synaptic balance implies specific connectivity among Dp neurons, despite the absence of an obvious topography. Precise synaptic balance stabilizes activity patterns in different directions of coding space and in time while preserving high bandwidth. The coordinated connectivity of excitatory and inhibitory subnetworks in Dp therefore supports fast recurrent memory operations.
Collapse
Affiliation(s)
- Peter Rupprecht
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; Faculty of Natural Sciences, University of Basel, 4003 Basel, Switzerland.
| | - Rainer W Friedrich
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; Faculty of Natural Sciences, University of Basel, 4003 Basel, Switzerland.
| |
Collapse
|
95
|
Nguyen QA, Nicoll RA. The GABA A Receptor β Subunit Is Required for Inhibitory Transmission. Neuron 2018; 98:718-725.e3. [PMID: 29706582 DOI: 10.1016/j.neuron.2018.03.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 01/24/2018] [Accepted: 03/26/2018] [Indexed: 11/15/2022]
Abstract
While the canonical assembly of a GABAA receptor contains two α subunits, two β subunits, and a fifth subunit, it is unclear which variants of each subunit are necessary for native receptors. We used CRISPR/Cas9 to dissect the role of the GABAA receptor β subunits in inhibitory transmission onto hippocampal CA1 pyramidal cells and found that deletion of all β subunits 1, 2, and 3 completely eliminated inhibitory responses. In addition, only knockout of β3, alone or in combination with another β subunit, impaired inhibitory synaptic transmission. We found that β3 knockout impairs inhibitory input from PV but not SOM expressing interneurons. Furthermore, expression of β3 alone on the background of the β1-3 subunit knockout was sufficient to restore synaptic and extrasynaptic inhibitory transmission. These findings reveal a crucial role for the β3 subunit in inhibitory transmission and identify a synapse-specific role of the β3 subunit in GABAergic synaptic transmission.
Collapse
Affiliation(s)
- Quynh-Anh Nguyen
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
96
|
Bourke AM, Bowen AB, Kennedy MJ. New approaches for solving old problems in neuronal protein trafficking. Mol Cell Neurosci 2018; 91:48-66. [PMID: 29649542 DOI: 10.1016/j.mcn.2018.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 11/16/2022] Open
Abstract
Fundamental cellular properties are determined by the repertoire and abundance of proteins displayed on the cell surface. As such, the trafficking mechanisms for establishing and maintaining the surface proteome must be tightly regulated for cells to respond appropriately to extracellular cues, yet plastic enough to adapt to ever-changing environments. Not only are the identity and abundance of surface proteins critical, but in many cases, their regulated spatial positioning within surface nanodomains can greatly impact their function. In the context of neuronal cell biology, surface levels and positioning of ion channels and neurotransmitter receptors play essential roles in establishing important properties, including cellular excitability and synaptic strength. Here we review our current understanding of the trafficking pathways that control the abundance and localization of proteins important for synaptic function and plasticity, as well as recent technological advances that are allowing the field to investigate protein trafficking with increasing spatiotemporal precision.
Collapse
Affiliation(s)
- Ashley M Bourke
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Aaron B Bowen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States.
| |
Collapse
|